Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Proc Natl Acad Sci U S A ; 114(2): 370-375, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28011764

ABSTRACT

Glioblastoma multiforme (GBM) is an intractable tumor despite therapeutic advances, principally because of its invasive properties. Radiation is a staple in therapeutic regimens, although cells surviving radiation can become more aggressive and invasive. Subtraction hybridization identified melanoma differentiation-associated gene 9 [MDA-9/Syntenin; syndecan-binding protein (SDCBP)] as a differentially regulated gene associated with aggressive cancer phenotypes in melanoma. MDA-9/Syntenin, a highly conserved double-PDZ domain-containing scaffolding protein, is robustly expressed in human-derived GBM cell lines and patient samples, with expression increasing with tumor grade and correlating with shorter survival times and poorer response to radiotherapy. Knockdown of MDA-9/Syntenin sensitizes GBM cells to radiation, reducing postradiation invasion gains. Radiation induces Src and EGFRvIII signaling, which is abrogated through MDA-9/Syntenin down-regulation. A specific inhibitor of MDA-9/Syntenin activity, PDZ1i (113B7), identified through NMR-guided fragment-based drug design, inhibited MDA-9/Syntenin binding to EGFRvIII, which increased following radiation. Both genetic (shmda-9) and pharmacological (PDZ1i) targeting of MDA-9/Syntenin reduced invasion gains in GBM cells following radiation. Although not affecting normal astrocyte survival when combined with radiation, PDZ1i radiosensitized GBM cells. PDZ1i inhibited crucial GBM signaling involving FAK and mutant EGFR, EGFRvIII, and abrogated gains in secreted proteases, MMP-2 and MMP-9, following radiation. In an in vivo glioma model, PDZ1i resulted in smaller, less invasive tumors and enhanced survival. When combined with radiation, survival gains exceeded radiotherapy alone. MDA-9/Syntenin (SDCBP) provides a direct target for therapy of aggressive cancers such as GBM, and defined small-molecule inhibitors such as PDZ1i hold promise to advance targeted brain cancer therapy.


Subject(s)
Glioblastoma/genetics , Neoplasm Invasiveness/genetics , Syntenins/genetics , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Melanoma/genetics , Mice , Mice, Nude , PDZ Domains/genetics , Signal Transduction/genetics , src-Family Kinases/genetics
2.
Chembiochem ; 17(7): 570-5, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26895508

ABSTRACT

Cell surface p32, the target of LyP-1 homing peptide, is upregulated in tumors and atherosclerotic plaques and has been widely used as a receptor for systemic delivery of payloads. Here, we identified an improved LyP-1-mimicking peptide (TT1, CKRGARSTC). We used this peptide in a fluorescence polarization-based high-throughput screening of a 50,000-compound chemical library and identified a panel of compounds that bind p32 with low micromolar affinity. Among the hits identified in the screen, two compounds were shown to specifically bind to p32 in multiple assays. One of these compounds was chosen for an in vivo study. Nanoparticles surface-functionalized with this compound specifically adhered to surfaces coated with recombinant p32 and, when injected intravenously, homed to p32-expressing breast tumors in mice. This compound provides a lead for the development of p32-targeted affinity ligands that circumvent some of the limitations of peptide-based probes in guided drug delivery.


Subject(s)
Aminopyridines/chemistry , Breast Neoplasms/drug therapy , Drug Delivery Systems , Ethylenediamines/chemistry , Mitochondrial Proteins/administration & dosage , Peptides, Cyclic/administration & dosage , Aminopyridines/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Carrier Proteins , Cell Line, Tumor , Ethylenediamines/pharmacology , Female , Humans , Ligands , Mice , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Nanoparticles/chemistry
3.
Proc Natl Acad Sci U S A ; 108(21): 8785-90, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555592

ABSTRACT

Limited options are available for treating patients with advanced prostate cancer (PC). Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24), an IL-10 family cytokine, exhibits pleiotropic anticancer activities without adversely affecting normal cells. We previously demonstrated that suppression of the prosurvival Bcl-2 family member, myeloid cell leukemia-1 (Mcl-1), is required for mda-7/IL-24-mediated apoptosis of prostate carcinomas. Here we demonstrate that pharmacological inhibition of Mcl-1 expression with the unique Apogossypol derivative BI-97C1, also called Sabutoclax, is sufficient to sensitize prostate tumors to mda-7/IL-24-induced apoptosis, whereas ABT-737, which lacks efficacy in inhibiting Mcl-1, does not sensitize mda-7/IL-24-mediated cytotoxicity. A combination regimen of tropism-modified adenovirus delivered mda-7/IL-24 (Ad.5/3-mda-7) and BI-97C1 enhances cytotoxicity in human PC cells, including those resistant to mda-7/IL-24 or BI-97C1 alone. The combination regimen causes autophagy that facilitates NOXA- and Bim-induced and Bak/Bax-mediated mitochondrial apoptosis. Treatment with Ad.5/3-mda-7 and BI-97C1 significantly inhibits the growth of human PC xenografts in nude mice and spontaneously induced PC in Hi-myc transgenic mice. Tumor growth inhibition correlated with increased TUNEL staining and decreased Ki-67 expression in both PC xenografts and prostates of Hi-myc mice. These findings demonstrate that pharmacological inhibition of Mcl-1 with the Apogossypol derivative, BI-97C1, sensitizes human PCs to mda-7/IL-24-mediated cytotoxicity, thus potentially augmenting the therapeutic benefit of this combinatorial approach toward PC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Genetic Therapy/methods , Gossypol/analogs & derivatives , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Cell Line, Tumor , Gossypol/pharmacology , Gossypol/therapeutic use , Humans , Interleukins/administration & dosage , Male , Mice , Mice, Nude , Myeloid Cell Leukemia Sequence 1 Protein , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Transfection , Xenograft Model Antitumor Assays
4.
J Biol Chem ; 286(46): 39829-35, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21953453

ABSTRACT

Recent characterization of Mcl-1 as the primary anti-apoptotic Bcl-2 family member expressed in solid tumors, coupled with its ability to enable therapeutic resistance, has provided the impetus for further study into how Mcl-1 is involved in apoptosis signaling. Here, we employ Sabutoclax, a potent and effective Mcl-1 antagonist, as a competing agent to screen a randomized 12-residue phage display library for peptides that bind strongly to the Bcl-2 homology 3 (BH3) binding groove of Mcl-1. Although the screen identified a number of α-helical peptides with canonical BH3 domain sequences, it also isolated a pair of unique peptide sequences. These sequences exhibit a reverse organization of conserved hydrophobic and acidic residues when compared with canonical BH3 sequences, and we therefore refer to them as reverse BH3 (rBH3) peptides. Furthermore, studies of the rBH3 peptides using NMR spectroscopy, fluorescence polarization displacement assays, and alanine scanning data all suggest that they bind to the BH3 binding groove of Mcl-1 selectively over Bcl-x(L). A search for proteins containing the rBH3 motif has identified a number of interesting Mcl-1 protein partners, some of which have previously been associated with apoptosis regulation involving Mcl-1. These findings provide insights into the development of more specific Mcl-1 antagonists and open the way to the identification of a previously unknown family of apoptosis-regulating and Mcl-1 interacting proteins.


Subject(s)
Peptide Fragments/chemistry , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins/chemistry , Amino Acid Motifs , Animals , Cell Line, Tumor , Hydrophobic and Hydrophilic Interactions , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Structure-Activity Relationship , bcl-X Protein/chemistry , bcl-X Protein/genetics , bcl-X Protein/metabolism
5.
Sci Rep ; 12(1): 14561, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36028520

ABSTRACT

Anti-IL17A therapies have proven effective for numerous inflammatory diseases including psoriasis, axial spondylitis and psoriatic arthritis. Modulating and/or antagonizing protein-protein interactions of IL17A cytokine binding to its cell surface receptors with oral therapies offers the promise to bring forward biologics-like efficacy in a pill to patients. We used an NMR-based fragment screen of recombinant IL17A to uncover starting points for small molecule IL17A antagonist discovery. By examining chemical shift perturbations in 2D [1H, 13C-HSQC] spectra of isotopically labeled IL17A, we discovered fragments binding the cytokine at a previously undescribed site near the IL17A C-terminal region, albeit with weak affinity (> 250 µM). Importantly this binding location was distinct from previously known chemical matter modulating cytokine responses. Subsequently through analog screening, we identified related compounds that bound symmetrically in this novel site with two copies. From this observation we employed a linking strategy via structure-based drug design and obtained compounds with increased binding affinity (< 50 nM) and showed functional inhibition of IL17A-induced cellular signaling (IC50~1 µM). We also describe a fluorescence-based probe molecule suitable to discern/screen for additional molecules binding in this C-terminal site.


Subject(s)
Arthritis, Psoriatic , Axial Spondyloarthritis , Interleukin-17 , Psoriasis , Cytokines , Drug Design , Humans , Interleukin-17/antagonists & inhibitors
6.
Mol Cancer Ther ; 8(4): 904-13, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19372563

ABSTRACT

Guided by a combination of nuclear magnetic resonance binding assays and computational docking studies, we synthesized a library of 5,5' substituted Apogossypol derivatives as potent Bcl-XL antagonists. Each compound was subsequently tested for its ability to inhibit Bcl-XL in an in vitro fluorescence polarization competition assay and exert single-agent proapoptotic activity in human cancer cell lines. The most potent compound BI79D10 binds to Bcl-XL, Bcl-2, and Mcl-1 with IC50 values of 190, 360, and 520 nmol/L, respectively, and potently inhibits cell growth in the H460 human lung cancer cell line with an EC50 value of 680 nmol/L, expressing high levels of Bcl-2. BI79D10 also effectively induces apoptosis of the RS11846 human lymphoma cell line in a dose-dependent manner and shows little cytotoxicity against bax-/-bak-/- mouse embryonic fibroblast cells, in which antiapoptotic Bcl-2 family proteins lack a cytoprotective phenotype, implying that BI79D10 has little off-target effects. BI79D10 displays in vivo efficacy in transgenic mice, in which Bcl-2 is overexpressed in splenic B cells. Together with its improved plasma and microsomal stability relative to Apogossypol, BI79D10 represents a lead compound for the development of novel apoptosis-based therapies for cancer.


Subject(s)
Gossypol/analogs & derivatives , Lung Neoplasms/drug therapy , Lymphoma/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Cell Survival/drug effects , Female , Fluorescence Polarization , Gossypol/chemical synthesis , Gossypol/chemistry , Gossypol/pharmacology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lymphoma/metabolism , Lymphoma/pathology , Magnetic Resonance Spectroscopy , Male , Membrane Proteins/metabolism , Membranes, Artificial , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Microsomes, Liver , Models, Molecular , Myeloid Cell Leukemia Sequence 1 Protein , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Tumor Cells, Cultured , bcl-2 Homologous Antagonist-Killer Protein/physiology , bcl-2-Associated X Protein/physiology , bcl-X Protein/metabolism
7.
J Med Chem ; 50(26): 6607-17, 2007 Dec 27.
Article in English | MEDLINE | ID: mdl-18038971

ABSTRACT

Peptidyl-prolyl cis-trans isomerases are a group of cytosolic enzymes initially characterized by their ability to catalyze the cis-trans isomerization of peptidyl-prolyl bonds. This represents a significant event for protein folding because cis-proline introduces critical bends within the protein conformation. FK506-binding proteins (FKBPs) represent one of the three families of enzymes sharing peptidyl-prolyl cis-trans isomerase activity. Inhibitors of FKBP12, in particular, have potent neurotrophic properties both in vivo and in vitro. Here, we describe a fragment-based unbiased nuclear magnetic resonance drug discovery approach for the identification of novel classes of chemical inhibitors against FKBP12. Compared to FK506, the fragment-based FKBP12 inhibitors developed herein possess significant advantages as drug candidates.


Subject(s)
Morpholines/chemical synthesis , Tacrolimus Binding Protein 1A/antagonists & inhibitors , Animals , Cell Line , Drug Design , Humans , In Vitro Techniques , Magnetic Resonance Spectroscopy , Microsomes, Liver/metabolism , Models, Molecular , Morpholines/chemistry , Morpholines/pharmacology , Neurites/drug effects , Neurites/physiology , Rats , Rats, Long-Evans , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Structure-Activity Relationship , Tacrolimus/chemistry , Tacrolimus/pharmacology , Tacrolimus Binding Protein 1A/chemistry
8.
Cell Chem Biol ; 24(3): 293-305, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28196613

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a progressive degenerative disease that affects motor neurons. Recent studies identified the receptor tyrosine kinase EphA4 as a disease-modifying gene that is critical for the progression of motor neuron degeneration. We report on the design and characterization of a family of EphA4 targeting agents that bind to its ligand binding domain with nanomolar affinity. The molecules exhibit excellent selectivity and display efficacy in a SOD1 mutant mouse model of ALS. Interestingly, the molecules appear to act as agonists for the receptor in certain surrogate cellular assays. While the exact mechanisms responsible for the therapeutic effect of the new agonists remain to be elucidated, we believe that the described agent represents both an invaluable pharmacological tool to further decipher the role of the EphA4 in ALS and potentially other human diseases, and a significant stepping stone for the development of novel treatments.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Receptor, EphA4/agonists , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Binding Sites , Cells, Cultured , Disease Models, Animal , Drug Design , Half-Life , Humans , Ligands , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Docking Simulation , Protein Binding , Protein Isoforms/metabolism , Protein Structure, Tertiary , Receptor, EphA4/chemistry , Receptor, EphA4/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Small Molecule Libraries/pharmacokinetics , Small Molecule Libraries/therapeutic use , Structure-Activity Relationship , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
9.
ChemMedChem ; 11(8): 919-27, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26592695

ABSTRACT

Recently we described a novel approach, named high-throughput screening (HTS) by NMR that allows the identification, from large combinatorial peptide libraries, of potent and selective peptide mimetics against a given target. Here, we deployed the "HTS by NMR" approach for the design of novel peptoid sequences targeting the N-terminal domain of Yersinia outer protein H (YopH-NT), a bacterial toxin essential for the virulence of Yersinia pestis. We aimed at disrupting the protein-protein interactions between YopH-NT and its cellular substrates, with the goal of inhibiting indirectly YopH enzymatic function. These studies resulted in a novel agent of sequence Ac-F-pY-cPG-d-P-NH2 (pY=phosphotyrosine; cPG=cyclopentyl glycine) with a Kd value against YopH-NT of 310 nm. We demonstrated that such a pharmacological inhibitor of YopH-NT results in the inhibition of the dephosphorylation by full-length YopH of a cellular substrate. Hence, potentially this agent represents a valuable stepping stone for the development of novel therapeutics against Yersinia infections. The data reported further demonstrate the utility of the HTS by NMR approach in deriving novel peptide mimetics targeting protein-protein interactions.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/metabolism , High-Throughput Screening Assays , Peptoids/pharmacology , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Nuclear Magnetic Resonance, Biomolecular , Peptoids/chemical synthesis , Peptoids/chemistry , Plague/drug therapy , Protein Binding/drug effects , Protein Domains/drug effects , Structure-Activity Relationship , Yersinia pestis/drug effects
10.
Chem Biol Drug Des ; 86(4): 663-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25676805

ABSTRACT

The emergence of drug-resistant strains of influenza virus makes exploring new classes of inhibitors that target universally conserved viral targets a highly important goal. The influenza A viral genome is made up of eight single-stranded RNA-negative segments. The RNA promoter, consisting of the conserved sequences at the 3' and 5' end of each RNA genomic segment, is universally conserved among influenza A virus strains and in all segments. Previously, we reported on the identification and NMR structure of DPQ (6,7-dimethoxy-2-(1-piperazinyl)-4-quinazolinamine) (compound 1) in complex with the RNA promoter. Here, we report on additional screening and SAR studies with compound 1, including ex vivo anti-influenza activity assays, resulted in improved cellular activity against influenza A virus in the micromolar range.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Influenza A virus/genetics , Piperazines/pharmacology , Promoter Regions, Genetic , Quinazolines/pharmacology , RNA, Viral/drug effects , Animals , Dogs , Drug Evaluation, Preclinical/methods , Influenza A virus/drug effects , Madin Darby Canine Kidney Cells/drug effects , Madin Darby Canine Kidney Cells/virology , Magnetic Resonance Spectroscopy , Molecular Structure , Peptide Library , Piperazines/chemistry , Quinazolines/chemistry , Structure-Activity Relationship , Virus Replication/drug effects
11.
Curr Top Med Chem ; 15(20): 2032-42, 2015.
Article in English | MEDLINE | ID: mdl-25986689

ABSTRACT

In recent years the ever so complex field of drug discovery has embraced novel design strategies based on biophysical fragment screening (fragment-based drug design; FBDD) using nuclear magnetic resonance spectroscopy (NMR) and/or structure-guided approaches, most often using X-ray crystallography and computer modeling. Experience from recent years unveiled that these methods are more effective and less prone to artifacts compared to biochemical high-throughput screening (HTS) of large collection of compounds in designing protein inhibitors. Hence these strategies are increasingly becoming the most utilized in the modern pharmaceutical industry. Nonetheless, there is still an impending need to develop innovative and effective strategies to tackle other more challenging targets such as those involving protein-protein interactions (PPIs). While HTS strategies notoriously fail to identify viable hits against such targets, few successful examples of PPIs antagonists derived by FBDD strategies exist. Recently, we reported on a new strategy that combines some of the basic principles of fragment-based screening with combinatorial chemistry and NMR-based screening. The approach, termed HTS by NMR, combines the advantages of combinatorial chemistry and NMR-based screening to rapidly and unambiguously identify bona fide inhibitors of PPIs. This review will reiterate the critical aspects of the approach with examples of possible applications.


Subject(s)
Drug Discovery , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Receptor, EphA4/chemistry , Small Molecule Libraries/chemistry , X-Linked Inhibitor of Apoptosis Protein/chemistry , Combinatorial Chemistry Techniques , Crystallography, X-Ray , Ephrin-A5/chemistry , High-Throughput Screening Assays , Humans , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Peptides/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Receptor, EphA4/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors
12.
Chem Biol ; 22(7): 876-887, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26165155

ABSTRACT

The development of novel, targeted delivery agents for anti-cancer therapies requires the design and optimization of potent and selective tumor-targeting agents that are stable and amenable to conjugation with chemotherapeutic drugs. While short peptides represent potentially an excellent platform for these purposes, they often get degraded and are eliminated too rapidly in vivo. In this study, we used a combination of nuclear magnetic resonance-guided structure-activity relationships along with biochemical and cellular studies to derive a novel tumor-homing agent, named 123B9, targeting the EphA2 tyrosine kinase receptor ligand-binding domain. Conjugating 123B9 to the chemotherapeutic drug paclitaxel (PTX) via a stable linker results in an agent that is significantly more effective than the unconjugated drug in both a pancreatic cancer xenograft model and a melanoma lung colonization and metastases model. Hence, 123B9 could represent a promising strategy for the development of novel targeted therapies for cancer.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Paclitaxel/analogs & derivatives , Receptor, EphA2/agonists , Amino Acid Sequence , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Cell Survival/drug effects , Drug Delivery Systems/methods , Female , Humans , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Mice , Mice, Nude , Models, Animal , Molecular Targeted Therapy , Paclitaxel/chemistry , Paclitaxel/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Rats , Receptor, EphA2/chemistry , Xenograft Model Antitumor Assays
13.
Pharmacogenetics ; 14(8): 557-67, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15284538

ABSTRACT

We found a significant inverse relationship between gamma-glutamyl hydrolase (GGH) activity and the accumulation of long-chain methotrexate polyglutamates (MTXPG4-7) in non-hyperdiploid B-lineage acute lymphoblastic leukaemia (ALL) cells after uniform treatment with high-dose methotrexate (HDMTX) (1 g/m i.v.). To identify genetic polymorphisms that alter the function of human GGH, we sequenced the GGH exons of genomic DNA from children with ALL, who had a 7.8-fold range of GGH activity in their ALL cells at diagnosis. A single nucleotide polymorphism (452C>T, T127I) was found among patients with low GGH activity, but not found in patients with high GGH activity. Computational modelling indicated that the T127I substitution alters the molecular surface conformation at the catalytic cleft-tail on GGH, which is predicted to alter binding affinity with long chain but not short-chain methotrexate polyglutamates. Enzyme kinetic analysis of heterologously expressed GGH revealed a significantly higher Km (2.7-fold) and lower catalytic efficiency (Vmax/Km reduced 67%) of the T127I variant compared to wild-type GGH using long-chain MTXPG5 as substrate, but not a significant change with short-chain MTXPG2. The 452C>T single nucleotide polymorphism (SNP) was also associated with lower GGH activity in hyperdiploid B-lineage and T lineage ALL cells. Caucasians [10.0%; 95% confidence interval (CI) 6.7-13.3%; n = 155] were found to have a significantly higher frequency of the Ile allele than African-Americans (4.4%; 95% CI 1.2-7.5%; n = 80) (P = 0.033). These studies demonstrate a substrate specific functional SNP (452C>T) in the human GGH gene that is associated with lower catalytic activity and higher accumulation of long-chain MTX-PG in leukaemia cells of patients treated with HDMTX.


Subject(s)
Methotrexate/analogs & derivatives , Methotrexate/metabolism , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/metabolism , Polymorphism, Single Nucleotide/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , gamma-Glutamyl Hydrolase/genetics , Catalysis , Cloning, Molecular , Diploidy , Humans , Kinetics , Mutagenesis, Site-Directed , Substrate Specificity
14.
Chem Biol ; 20(1): 19-33, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23352136

ABSTRACT

Fragment-based ligand design (FBLD) approaches have become more widely used in drug discovery projects from both academia and industry, and are even often preferred to traditional high-throughput screening (HTS) of large collection of compounds (>10(5)). A key advantage of FBLD approaches is that these often rely on robust biophysical methods such as NMR spectroscopy for detection of ligand binding, hence are less prone to artifacts that too often plague the results from HTS campaigns. In this article, we introduce a screening strategy that takes advantage of both the robustness of protein NMR spectroscopy as the detection method, and the basic principles of combinatorial chemistry to enable the screening of large libraries of fragments (>10(5) compounds) preassembled on a common backbone. We used the method to identify compounds that target protein-protein interactions.


Subject(s)
Combinatorial Chemistry Techniques/methods , Drug Design , Magnetic Resonance Spectroscopy/methods , Receptor, EphA4/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , High-Throughput Screening Assays/methods , Humans , Ligands , Models, Molecular , Protein Structure, Tertiary , Receptor, EphA4/chemistry , Structure-Activity Relationship , X-Linked Inhibitor of Apoptosis Protein/chemistry , X-Linked Inhibitor of Apoptosis Protein/metabolism
15.
Chem Biol ; 20(8): 973-82, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23891150

ABSTRACT

The E3 ubiquitin ligase Siah regulates key cellular events that are central to cancer development and progression. A promising route to Siah inhibition is disrupting its interactions with adaptor proteins. However, typical of protein-protein interactions, traditional unbiased approaches to ligand discovery did not produce viable hits against this target, despite considerable effort and a multitude of approaches. Ultimately, a rational structure-based design strategy was successful for the identification of Siah inhibitors in which peptide binding drives specific covalent bond formation with the target. X-ray crystallography, mass spectrometry, and functional data demonstrate that these peptide mimetics are efficient covalent inhibitors of Siah and antagonize Siah-dependent regulation of Erk and Hif signaling in the cell. The proposed strategy may result useful as a general approach to the design of peptide-based inhibitors of other protein-protein interactions.


Subject(s)
Drug Design , Enzyme Inhibitors/chemistry , Nuclear Proteins/antagonists & inhibitors , Peptides/chemistry , Peptidomimetics/chemistry , Ubiquitin-Protein Ligases/antagonists & inhibitors , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Peptides/pharmacology , Peptidomimetics/pharmacology , Protein Interaction Maps/drug effects , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
16.
Clin Cancer Res ; 19(1): 128-37, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23155185

ABSTRACT

PURPOSE: YSA is an EphA2-targeting peptide that effectively delivers anticancer agents to prostate cancer tumors. Here, we report on how we increased the drug-like properties of this delivery system. EXPERIMENTAL DESIGN: By introducing non-natural amino acids, we have designed two new EphA2 targeting peptides: YNH, where norleucine and homoserine replace the two methionine residues of YSA, and dYNH, where a D-tyrosine replaces the L-tyrosine at the first position of the YNH peptide. We describe the details of the synthesis of YNH and dYNH paclitaxel conjugates (YNH-PTX and dYNH-PTX) and their characterization in cells and in vivo. RESULTS: dYNH-PTX showed improved stability in mouse serum and significantly reduced tumor size in a prostate cancer xenograft model and also reduced tumor vasculature in a syngeneic orthotopic allograft mouse model of renal cancer compared with vehicle or paclitaxel treatments. CONCLUSION: This study reveals that targeting EphA2 with dYNH drug conjugates could represent an effective way to deliver anticancer agents to a variety of tumor types.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Drug Delivery Systems , Neoplasms/genetics , Paclitaxel/administration & dosage , Peptides , Receptor, EphA2/genetics , Animals , Antineoplastic Agents, Phytogenic/chemistry , Disease Models, Animal , Female , Gene Expression , Humans , Male , Mice , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Paclitaxel/chemistry , Peptides/chemistry , Receptor, EphA2/metabolism , Transplantation, Homologous , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
17.
ChemMedChem ; 7(12): 2227-35, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23139022

ABSTRACT

Successful replication of the influenza A virus requires both viral proteins and host cellular factors. In this study we used a cellular assay to screen for small molecules capable of interfering with any of such necessary viral or cellular components. We used an established reporter assay to assess influenza viral replication by monitoring the activity of co-expressed luciferase. We screened a diverse chemical compound library, resulting in the identification of compound 7, which inhibits a novel yet elusive target. Quantitative real-time PCR studies confirmed the dose-dependent inhibitory activity of compound 7 in a viral replication assay. Furthermore, we showed that compound 7 is effective in rescuing high-dose influenza infection in an in vivo mouse model. As oseltamivir-resistant influenza strains emerge, compound 7 could be further investigated as a new and potentially suitable scaffold for the development of anti-influenza agents that act on novel targets.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Influenza A Virus, H1N1 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Small Molecule Libraries/chemistry , Small Molecule Libraries/therapeutic use , Virus Replication/drug effects , Animals , Antiviral Agents/pharmacology , Female , Humans , Influenza A Virus, H1N1 Subtype/physiology , Influenza, Human/drug therapy , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/virology , Real-Time Polymerase Chain Reaction , Small Molecule Libraries/pharmacology , Tetrazoles/chemistry , Tetrazoles/pharmacology , Tetrazoles/therapeutic use
18.
Anticancer Agents Med Chem ; 12(9): 1143-55, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22931411

ABSTRACT

Structure-based modeling combined with rational drug design, and high throughput screening approaches offer significant potential for identifying and developing lead compounds with therapeutic potential. The present review focuses on these two approaches using explicit examples based on specific derivatives of Gossypol generated through rational design and applications of a cancer-specificpromoter derived from Progression Elevated Gene-3. The Gossypol derivative Sabutoclax (BI-97C1) displays potent anti-tumor activity against a diverse spectrum of human tumors. The model of the docked structure of Gossypol bound to Bcl-XL provided a virtual structure-activity-relationship where appropriate modifications were predicted on a rational basis. These structure-based studies led to the isolation of Sabutoclax, an optically pure isomer of Apogossypol displaying superior efficacy and reduced toxicity. These studies illustrate the power of combining structure-based modeling with rational design to predict appropriate derivatives of lead compounds to be empirically tested and evaluated for bioactivity. Another approach to cancer drug discovery utilizes a cancer-specific promoter as readouts of the transformed state. The promoter region of Progression Elevated Gene-3 is such a promoter with cancer-specific activity. The specificity of this promoter has been exploited as a means of constructing cancer terminator viruses that selectively kill cancer cells and as a systemic imaging modality that specifically visualizes in vivo cancer growth with no background from normal tissues. Screening of small molecule inhibitors that suppress the Progression Elevated Gene-3-promoter may provide relevant lead compounds for cancer therapy that can be combined with further structure-based approaches leading to the development of novel compounds for cancer therapy.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Design , Drug Screening Assays, Antitumor/methods , Gossypol/analogs & derivatives , Gossypol/pharmacology , Neoplasms/drug therapy , Animals , Drug Screening Assays, Antitumor/economics , High-Throughput Screening Assays , Humans , Neoplasms/genetics , Promoter Regions, Genetic/drug effects
19.
J Med Chem ; 54(17): 6000-13, 2011 Sep 08.
Article in English | MEDLINE | ID: mdl-21797225

ABSTRACT

Overexpression of antiapoptotic members of the Bcl-2 family proteins, such as Bcl-x(L) and Mfl-1, has been shown to be involved in resistance to chemotherapeutic drugs in many forms of cancers. Recent efforts from the Abbott Laboratories resulted in the development of the acylsulfonamide compound and clinical candidate that targets selectively Bcl-2, Bcl-x(L), and Bcl-w while it is not active against Mcl-1 and Bfl-1. However, early clinical and preclinical studies suggest that pan-Bcl-2 antagonists, targeting simultaneously Mcl-1, Bcl-xL, and possibly all other four antiapoptotic Bcl-2 proteins, may result in more efficacious drugs. Here, following an NMR fragment-based approach, SAR by ILOEs, we report on compounds that exhibit nanomolar affinities for both Bcl-x(L) and Mcl-1 in vitro. We believe that these molecules can be used as useful starting point for the development of novel Bcl-2 antagonists, in particular targeting Mcl-1.


Subject(s)
Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/chemistry , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors , Acylation , Humans , Magnetic Resonance Spectroscopy , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/metabolism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , bcl-X Protein/metabolism
20.
Chem Biol Drug Des ; 78(4): 667-78, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21791013

ABSTRACT

Eph receptor tyrosine kinases and ephrin ligands control many physiological and pathological processes, and molecules interfering with their interaction are useful probes to elucidate their complex biological functions. Moreover, targeting Eph receptors might enable new strategies to inhibit cancer progression and pathological angiogenesis as well as promote nerve regeneration. Because our previous work suggested the importance of the salicylic acid group in antagonistic small molecules targeting Eph receptors, we screened a series of salicylic acid derivatives to identify novel Eph receptor antagonists. This identified a disalicylic acid-furanyl derivative that inhibits ephrin-A5 binding to EphA4 with an IC(50) of 3 µm in ELISAs. This compound, which appears to bind to the ephrin-binding pocket of EphA4, also targets several other Eph receptors. Furthermore, it inhibits EphA2 and EphA4 tyrosine phosphorylation in cells stimulated with ephrin while not affecting phosphorylation of EphB2, which is not a target receptor. In endothelial cells, the disalicylic acid-furanyl derivative inhibits EphA2 phosphorylation in response to TNFα and capillary-like tube formation on Matrigel, two effects that depend on EphA2 interaction with endogenous ephrin-A1. These findings suggest that salicylic acid derivatives could be used as starting points to design new small molecule antagonists of Eph receptors.


Subject(s)
Ephrins/metabolism , Protein Binding/drug effects , Receptors, Eph Family/antagonists & inhibitors , Receptors, Eph Family/metabolism , Salicylates/chemistry , Salicylates/pharmacology , Animals , Binding Sites , Cell Line , Haplorhini , Humans , Mice , Models, Molecular , Receptors, Eph Family/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL