Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Med Chem ; 67(5): 3437-3447, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38363074

ABSTRACT

Human African trypanosomiasis (HAT), a neglected tropical disease caused by Trypanosoma brucei gambiense (Tbg) or Trypanosoma brucei rhodesiense (Tbr), remains a significant public health concern with over 55 million people at risk of infection. Current treatments for HAT face the challenges of poor efficacy, drug resistance, and toxicity. This study presents the synthesis and evaluation of chloronitrobenzamides (CNBs) against Trypanosoma species, identifying previously reported compound 52 as a potent and selective orally bioavailable antitrypanosomal agent. 52 was well tolerated in vivo and demonstrated favorable oral pharmacokinetics, maintaining plasma concentrations surpassing the cellular EC50 for over 24 h and achieving peak brain concentrations exceeding 7 µM in rodents after single oral administration (50 mg/kg). Treatment with 52 significantly extended the lifespan of mice infected with Trypanosoma congolense and T. brucei rhodesiense. These results demonstrate that 52 is a strong antitrypanosomal lead with potential for developing treatments for both human and animal African trypanosomiasis.


Subject(s)
Trypanocidal Agents , Trypanosoma brucei brucei , Trypanosomiasis, African , Humans , Animals , Mice , Trypanosomiasis, African/drug therapy , Trypanosoma brucei rhodesiense , Trypanosoma brucei gambiense , Trypanocidal Agents/toxicity , Trypanocidal Agents/therapeutic use
2.
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
3.
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
4.
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
5.
J Med Chem ; 59(17): 7950-62, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27505686

ABSTRACT

Phenotypic whole-cell screening in erythrocytic cocultures of Plasmodium falciparum identified a series of dihydroisoquinolones that possessed potent antimalarial activity against multiple resistant strains of P. falciparum in vitro and show no cytotoxicity to mammalian cells. Systematic structure-activity studies revealed relationships between potency and modifications at N-2, C-3, and C-4. Careful structure-property relationship studies, coupled with studies of metabolism, addressed the poor aqueous solubility and metabolic vulnerability, as well as potential toxicological effects, inherent in the more potent primary screening hits such as 10b. Analogues 13h and 13i, with structural modifications at each site, were shown to possess excellent antimalarial activity in vivo. The (+)-(3S,4S) enantiomer of 13i and similar analogues were identified as the more potent. On the basis of these studies, we have selected (+)-13i for further study as a preclinical candidate.


Subject(s)
Anilides/chemistry , Antimalarials/chemistry , Isoquinolines/chemistry , Plasmodium falciparum/drug effects , Anilides/chemical synthesis , Anilides/pharmacology , Anilides/toxicity , Animals , Antimalarials/chemical synthesis , Antimalarials/pharmacology , Antimalarials/toxicity , Coculture Techniques , Erythrocytes/cytology , Erythrocytes/parasitology , Humans , Isoquinolines/chemical synthesis , Isoquinolines/pharmacology , Isoquinolines/toxicity , Mice , Microsomes, Liver/metabolism , Plasmodium falciparum/physiology , Solubility , Stereoisomerism , Structure-Activity Relationship
6.
J Med Chem ; 59(2): 559-77, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26632965

ABSTRACT

We previously reported a novel inhibitor of the ataxia-telangiectasia mutated (ATM) kinase, which is a target for novel radiosensitizing drugs. While our initial lead, compound 4, was relatively potent and nontoxic, it exhibited poor stability to oxidative metabolism and relatively poor selectivity against other kinases. The current study focused on balancing potency and selectivity with metabolic stability through structural modification to the metabolized site on the quinazoline core. We performed extensive structure-activity and structure-property relationship studies on this quinazoline ATM kinase inhibitor in order to identify structural variants with enhanced selectivity and metabolic stability. We show that, while the C-7-methoxy group is essential for potency, replacing the C-6-methoxy group considerably improves metabolic stability without affecting potency. Promising analogues 20, 27g, and 27n were selected based on in vitro pharmacology and evaluated in murine pharmacokinetic and tolerability studies. Compound 27g possessed significantly improve pharmacokinetics relative to that of 4. Compound 27g was also significantly more selective against other kinases than 4. Therefore, 27g is a good candidate for further development as a potential radiosensitizer.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Radiation-Sensitizing Agents/chemical synthesis , Radiation-Sensitizing Agents/pharmacology , Animals , Colony-Forming Units Assay , Drug Delivery Systems , Drug Design , Female , Humans , In Vitro Techniques , MCF-7 Cells , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Quinazolines/chemical synthesis , Quinazolines/pharmacology , Radiation-Sensitizing Agents/pharmacokinetics , Structure-Activity Relationship , Substrate Specificity
7.
Proc Natl Acad Sci U S A ; 111(50): E5455-62, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25453091

ABSTRACT

Drug discovery for malaria has been transformed in the last 5 years by the discovery of many new lead compounds identified by phenotypic screening. The process of developing these compounds as drug leads and studying the cellular responses they induce is revealing new targets that regulate key processes in the Plasmodium parasites that cause malaria. We disclose herein that the clinical candidate (+)-SJ733 acts upon one of these targets, ATP4. ATP4 is thought to be a cation-transporting ATPase responsible for maintaining low intracellular Na(+) levels in the parasite. Treatment of parasitized erythrocytes with (+)-SJ733 in vitro caused a rapid perturbation of Na(+) homeostasis in the parasite. This perturbation was followed by profound physical changes in the infected cells, including increased membrane rigidity and externalization of phosphatidylserine, consistent with eryptosis (erythrocyte suicide) or senescence. These changes are proposed to underpin the rapid (+)-SJ733-induced clearance of parasites seen in vivo. Plasmodium falciparum ATPase 4 (pfatp4) mutations that confer resistance to (+)-SJ733 carry a high fitness cost. The speed with which (+)-SJ733 kills parasites and the high fitness cost associated with resistance-conferring mutations appear to slow and suppress the selection of highly drug-resistant mutants in vivo. Together, our data suggest that inhibitors of PfATP4 have highly attractive features for fast-acting antimalarials to be used in the global eradication campaign.


Subject(s)
Antimalarials/pharmacology , Calcium-Transporting ATPases/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Isoquinolines/pharmacology , Malaria/drug therapy , Models, Molecular , Plasmodium/drug effects , Antimalarials/pharmacokinetics , Calcium-Transporting ATPases/genetics , Cellular Senescence/drug effects , Drug Discovery , Drug Resistance/genetics , Erythrocytes/drug effects , Flow Cytometry , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , High-Throughput Screening Assays , Isoquinolines/pharmacokinetics , Molecular Structure
8.
Bioorg Med Chem Lett ; 23(14): 4127-31, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23746473

ABSTRACT

We previously reported the phenylchloronitrobenzamides (PCNBs), a novel class of compounds active against the species of trypanosomes that cause Human African Trypanosomiasis (HAT). Herein, we explored the potential to adjust the reactivity of the electrophilic chloronitrobenzamide core. These studies identified compound 7d that potently inhibited the growth of trypanosomes (EC50=120nM for Trypanosoma b. brucei, 18nM for Trypanosoma b. rhodesiense, and 38nM for Trypanosoma b. gambiense) without significant cytotoxicity against mammalian cell lines (EC50>25µM for HepG2, HEK293, Raji, and BJ cell lines) and also had good stability in microsomal models (t1/2>4h in both human and mouse). Overall these properties indicate the compound 7d and its analogs are worth further exploration as potential leads for HAT.


Subject(s)
Benzamides/chemistry , Trypanocidal Agents/chemistry , Trypanosoma brucei brucei/drug effects , Animals , Benzamides/chemical synthesis , Benzamides/toxicity , Cell Line , Hep G2 Cells , Humans , Mice , Microsomes/metabolism , Solubility , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/toxicity
9.
J Med Chem ; 56(7): 2850-60, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23484493

ABSTRACT

We previously reported the discovery of the activity of chloronitrobenzamides (CNBs) against bloodstream forms of Trypanosoma brucei . Herein we disclose extensive structure-activity relationship and structure-property relationship studies aimed at identification of tractable early leads for clinical development. These studies revealed a promising lead compound, 17b, that exhibited nanomolar potency against T. brucei (EC50 = 27 nM for T. b. brucei, 7 nM for T. b. rhodesiense, and 2 nM for T. b. gambiense ) with excellent selectivity for parasite cells relative to mammalian cell lines (EC50 > 25 µM). In addition compound 17b displayed suitable physiochemical characteristics and microsomal stability (t1/2 > 4 h for human and mouse) to justify pursuing in vivo studies.


Subject(s)
Benzamides/pharmacology , Trypanosomiasis, African/drug therapy , Animals , Benzamides/chemistry , Cell Line , Drug Evaluation, Preclinical , Humans , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...