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










Publication year range
1.
Bioorg Med Chem Lett ; 47: 128111, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34353608

ABSTRACT

Flavaglines such as silvestrol (1) and rocaglamide (2) constitute an interesting class of natural products with promising anticancer activities. Their mode of action is based on inhibition of eukaryotic initiation factor 4A (eIF4A) dependent translation through formation of a stable ternary complex with eIF4A and mRNA, thus blocking ribosome scanning. Herein we describe initial SAR studies in a novel series of 1-aminomethyl substituted flavagline-inspired eIF4A inhibitors. We discovered that a variety of N-substitutions at the 1-aminomethyl group are tolerated, making this position pertinent for property and ADME profile tuning. The findings presented herein are relevant to future drug design efforts towards novel eIF4A inhibitors with drug-like properties.


Subject(s)
Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Biological Products/pharmacology , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Triterpenes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzofurans/chemical synthesis , Benzofurans/chemistry , Biological Products/chemical synthesis , Biological Products/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Eukaryotic Initiation Factor-4A/metabolism , Humans , Molecular Structure , Structure-Activity Relationship , Triterpenes/chemical synthesis , Triterpenes/chemistry
2.
Mol Cancer Ther ; 20(1): 26-36, 2021 01.
Article in English | MEDLINE | ID: mdl-33037136

ABSTRACT

The PI3K/AKT/mTOR pathway is often activated in lymphoma through alterations in PI3K, PTEN, and B-cell receptor signaling, leading to dysregulation of eIF4A (through its regulators, eIF4B, eIF4G, and PDCD4) and the eIF4F complex. Activation of eIF4F has a direct role in tumorigenesis due to increased synthesis of oncogenes that are dependent on enhanced eIF4A RNA helicase activity for translation. eFT226, which inhibits translation of specific mRNAs by promoting eIF4A1 binding to 5'-untranslated regions (UTR) containing polypurine and/or G-quadruplex recognition motifs, shows potent antiproliferative activity and significant in vivo efficacy against a panel of diffuse large B-cell lymphoma (DLBCL), and Burkitt lymphoma models with ≤1 mg/kg/week intravenous administration. Evaluation of predictive markers of sensitivity or resistance has shown that activation of eIF4A, mediated by mTOR signaling, correlated with eFT226 sensitivity in in vivo xenograft models. Mutation of PTEN is associated with reduced apoptosis in vitro and diminished efficacy in vivo in response to eFT226. In models evaluated with PTEN loss, AKT was stimulated without a corresponding increase in mTOR activation. AKT activation leads to the degradation of PDCD4, which can alter eIF4F complex formation. The association of eFT226 activity with PTEN/PI3K/mTOR pathway regulation of mRNA translation provides a means to identify patient subsets during clinical development.


Subject(s)
Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Oncogenes , Protein Biosynthesis/genetics , RNA, Messenger/genetics , Animals , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Eukaryotic Initiation Factor-4A/metabolism , Female , Humans , Mice, Inbred NOD , Mice, SCID , PTEN Phosphohydrolase/metabolism , RNA, Messenger/metabolism , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
3.
Org Lett ; 22(16): 6257-6261, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32806219

ABSTRACT

Rocaglates, rocaglamides, and related flavagline natural products exert their remarkable anticancer activity through inhibition of eukaryotic initiation factor 4A (eIF4A) but generally display suboptimal drug-like properties. In our efforts to identify potent drug-like eIF4A inhibitors, we developed synthetic strategies for diastereoselectively functionalizing the C1 position of aza-rocaglamide scaffolds (cf. 14 and 18), which proceed via retention or inversion of configuration at C1 depending on the C2 substituent (cf. 15 and 21) and ultimately enabled the discovery of novel and potent eIF4A inhibitors such as 25.


Subject(s)
Benzofurans/chemistry , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Binding Sites , Biological Products , Eukaryotic Initiation Factor-4A/metabolism , Humans , Molecular Structure
4.
J Med Chem ; 63(11): 5879-5955, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32470302

ABSTRACT

Dysregulation of protein translation is a key driver for the pathogenesis of many cancers. Eukaryotic initiation factor 4A (eIF4A), an ATP-dependent DEAD-box RNA helicase, is a critical component of the eIF4F complex, which regulates cap-dependent protein synthesis. The flavagline class of natural products (i.e., rocaglamide A) has been shown to inhibit protein synthesis by stabilizing a translation-incompetent complex for select messenger RNAs (mRNAs) with eIF4A. Despite showing promising anticancer phenotypes, the development of flavagline derivatives as therapeutic agents has been hampered because of poor drug-like properties as well as synthetic complexity. A focused effort was undertaken utilizing a ligand-based design strategy to identify a chemotype with optimized physicochemical properties. Also, detailed mechanistic studies were undertaken to further elucidate mRNA sequence selectivity, key regulated target genes, and the associated antitumor phenotype. This work led to the design of eFT226 (Zotatifin), a compound with excellent physicochemical properties and significant antitumor activity that supports clinical development.


Subject(s)
Benzofurans/chemistry , Drug Design , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Animals , Benzofurans/pharmacokinetics , Benzofurans/therapeutic use , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Crystallography, X-Ray , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Female , Half-Life , Humans , Ligands , Mice , Mice, Nude , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Structure, Tertiary , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Rats , Structure-Activity Relationship
5.
J Med Chem ; 61(8): 3516-3540, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29526098

ABSTRACT

Dysregulated translation of mRNA plays a major role in tumorigenesis. Mitogen-activated protein kinase interacting kinases (MNK)1/2 are key regulators of mRNA translation integrating signals from oncogenic and immune signaling pathways through phosphorylation of eIF4E and other mRNA binding proteins. Modulation of these key effector proteins regulates mRNA, which controls tumor/stromal cell signaling. Compound 23 (eFT508), an exquisitely selective, potent dual MNK1/2 inhibitor, was designed to assess the potential for control of oncogene signaling at the level of mRNA translation. The crystal structure-guided design leverages stereoelectronic interactions unique to MNK culminating in a novel pyridone-aminal structure described for the first time in the kinase literature. Compound 23 has potent in vivo antitumor activity in models of diffuse large cell B-cell lymphoma and solid tumors, suggesting that controlling dysregulated translation has real therapeutic potential. Compound 23 is currently being evaluated in Phase 2 clinical trials in solid tumors and lymphoma. Compound 23 is the first highly selective dual MNK inhibitor targeting dysregulated translation being assessed clinically.


Subject(s)
Antineoplastic Agents/therapeutic use , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/therapeutic use , Pyridones/therapeutic use , Pyrimidines/therapeutic use , Spiro Compounds/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Catalytic Domain , Cell Line, Tumor , Drug Design , Eukaryotic Initiation Factor-4E/chemistry , Eukaryotic Initiation Factor-4E/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Molecular Structure , Phosphorylation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Pyridines/chemical synthesis , Pyridines/chemistry , Pyridines/pharmacology , Pyridones/chemical synthesis , Pyridones/chemistry , Pyridones/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Serine/chemistry , Signal Transduction/drug effects , Spiro Compounds/chemical synthesis , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Xenograft Model Antitumor Assays
6.
J Med Chem ; 57(8): 3382-400, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24673104

ABSTRACT

A structure-based drug design strategy was used to optimize a novel benzolactam series of HSP90α/ß inhibitors to achieve >1000-fold selectivity versus the HSP90 endoplasmic reticulum and mitochondrial isoforms (GRP94 and TRAP1, respectively). Selective HSP90α/ß inhibitors were found to be equipotent to pan-HSP90 inhibitors in promoting the clearance of mutant huntingtin protein (mHtt) in vitro, however with less cellular toxicity. Improved tolerability profiles may enable the use of HSP90α/ß selective inhibitors in treating chronic neurodegenerative indications such as Huntington's disease (HD). A potent, selective, orally available HSP90α/ß inhibitor was identified (compound 31) that crosses the blood-brain barrier. Compound 31 demonstrated proof of concept by successfully reducing brain Htt levels following oral dosing in rats.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Huntington Disease/drug therapy , Animals , Drug Design , HSP90 Heat-Shock Proteins/chemistry , Humans , Male , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 24(1): 204-8, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24332488

ABSTRACT

HSP90 continues to be a target of interest for neurodegeneration indications. Selective knockdown of the HSP90 cytosolic isoforms α and ß is sufficient to reduce mutant huntingtin protein levels in vitro. Chemotype-dependent binding conformations of HSP90α/ß appear to strongly influence isoform selectivity. The rational design of HSP90α/ß inhibitors selective versus the mitochondrial (TRAP1) and endoplasmic reticulum (GRP94) isoforms offers a potential mitigating strategy for mechanism-based toxicities. Better tolerated HSP90 inhibitors would be attractive for targeting chronic neurodegenerative diseases such as Huntington's disease.


Subject(s)
Drug Design , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Neurodegenerative Diseases/drug therapy , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure , Protein Isoforms/antagonists & inhibitors
8.
Bioorg Med Chem Lett ; 20(16): 4819-24, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20663667

ABSTRACT

We have optimized a novel series of potent p38 MAP kinase inhibitors based on an alpha-ketoamide scaffold through structure based design that due to their extended molecular architecture bind, in addition to the ATP site, to an allosteric pocket. In vitro ADME, in vivo PK and efficacy studies show these compounds to have drug-like characteristics and have resulted in the nomination of a development candidate which is currently in phase II clinical trials for the oral treatment of inflammatory conditions.


Subject(s)
Amides/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Protein Kinase Inhibitors/chemistry , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Administration, Oral , Allosteric Site , Amides/chemical synthesis , Amides/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Binding Sites , Cell Line , Computer Simulation , Humans , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Rats , Structure-Activity Relationship , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Bioorg Med Chem Lett ; 18(6): 1772-7, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18325768

ABSTRACT

We have identified a novel series of potent p38 MAP kinase inhibitors through structure-based design which due to their extended molecular architecture bind, in addition to the ATP site, to an allosteric pocket. In vitro ADME and in vivo PK studies show these compounds to have drug-like characteristics which could result in the development of an oral treatment for inflammatory conditions.


Subject(s)
Amides/chemical synthesis , Drug Design , Protein Kinase Inhibitors/chemical synthesis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Allosteric Site , Amides/chemistry , Amides/pharmacokinetics , Amides/pharmacology , Animals , Binding Sites , Cells, Cultured , Crystallography, X-Ray , Humans , Lipopolysaccharides/pharmacology , Models, Molecular , Molecular Structure , Monocytes/cytology , Monocytes/drug effects , Phosphorylation/drug effects , Protein Binding , Protein Conformation , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Rats , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Am Chem Soc ; 127(29): 10191-6, 2005 Jul 27.
Article in English | MEDLINE | ID: mdl-16028929

ABSTRACT

We describe a general method for the mimicry of one face of an alpha-helix based on a terphenyl scaffold that spatially projects functionality in a manner similar to that of two turns of an alpha-helix. The synthetic scaffold reduces the flexibility and molecular weight of the mimicked protein secondary structure. We have applied this design to the development of antagonists of the alpha-helix binding protein Bcl-x(L). Using a sequential synthetic strategy, we have prepared a library of terphenyl derivatives to mimic the helical region of the Bak BH3 domain that binds Bcl-x(L). Fluorescence polarization assays were carried out to evaluate the ability of terphenyl derivatives to displace the Bcl-x(L)-bound Bak peptide. Terphenyl 14 exhibited good in vitro affinity with a K(i) value of 0.114 muM. These terphenyl derivatives were more selective at disrupting the Bcl-x(L)/Bak over the HDM2/p53 interaction, which involves binding of the N-terminal alpha-helix of p53 to HDM2. Structural studies using NMR spectroscopy and computer-aided docking simulations suggested that the helix binding area on the surface of Bcl-x(L) is the target for the synthetic ligands. Treatment of human embryonic kidney 293 (HEK293) cells with terphenyl derivatives resulted in the disruption of the binding of Bcl-x(L) to Bax in intact cells.


Subject(s)
Membrane Proteins/chemistry , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/chemistry , Terphenyl Compounds/chemistry , Terphenyl Compounds/pharmacology , Biomimetic Materials/chemical synthesis , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Line , Crystallography, X-Ray , Fluorescence Polarization , Humans , Kinetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Models, Molecular , Molecular Weight , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Structure, Secondary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/pharmacology , Structure-Activity Relationship , Terphenyl Compounds/chemical synthesis , bcl-2 Homologous Antagonist-Killer Protein , bcl-X Protein
11.
Mol Divers ; 8(2): 89-100, 2004.
Article in English | MEDLINE | ID: mdl-15209160

ABSTRACT

The design of synthetic agents to disrupt protein-protein interactions has received relatively little attention in recent years. In this review we describe strategies for targeting different types of protein surfaces using mimetics of protein secondary or tertiary structure. In this way strong and selective binding to a protein surface has be achieved and disruption of clinically important protein-protein interactions has been demonstrated in models of human disease.


Subject(s)
Biochemistry/methods , Proteins/chemistry , Proteins/metabolism , Amino Acid Sequence , Chymotrypsin/antagonists & inhibitors , Chymotrypsin/drug effects , Chymotrypsin/metabolism , Cytochrome-c Peroxidase/chemistry , Cytochrome-c Peroxidase/drug effects , Cytochrome-c Peroxidase/metabolism , Cytochromes c/chemistry , Cytochromes c/drug effects , Cytochromes c/metabolism , Molecular Mimicry , Molecular Sequence Data , Organic Chemicals/chemistry , Organic Chemicals/metabolism , Organic Chemicals/pharmacology , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/metabolism , Protein Conformation , Proteins/drug effects , Receptors, Platelet-Derived Growth Factor/chemistry , Receptors, Platelet-Derived Growth Factor/drug effects , Receptors, Platelet-Derived Growth Factor/metabolism
14.
AIDS Res Hum Retroviruses ; 18(14): 989-97, 2002 Sep 20.
Article in English | MEDLINE | ID: mdl-12396451

ABSTRACT

XTT can be metabolically reduced by mitochondrial dehydrogenase in viable cells to a water-soluble formazan product. Thus XTT has been widely used to evaluate cell viability and to screen anti-HIV agents and the cytotoxicity of these agents. The present studies demonstrated that XTT formazan derived from XTT in cell culture significantly inhibits the fusion of HIV-1-infected cells with uninfected cells. Synthetic XTT formazan effectively inhibited the replication of laboratory-adapted and primary HIV-1 isolates and cell-to-cell fusion with low cytotoxicity. It blocks the six-helix bundle formation between peptides derived from the N- and C-terminal heptad repeat regions of the gp41 ectodomain (designated N- and C-peptides, respectively). Analysis by a computer-aided docking program indicates that XTT formazan may bind to the highly conserved hydrophobic pocket on the surface of the central trimeric coiled coil of gp41. These results suggest that XTT formazan inhibits HIV-1 entry by targeting the alpha-helical coiled-coil domain of gp41. This small molecular nonpeptide antiviral compound can be used as a lead for designing more effective HIV-1 entry inhibitors targeting the fusion stage of HIV-1 infection. But because XTT formazan itself has anti-HIV-1 activity, caution should be exercised when XTT is used to evaluate HIV-1 infectivity.


Subject(s)
Formazans/pharmacology , HIV Infections/virology , HIV-1/drug effects , HIV-1/pathogenicity , Virus Replication/drug effects , Animals , Binding Sites , Cell Fusion , Cell Line , Cell Survival , Circular Dichroism , Formazans/chemistry , Formazans/metabolism , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/metabolism , Humans , Membrane Fusion/drug effects , Methylphenazonium Methosulfate/pharmacology , Mice , Models, Molecular
15.
J Am Chem Soc ; 124(40): 11838-9, 2002 Oct 09.
Article in English | MEDLINE | ID: mdl-12358513

ABSTRACT

The rational design of low-molecular weight ligands that disrupt protein-protein interactions is still a challenging goal in medicinal chemistry. Our approach to this problem involves the design of molecular scaffolds that mimic the surface functionality projected along one face of an alpha-helix. Using a terphenyl scaffold, which in a staggered conformation closely reproduces the projection of functionality on the surface of an alpha-helix, we designed mimics of the pro-apoptotic alpha-helical Bak-peptide as inhibitors of the Bak/Bcl-xL interaction. This led to the development of a potent Bcl-xL antagonist (KD = 114 nM), whose binding affinity for Bcl-xL was assessed by a fluorescence polarization assay. To determine the binding site of the developed inhibitor we used docking studies and an HSQC-NMR experiment with 15N-labeled Bcl-xL protein. These studies suggest that the inhibitor is binding in the same hydrophobic cleft as the Bak- and Bad-peptides.


Subject(s)
Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Terphenyl Compounds/chemistry , Binding Sites , Fluorescence Polarization , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Molecular Mimicry , Protein Structure, Secondary , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Terphenyl Compounds/chemical synthesis , Terphenyl Compounds/pharmacology , bcl-2 Homologous Antagonist-Killer Protein , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...