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










Publication year range
1.
J Cyst Fibros ; 20(2): 250-256, 2021 03.
Article in English | MEDLINE | ID: mdl-33293212

ABSTRACT

BACKGROUND: This is the first-in-human study of icenticaftor, an oral potentiator of the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) channel. Restoration of CFTR activity has shown significant clinical benefits, but more studies are needed to address all CFTR mutations. METHODS: Safety, pharmacodynamics/pharmacokinetics of icenticaftor were evaluated in a randomized, double-blind, placebo-controlled study in healthy volunteers. Efficacy was assessed in adult CF patients with ≥1 pre-specified CFTR Class III or IV mutation (150 and 450 mg bid), or homozygous for F508del mutation (450 mg bid). Primary efficacy endpoint was change from baseline in lung clearance index (LCI2.5). Secondary endpoints included %predicted FEV1 and sweat chloride level. RESULTS: Class IV mutations were present in 22 patients, Class III in 2 (both S549N), and 25 were homozygous for F508del. Icenticaftor was well-tolerated in healthy and CF subjects with no unexpected events or discontinuations in the CF groups. The most frequent study-drug related adverse events in CF patients were nausea (12.2%), headache (10.2%), and fatigue (6.1%). Icenticaftor 450 mg bid for 14 days showed significant improvements in all endpoints versus placebo in patients with Class III and IV mutations; mean %predicted FEV1 increased by 6.46%, LCI2.5 decreased by 1.13 points and sweat chloride decreased by 8.36 mmol/L. No significant efficacy was observed in patients homozygous for a single F508del. CONCLUSIONS: Icenticaftor was safe and well-tolerated in healthy volunteers and CF patients, and demonstrated clinically meaningful changes in lung function and sweat chloride level in CF patients with Class III and IV CFTR mutations. ClinicalTrials.gov: NCT02190604.


Subject(s)
Amides/therapeutic use , Chloride Channel Agonists/therapeutic use , Cystic Fibrosis/drug therapy , Pyridines/therapeutic use , Adult , Cystic Fibrosis/genetics , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Double-Blind Method , Female , Humans , Male , Mutation , Respiratory Function Tests
2.
J Med Chem ; 63(21): 12511-12525, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32658473

ABSTRACT

Multidrug resistant Gram-negative bacterial infections are an increasing public health threat due to rapidly rising resistance toward ß-lactam antibiotics. The hydrolytic enzymes called ß-lactamases are responsible for a large proportion of the resistance phenotype. ß-Lactamase inhibitors (BLIs) can be administered in combination with ß-lactam antibiotics to negate the action of the ß-lactamases, thereby restoring activity of the ß-lactam. Newly developed BLIs offer some advantage over older BLIs in terms of enzymatic spectrum but are limited to the intravenous route of administration. Reported here is a novel, orally bioavailable diazabicyclooctane (DBO) ß-lactamase inhibitor. This new DBO, ETX1317, contains an endocyclic carbon-carbon double bond and a fluoroacetate activating group and exhibits broad spectrum activity against class A, C, and D serine ß-lactamases. The ester prodrug of ETX1317, ETX0282, is orally bioavailable and, in combination with cefpodoxime proxetil, is currently in development as an oral therapy for multidrug resistant and carbapenem-resistant Enterobacterales infections.


Subject(s)
Anti-Bacterial Agents/chemistry , Azabicyclo Compounds/chemistry , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/chemistry , Administration, Oral , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Azabicyclo Compounds/metabolism , Azabicyclo Compounds/pharmacology , Azabicyclo Compounds/therapeutic use , Drug Design , Drug Evaluation, Preclinical , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Half-Life , Humans , Mice , Microbial Sensitivity Tests , Penicillin-Binding Proteins/chemistry , Penicillin-Binding Proteins/metabolism , Prodrugs/chemistry , Prodrugs/metabolism , Protein Binding , Rats , Skin Diseases/drug therapy , Skin Diseases/pathology , Skin Diseases/veterinary , Structure-Activity Relationship , beta-Lactamase Inhibitors/metabolism , beta-Lactamase Inhibitors/pharmacology , beta-Lactamase Inhibitors/therapeutic use , beta-Lactamases/metabolism
3.
ACS Infect Dis ; 1(7): 310-6, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-27622821

ABSTRACT

Bacterially expressed ß-lactamases are rapidly eroding the clinical utility of the important ß-lactam class of antibacterials, significantly impairing our ability to fight serious bacterial infections. This paper describes a study of oxaborole-derived ß-lactamase inhibitors in which crystal structures and computational modeling aided in the rational design of analogues with improved spectrum of activity against class A, C, and D enzymes. Crystal structures of two of these inhibitors covalently bound to two different serine ß-lactamases, class C Pseudomonas aeruginosa AmpC and class D OXA-10, are described herein. Improved physicochemical properties as well as increased activity against an array of ß-lactamases resulted in substantial restoration of susceptibility to ceftazidime in Escherichia coli and Klebsiella pneumoniae.

4.
ACS Med Chem Lett ; 5(8): 915-20, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-25147614

ABSTRACT

We present a comprehensive study of C6-alkylidene containing oxapenems. We show that this class of ß-lactamase inhibitors possesses an unprecedented spectrum with activity against class A, C, and D enzymes. Surprisingly, this class of compounds displayed significant photolytic instability in addition to the known hydrolytic instability. Quantum mechanical calculations were used to develop models to predict the stability of new analogues.

5.
Pharm Pat Anal ; 3(1): 87-112, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24354981

ABSTRACT

This article reviews 101 patent applications published in 2011 that disclosed small-molecule antibacterials and reported bacterial growth inhibition, in which the compounds were not similarly disclosed to be toxic to fungal or mammalian cells. The patent applications were analyzed according to their biological target and/or antibacterial class. Protein synthesis inhibitors included ligands of the 50S ribosome subunit (oxazolidinones, macrolides/ketolides and pleuromutilins), the 30S ribosome subunit (aminoglycosides and tetracyclines) and nonribosomal targets. DNA synthesis inhibitors included ligands of topoisomerase type II and type IV. Inhibitors directed at the bacterial cell envelope included those that act on cell envelope synthesis (LpxC inhibitors, penicillin-binding protein inhibitors and glycopeptides) as well as membrane disruptors (lantibiotics). Other antibacterial targets included cell division (FtsZ and WalR) and fatty acid biosynthesis (FabH/I). Compounds for which the targets are unknown or undisclosed are also covered, as are compounds aimed at overcoming resistance mechanisms (efflux inhibitors, ß-lactamase inhibitors).


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Patents as Topic , Animals , Humans , Macrolides/chemistry , Macrolides/therapeutic use , Nucleic Acid Synthesis Inhibitors/chemistry , Nucleic Acid Synthesis Inhibitors/therapeutic use , Oxazolidinones/chemistry , Oxazolidinones/therapeutic use , Patents as Topic/legislation & jurisprudence , beta-Lactamases/chemistry , beta-Lactamases/therapeutic use
6.
Pharm Pat Anal ; 1(5): 601-20, 2012 Nov.
Article in English | MEDLINE | ID: mdl-24236928

ABSTRACT

This review summarizes patent applications from 2010 for small molecules for which there is a claim of antibacterial activity. The primary criterion for inclusion in this analysis was reporting of cellular antibacterial activity data (MICs) for at least one compound. Patent applications are reviewed according to their biological target and antibacterial class. Protein synthesis inhibitors disclosed in this period include inhibitors of the 50S ribosome subunit (oxazolidinones, macrolides/ketolides and pleuromutilins), 30S ribosome subunit (aminoglycosides and tetracyclines) and nonribosomal targets (PDF inhibitors). DNA synthesis inhibitors include inhibitors of GyrA/ParC and GyrB/ParE. Cell envelope disruptors disclosed in 2010 cover both inhibitors of cell-envelope synthesis (LpxC inhibitors, ß-lactams and glycopeptides), as well as membrane disruptors (lipopeptides and polymyxins). Other antibacterial classes covered in this review include rifamycins and antibacterial peptides. Patent applications for compounds aimed at overcoming resistance mechanisms (efflux inhibitors and ß-lactamase inhibitors) are also described.


Subject(s)
Anti-Bacterial Agents , Animals , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Humans , Nucleic Acid Synthesis Inhibitors/pharmacology , Patents as Topic , Protein Synthesis Inhibitors/pharmacology
7.
Expert Opin Ther Pat ; 21(7): 1109-27, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21591993

ABSTRACT

INTRODUCTION: The mammalian target of rapamycin (mTOR) is a protein kinase and a key component of the PI3K/Akt/mTOR signaling pathway, and is deregulated in half of all human cancers. Rapamycin and its analogs (rapalogs) are allosteric inhibitors of one functional mTOR complex, mTORC1, and are clinically proven therapeutic agents for the treatment of certain cancers. However, rapalogs mainly partially inhibit mTORC1, while ATP competitive inhibitors suppress both mTORC1 and mTORC2, and therefore may offer advantages in the clinic. Recently, small-molecule inhibitors have entered clinical trials that are mTOR-selective or dual mTOR/PI3K inhibitors. AREAS COVERED: This review focuses on ATP-competitive mTOR inhibitors that have appeared in the patent literature in 2010. Many inhibitors with new structural motifs have been discovered as well as inhibitors that are related to previously disclosed structures. This review endeavors to put into perspective the diverse structural elements that make up these compounds. Patent applications are covered that include either selective mTOR inhibitors or dual mTOR/PI3K inhibitors. EXPERT OPINION: The PI3K/mTOR signaling pathway is an exciting target for the development of pharmaceuticals to treat cancer and other diseases, due to the unique combination of a clinically and commercially validated pathway approach (i.e., rapalogs), combined with a biological rationale for further increased efficacy (i.e., ATP-competitive inhibitors). With the number of candidate drugs currently in development or at earlier stages of the drug discovery pipeline, we are bound to see small-molecule inhibitors reach pivotal trials, and hopefully the market, in the near future.


Subject(s)
Adenosine Triphosphate/metabolism , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Binding, Competitive , Drug Delivery Systems , Drug Design , Humans , Neoplasms/drug therapy , Neoplasms/physiopathology , Patents as Topic
8.
Curr Opin Drug Discov Devel ; 13(4): 428-40, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20597028

ABSTRACT

mTOR is a serine-threonine kinase that plays a key role in the regulation of cellular growth. The mTOR pathway consists of two distinct complexes: mTOR/Raptor (mTORC1) and mTOR/Rictor (mTORC2). In response to changes in the levels of insulin, nutrients and energy supply, signaling through these complexes affects a variety of processes, including protein translation and cell proliferation. The efficacy of derivatives of the natural product rapamycin (sirolimus), which functions as an allosteric inhibitor of mTORC1, has validated mTOR inhibition as an anticancer treatment. More recently, extensive efforts have been focused on the discovery of ATP-competitive inhibitors of mTOR that would inhibit both mTORC1 and mTORC2 and may provide additional clinical benefit. This review provides a summary of recent research efforts in this field, focusing on mTOR inhibitors that are selective for mTOR over the related lipid kinase PI3K.


Subject(s)
Adenosine Triphosphate/pharmacology , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases , Signal Transduction/drug effects , Animals , Humans , Immunosuppressive Agents , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Purines/chemistry , Purines/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
9.
Bioorg Med Chem Lett ; 20(8): 2644-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20227881

ABSTRACT

Incorporation of bridged morpholines in monocyclic triazine PI3K/mTOR inhibitors gave compounds with increased mTOR selectivity relative to the corresponding morpholine analogs. Compounds with ureidophenyl groups gave highly potent and selective mTOR inhibitors. Potency and selectivity was demonstrated both in vitro and in vivo through biomarker suppression studies. Select compounds exhibited potent inhibition of tumor growth in nude mouse xenograft assays upon PO and IV administration.


Subject(s)
Adenosine Triphosphate/chemistry , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Morpholines/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Triazines/chemistry , Triazines/pharmacology , Animals , Drug Discovery , Inhibitory Concentration 50 , Mice , Mice, Nude , TOR Serine-Threonine Kinases
10.
Bioorg Med Chem Lett ; 20(8): 2648-53, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20223663

ABSTRACT

Isosteric replacement of one of the 3,5-ethylene-bridged morpholines in 2-arylureidophenyl-4,6-di(3-oxa-8-azabicyclo[3.2.1]octan-8-yl)triazines led to significant improvements in human microsomal stability. 3-R-Me-morpholine and tetrahydropyran were identified as preferred isosteres for the bridged morpholine. Combination of tetrahydropyran substitution with an N-Me-piperazinophenylureido group led to 27, that selectively suppressed mTOR biomarkers in vivo and possessed excellent efficacy in a murine xenograft model.


Subject(s)
Microsomes/drug effects , Triazines/pharmacology , Animals , Humans , Inhibitory Concentration 50 , Mice , Microsomes/metabolism , Transplantation, Heterologous , Triazines/chemistry
11.
Bioorg Med Chem Lett ; 20(8): 2654-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20223664

ABSTRACT

Potent inhibitors of the mammalian target of rapamycin (mTOR) which contain the triazine scaffold and the (R)-3-methyl morpholine moiety have been identified. Such compounds also demonstrated good selectivity over the related lipid kinase PI3Kalpha. Incorporation of additional functionality at the 4-position of the arylureidophenyl ring resulted in compounds with enhanced cellular activity.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Morpholines/chemistry , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Triazines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases , Triazines/chemistry
12.
Cancer Res ; 70(2): 621-31, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068177

ABSTRACT

The mammalian target of rapamycin (mTOR) is a major component of the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway that is dysregulated in 50% of all human malignancies. Rapamycin and its analogues (rapalogs) partially inhibit mTOR through allosteric binding to mTOR complex 1 (mTORC1) but not mTOR complex 2 (mTORC2), an emerging player in cancer. Here, we report WYE-125132 (WYE-132), a highly potent, ATP-competitive, and specific mTOR kinase inhibitor (IC(50): 0.19 +/- 0.07 nmol/L; >5,000-fold selective versus PI3Ks). WYE-132 inhibited mTORC1 and mTORC2 in diverse cancer models in vitro and in vivo. Importantly, consistent with genetic ablation of mTORC2, WYE-132 targeted P-AKT(S473) and AKT function without significantly reducing the steady-state level of the PI3K/PDK1 activity biomarker P-AKT(T308), highlighting a prominent and direct regulation of AKT by mTORC2 in cancer cells. Compared with the rapalog temsirolimus/CCI-779, WYE-132 elicited a substantially stronger inhibition of cancer cell growth and survival, protein synthesis, cell size, bioenergetic metabolism, and adaptation to hypoxia. Oral administration of WYE-132 to tumor-bearing mice showed potent single-agent antitumor activity against MDA361 breast, U87MG glioma, A549 and H1975 lung, as well as A498 and 786-O renal tumors. An optimal dose of WYE-132 achieved a substantial regression of MDA361 and A549 large tumors and caused complete regression of A498 large tumors when coadministered with bevacizumab. Our results further validate mTOR as a critical driver for tumor growth, establish WYE-132 as a potent and profound anticancer agent, and provide a strong rationale for clinical development of specific mTOR kinase inhibitors as new cancer therapy.


Subject(s)
Neoplasms/drug therapy , Phenylurea Compounds/pharmacology , Pyrazoles/pharmacology , Sirolimus/analogs & derivatives , Transcription Factors/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Hypoxia/drug effects , Female , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proteins , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases
13.
Bioorg Med Chem Lett ; 20(4): 1440-4, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20089401

ABSTRACT

A series of pyrazolopyrimidine mammalian Target Of Rapamycin (mTOR) inhibitors with various substituents at the 1-position have been prepared, resulting in compounds with excellent potency, selectivity and microsomal stability. Combination of a 1-cyclohexyl ketal group with a 2,6-ethylene bridged morpholine in the 4-position and a ureidophenyl group in the 6-positon resulted in compound 8a, that selectively suppressed key mTOR biomarkers in vivo for at least 8h following iv administration and showed excellent oral activity in a xenograft tumor model.


Subject(s)
Adenosine Triphosphate/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Pyrazoles/chemistry , Pyridines/chemistry , Adenosine Triphosphate/chemistry , Administration, Oral , Animals , Binding, Competitive , Drug Stability , Humans , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Microsomes/enzymology , Molecular Structure , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/chemical synthesis , Pyridines/chemical synthesis , Structure-Activity Relationship , TOR Serine-Threonine Kinases , Xenograft Model Antitumor Assays
14.
Bioorg Med Chem Lett ; 20(2): 640-3, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19963384

ABSTRACT

The morpholine hinge-region binding group on a series of pyrazolopyrimidine and thienopyrimidine mammalian target of rapamycin (mTOR) inhibitors was replaced with 3,6-dihydro-2H-pyran (DHP), giving compounds of equivalent potency and selectivity versus PI3K. These results establish the DHP group as a hinge-region binding motif for the preparation of highly potent and selective mTOR inhibitors.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Morpholines/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrans/chemistry , Pyrazoles/chemistry , Pyridines/chemistry , Pyrimidines/chemistry , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Binding, Competitive , Computer Simulation , Drug Discovery , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Microsomes/metabolism , Models, Molecular , Protein Serine-Threonine Kinases/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases
15.
Bioorg Med Chem Lett ; 20(1): 375-9, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19897362

ABSTRACT

2-Aryl-4-morpholinothieno[3,2-d]pyrimidines are known PI3K inhibitors. This class of compounds also potently inhibited the homologous enzyme mTOR. Replacement of the morpholine group in these compounds with an 8-oxa-3-azabicyclo[3.2.1]octane group led to mTOR inhibitors with selectivity over PI3K. Optimization of the 2-aryl substituent led to the discovery of 2-(4-ureidophenyl)-thienopyrimidines as highly potent (IC(50) <1nM) mTOR inhibitors with excellent selectivity (up to >1000-fold) over PI3K and good potency in a cellular proliferation assay (IC(50) <50nM).


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/chemistry , Tropanes/chemistry , Cell Line, Tumor , Drug Discovery , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Morpholines/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Structure-Activity Relationship , TOR Serine-Threonine Kinases
16.
J Med Chem ; 52(24): 7942-5, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-19916508

ABSTRACT

Dramatic improvements in mTOR-targeting selectivity were achieved by replacing morpholine in pyrazolopyrimidine inhibitors with bridged morpholines. Analogues with subnanomolar mTOR IC(50) values and up to 26000-fold selectivity versus PI3Kalpha were prepared. Chiral morpholines gave inhibitors whose enantiomers had different selectivity and potency profiles. Molecular modeling suggests that a single amino acid difference between PI3K and mTOR (Phe961Leu) accounts for the profound selectivity seen by creating a deeper pocket in mTOR that can accommodate bridged morpholines.


Subject(s)
Morpholines/pharmacology , Protein Kinases/chemistry , Models, Molecular , Morpholines/chemical synthesis , Morpholines/chemistry , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinases/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Structure-Activity Relationship , TOR Serine-Threonine Kinases
17.
Bioorg Med Chem Lett ; 19(24): 6830-5, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19896845

ABSTRACT

A series of highly potent and selective pyrazolopyrimidine mTOR inhibitors which contain water-solubilizing groups attached to the 6-arylureidophenyl moiety have been prepared. Such derivatives displayed superior potency to those in which these appendages were attached to alternative sites. In comparison to unfunctionalized arylureido compounds, these analogs demonstrated enhanced cellular potency and significantly improved stability towards human microsomes, resulting in an mTOR inhibitor with impressive efficacy in a xenograft model with an intermittent dosing regimen.


Subject(s)
Antineoplastic Agents/chemistry , Intracellular Signaling Peptides and Proteins/drug effects , Microsomes, Liver/enzymology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/drug effects , Pyrazoles/chemistry , Pyrimidines/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Drug Discovery , Drug Stability , Humans , Mice , Mice, Nude , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/pharmacokinetics , Solubility , TOR Serine-Threonine Kinases , Water/chemistry , Xenograft Model Antitumor Assays
18.
J Med Chem ; 52(24): 8010-24, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-19894727

ABSTRACT

Design and synthesis of a series of 4-morpholino-6-aryl-1H-pyrazolo[3,4-d]pyrimidines as potent and selective inhibitors of the mammalian target of rapamycin (mTOR) are described. Optimization of the 6-aryl substituent led to the discovery of inhibitors carrying 6-ureidophenyl groups, the first reported active site inhibitors of mTOR with subnanomolar inhibitory concentrations. The data presented in this paper show that 6-arylureidophenyl substituents led to potent mixed inhibitors of mTOR and phosphatidylinositol 3-kinase alpha (PI3K-alpha), whereas 6-alkylureidophenyl appendages gave highly selective mTOR inhibitors. Combination of 6-alkylureidophenyl groups with 1-carbamoylpiperidine substitution resulted in compounds with subnanomolar IC(50) against mTOR and greater than 1000-fold selectivity over PI3K-alpha. In addition, structure based drug design resulted in the preparation of several 6-arylureidophenyl-1H-pyrazolo[3,4-d]pyrimidines, substituted in the 4-position of the arylureido moiety with water solubilizing groups. These compounds combined potent mTOR inhibition (IC(50) < 1 nM) with unprecedented activity in cellular proliferation assays (IC(50) < 1 nM).


Subject(s)
Adenosine Triphosphate/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Pyrimidines/pharmacology , Adenosine Triphosphate/metabolism , Binding, Competitive , Cell Growth Processes/drug effects , Cell Line, Tumor , Humans , Male , Models, Molecular , Morpholines/chemical synthesis , Morpholines/chemistry , Morpholines/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinases/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , TOR Serine-Threonine Kinases
19.
J Med Chem ; 52(22): 7081-9, 2009 Nov 26.
Article in English | MEDLINE | ID: mdl-19848404

ABSTRACT

The mammalian target of rapamycin (mTOR) is a central regulator of cell growth, metabolism, and angiogenesis and an emerging target in cancer research. High throughput screening (HTS) of our compound collection led to the identification of 3-(4-morpholin-4-yl-1-piperidin-4-yl-1H-pyrazolo[3,4-d]pyrimidin-6-yl)phenol (5a), a modestly potent and nonselective inhibitor of mTOR and phosphoinositide 3-kinase (PI3K). Optimization of compound 5a, employing an mTOR homology model based on an X-ray crystal structure of closely related PI3Kgamma led to the discovery of 6-(1H-indol-5-yl)-4-morpholin-4-yl-1-[1-(pyridin-3-ylmethyl)piperidin-4-yl]-1H-pyrazolo[3,4-d]pyrimidine (5u), a potent and selective mTOR inhibitor (mTOR IC(50) = 9 nM; PI3Kalpha IC(50) = 1962 nM). Compound 5u selectively inhibited cellular biomarker of mTORC1 (P-S6K, P-4EBP1) and mTORC2 (P-AKT S473) over the biomarker of PI3K/PDK1 (P-AKT T308) and did not inhibit PI3K-related kinases (PIKKs) in cellular assays. These pyrazolopyrimidines represent an exciting new series of mTOR-selective inhibitors with potential for development for cancer therapy.


Subject(s)
Drug Discovery , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Pyrimidines/pharmacology , Binding, Competitive , Cell Line, Tumor , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Molecular Weight , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinases/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/metabolism , Signal Transduction/drug effects , Substrate Specificity , TOR Serine-Threonine Kinases
20.
J Med Chem ; 52(16): 5013-6, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19645448

ABSTRACT

The mammalian target of rapamycin (mTOR), a central regulator of growth, survival, and metabolism, is a validated target for cancer therapy. Rapamycin and its analogues, allosteric inhibitors of mTOR, only partially inhibit one mTOR protein complex. ATP-competitive, global inhibitors of mTOR that have the potential for enhanced anticancer efficacy are described. Structural features leading to potency and selectivity were identified and refined leading to compounds with in vivo efficacy in tumor xenograft models.


Subject(s)
Adenosine Triphosphate/physiology , Antineoplastic Agents/chemical synthesis , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/chemical synthesis , Pyrimidines/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Carbamates/chemical synthesis , Carbamates/chemistry , Carbamates/pharmacology , Cell Line, Tumor , Class Ib Phosphatidylinositol 3-Kinase , Crystallography, X-Ray , Drug Design , Intracellular Signaling Peptides and Proteins/chemistry , Isoenzymes/chemistry , Mice , Mice, Nude , Microsomes/metabolism , Models, Molecular , Phosphatidylinositol 3-Kinases/chemistry , Protein Serine-Threonine Kinases/chemistry , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Structure-Activity Relationship , TOR Serine-Threonine Kinases , Urea/analogs & derivatives , Urea/chemical synthesis , Urea/chemistry , Urea/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...