Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Antibodies (Basel) ; 9(1)2020 Jan 08.
Article in English | MEDLINE | ID: mdl-31936270

ABSTRACT

Monoclonal antibodies have evolved from research tools to powerful therapeutics in the past 30 years. Clinical success rates of antibodies have exceeded expectations, resulting in heavy investment in biologics discovery and development in addition to traditional small molecules across the industry. However, protein therapeutics cannot drug targets intracellularly and are limited to soluble and cell-surface antigens. Tremendous strides have been made in antibody discovery, protein engineering, formulation, and delivery devices. These advances continue to push the boundaries of biologics to enable antibody conjugates to take advantage of the target specificity and long half-life from an antibody, while delivering highly potent small molecule drugs. While the "magic bullet" concept produced the first wave of antibody conjugates, these entities were met with limited clinical success. This review summarizes the advances and challenges in the field to date with emphasis on antibody conjugation, linker-payload chemistry, novel payload classes, absorption, distribution, metabolism, and excretion (ADME), and product developability. We discuss lessons learned in the development of oncology antibody conjugates and look towards future innovations enabling other therapeutic indications.

2.
Br J Clin Pharmacol ; 85(5): 935-948, 2019 05.
Article in English | MEDLINE | ID: mdl-30677788

ABSTRACT

AIMS: Erythropoiesis-stimulating agents used to treat anaemia in patients with chronic kidney disease (CKD) have been associated with cardiovascular adverse events. Hepcidin production, controlled by bone morphogenic protein 6 (BMP6), regulates iron homeostasis via interactions with the iron transporter, ferroportin. High hepcidin levels are thought to contribute to increased iron sequestration and subsequent anaemia in CKD patients. To investigate alternative therapies to erythropoiesis-stimulating agents for CKD patients, monoclonal antibodies, LY3113593 and LY2928057, targeting BMP6 and ferroportin respectively, were tested in CKD patients. METHODS: Preclinical in vitro/vivo data and clinical data in healthy subjects and CKD patients were used to illustrate the translation of pharmacological properties of LY3113593 and LY2928057, highlighting the novelty of targeting these nodes within the hepcidin-ferroportin pathway. RESULTS: LY2928057 bound ferroportin and blocked interactions with hepcidin, allowing iron efflux, leading to increased serum iron and transferrin saturation levels and increased hepcidin in monkeys and humans. In CKD patients, LY2928057 led to slower haemoglobin decline and reduction in ferritin (compared to placebo). Serum iron increase was (mean [90% confidence interval]) 1.98 [1.46-2.68] and 1.36 [1.22-1.51] fold-relative to baseline following LY2928057 600 mg and LY311593 150 mg respectively in CKD patients. LY3113593 specifically blocked BMP6 binding to its receptor and produced increases in iron and transferrin saturation and decreases in hepcidin preclinically and clinically. In CKD patients, LY3113593 produced an increase in haemoglobin and reduction in ferritin (compared to placebo). CONCLUSION: LY3113593 and LY2928057 pharmacological effects (serum iron and ferritin) were translated from preclinical-to-clinical development. Such interventions may lead to new CKD anaemia treatments.


Subject(s)
Anemia/drug therapy , Hematologic Agents/pharmacology , Hepcidins/metabolism , Renal Insufficiency, Chronic/complications , Signal Transduction/drug effects , Adult , Anemia/blood , Anemia/etiology , Anemia/metabolism , Animals , Bone Morphogenetic Protein 6/antagonists & inhibitors , Bone Morphogenetic Protein 6/metabolism , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Ferritins/blood , Ferritins/metabolism , Healthy Volunteers , Hematologic Agents/therapeutic use , Hemoglobins/analysis , Humans , Iron/blood , Iron/metabolism , Macaca fascicularis , Male , Mice , Middle Aged , Rats , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/metabolism , Treatment Outcome , Young Adult
3.
MAbs ; 7(6): 1084-93, 2015.
Article in English | MEDLINE | ID: mdl-26337808

ABSTRACT

The application of protein engineering technologies toward successfully improving antibody pharmacokinetics has been challenging due to the multiplicity of biochemical factors that influence monoclonal antibody (mAb) disposition in vivo. Physiological factors including interactions with the neonatal Fc receptor (FcRn) and specific antigen binding properties of mAbs, along with biophysical properties of the mAbs themselves play a critical role. It has become evident that applying an integrated approach to understand the relative contribution of these factors is critical to rationally guide and apply engineering strategies to optimize mAb pharmacokinetics. The study presented here evaluated the influence of unintended non-specific interactions on the disposition of mAbs whose clearance rates are governed predominantly by either non-specific (FcRn) or target-mediated processes. The pharmacokinetics of 8 mAbs representing a diverse range of these properties was evaluated in cynomolgus monkeys. Results revealed complementarity-determining region (CDR) charge patch engineering to decrease charge-related non-specific binding can have a significant impact on improving the clearance. In contrast, the influence of enhanced in vitro FcRn binding was mixed, and related to both the strength of charge interaction and the general mechanism predominant in governing the clearance of the particular mAb. Overall, improved pharmacokinetics through enhanced FcRn interactions were apparent for a CDR charge-patch normalized mAb which was affected by non-specific clearance. The findings in this report are an important demonstration that mAb pharmacokinetics requires optimization on a case-by-case basis to improve the design of molecules with increased therapeutic application.


Subject(s)
Antibodies, Monoclonal, Humanized/metabolism , Complementarity Determining Regions/metabolism , Histocompatibility Antigens Class I/metabolism , Protein Engineering/methods , Receptors, Fc/metabolism , Animals , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibody Affinity/immunology , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Macaca fascicularis , Metabolic Clearance Rate , Mice , Protein Binding/immunology , Receptors, Fc/immunology
4.
Exp Neurol ; 267: 165-76, 2015 May.
Article in English | MEDLINE | ID: mdl-25771799

ABSTRACT

Several lines of evidence indicate that Glial cell line-derived neurotrophic factor (GDNF) is a trophic factor for dopaminergic neurons. Direct parenchymal administration of GDNF is robustly neuroprotective and neurorestorative in multiple neurotoxin-based animal models (rat and non-human primate (NHP)) of Parkinson's Disease (PD), suggesting its potential as a therapeutic agent. Although small, open-label clinical trials of intra-putamenal administration of bacteria-derived, full length, wild type GDNF (GDNFwt) were efficacious in improving standardized behavioral scores, a double-blinded, randomized controlled trial failed to do so. We hypothesize that the lack of clinical efficacy of GDNFwt in the larger randomized trial was due to poor bio-distribution in the putamen and/or poor chemical stability while in the delivery device for prolonged time periods at 37°C. The development of neutralizing antibodies in some patients may also have been a contributing factor. GDNFv is an engineered form of GDNFwt, expressed and purified from mammalian cells, designed to overcome these limitations, including removal of the N-terminal heparin-binding domain to improve its diffusivity in brain parenchyma by reducing its binding to extracellular matrix (ECM), and key amino acid substitutions to improve chemical stability. Intra-striatal administration of a single injection of GDNFv in the rat produced significantly greater brain distribution than GDNFwt, consistent with reduced binding to ECM. Using liquid chromatography/mass spectrometry (LS/MS) methods GDNFv was shown to have improved chemical stability compared to GDNFwt when stored at 37°C for 4weeks. In addition, GDNFv resulted in lower predicted clinical immunogenicity compared to GDNFwt, as demonstrated by reduced CD4+ T cell proliferation and reduced IL-2-induced secretion in peripheral blood mononucleated cells collected from volunteers representing the world's major histocompatibility complex (MHC) haplotypes. GDNFv was demonstrated to be pharmacologically equivalent to GDNFwt in the key parameters in vitro of GFRα1 receptor binding, c-Ret phosphorylation, neurite outgrowth, and in vivo in its ability to increase dopamine turnover (DA). GDNFv protected dopamine nerve terminals and neurons in a 6-hydroxy-dopamine (6-OHDA) rat model. In summary, we empirically demonstrate the superior properties of GDNFv compared to GDNFwt through enhanced bio-distribution and chemical stability concurrently with decreased predicted clinical immunogenicity while maintaining pharmacological and neurotrophic activity. These data indicate that GDNFv is an improved version of GDNF suitable for clinical assessment as a targeted regenerative therapy for PD.


Subject(s)
Brain/metabolism , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Immunogenetic Phenomena/genetics , Mutation/genetics , Animals , Brain/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrinolytic Agents/pharmacology , Heparin/pharmacology , Humans , Male , Nerve Tissue Proteins/metabolism , Neurites/drug effects , Oxidopamine/toxicity , PC12 Cells , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Rats , Rats, Sprague-Dawley
5.
MAbs ; 7(3): 483-93, 2015.
Article in English | MEDLINE | ID: mdl-25695748

ABSTRACT

Lowering the isoelectric point (pI) through engineering the variable region or framework of an IgG can improve its exposure and half-life via a reduction in clearance mediated through non-specific interactions. As such, net charge is a potentially important property to consider in developing therapeutic IgG molecules having favorable pharmaceutical characteristics. Frequently, it may not be possible to shift the pI of monoclonal antibodies (mAbs) dramatically without the introduction of other liabilities such as increased off-target interactions or reduced on-target binding properties. In this report, we explored the influence of more subtle modifications of molecular charge on the in vivo properties of an IgG1 and IgG4 monoclonal antibody. Molecular surface modeling was used to direct residue substitutions in the complementarity-determining regions (CDRs) to disrupt positive charge patch regions, resulting in a reduction in net positive charge without affecting the overall pI of the mAbs. The effect of balancing the net positive charge on non-specific binding was more significant for the IgG4 versus the IgG1 molecule that we examined. This differential effect was connected to the degree of influence on cellular degradation in vitro and in vivo clearance, distribution and metabolism in mice. In the more extreme case of the IgG4, balancing the charge yielded an ∼7-fold improvement in peripheral exposure, as well as significantly reduced tissue catabolism and subsequent excretion of proteolyzed products in urine. Balancing charge on the IgG1 molecule had a more subtle influence on non-specific binding and yielded only a modest alteration in clearance, distribution and elimination. These results suggest that balancing CDR charge without affecting the pI can lead to improved mAb pharmacokinetics, the magnitude of which is likely dependent on the relative influence of charge imbalance and other factors affecting the molecule's disposition.


Subject(s)
Antibodies, Monoclonal, Humanized , Antibody Specificity/genetics , Complementarity Determining Regions , Immunoglobulin G , Models, Molecular , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacology , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , Complementarity Determining Regions/pharmacology , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Isoelectric Point , Mice
6.
PLoS One ; 3(7): e2486, 2008 Jul 02.
Article in English | MEDLINE | ID: mdl-18596935

ABSTRACT

BACKGROUND: Phosphoprotein phosphatase 2A (PP2A), a major serine-threonine protein phosphatase in eukaryotes, is an oligomeric protein comprised of structural (A) and catalytic (C) subunits to which a variable regulatory subunit (B) can associate. The C subunit contains a methyl ester post-translational modification on its C-terminal leucine residue, which is removed by a specific methylesterase (PME-1). Methylesterification is thought to control the binding of different B subunits to AC dimers, but little is known about its physiological significance in vivo. METHODOLOGY/PRINCIPAL FINDINGS: Here, we show that targeted disruption of the PME-1 gene causes perinatal lethality in mice, a phenotype that correlates with a virtually complete loss of the demethylated form of PP2A in the nervous system and peripheral tissues. Interestingly, PP2A catalytic activity over a peptide substrate was dramatically reduced in PME-1(-/-) tissues, which also displayed alterations in phosphoproteome content. CONCLUSIONS: These findings suggest a role for the demethylated form of PP2A in maintenance of enzyme function and phosphorylation networks in vivo.


Subject(s)
Carboxylic Ester Hydrolases/genetics , Protein Phosphatase 2/metabolism , Animals , Catalytic Domain , Gene Targeting , Genes, Lethal , Methylation , Mice , Mice, Transgenic , Models, Genetic
7.
J Med Chem ; 50(14): 3359-68, 2007 Jul 12.
Article in English | MEDLINE | ID: mdl-17559203

ABSTRACT

A systematic study of the structure-activity relationships of 2b (OL-135), a potent inhibitor of fatty acid amide hydrolase (FAAH), is detailed targeting the C2 acyl side chain. A series of aryl replacements or substituents for the terminal phenyl group provided effective inhibitors (e.g., 5c, aryl = 1-napthyl, Ki = 2.6 nM), with 5hh (aryl = 3-ClPh, Ki = 900 pM) being 5-fold more potent than 2b. Conformationally restricted C2 side chains were examined, and many provided exceptionally potent inhibitors, of which 11j (ethylbiphenyl side chain) was established to be a 750 pM inhibitor. A systematic series of heteroatoms (O, NMe, S), electron-withdrawing groups (SO, SO2), and amides positioned within and hydroxyl substitutions on the linking side chain were investigated, which typically led to a loss in potency. The most tolerant positions provided effective inhibitors (12p, 6-position S, Ki = 3 nM, or 13d, 2-position OH, Ki = 8 nM) comparable in potency to 2b. Proteome-wide screening of selected inhibitors from the systematic series of >100 candidates prepared revealed that they are selective for FAAH over all other mammalian serine proteases.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Oxazoles/chemistry , Oxazoles/pharmacology , Amidohydrolases/metabolism , Animals , Magnetic Resonance Spectroscopy , Rats , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
8.
J Med Chem ; 50(5): 1058-68, 2007 Mar 08.
Article in English | MEDLINE | ID: mdl-17279740

ABSTRACT

A study of the structure-activity relationships (SAR) of 2f (OL-135), a potent inhibitor of fatty acid amide hydrolase (FAAH), is detailed, targeting the 5-position of the oxazole. Examination of a series of substituted benzene derivatives (12-14) revealed that the optimal position for substitution was the meta-position with selected members approaching or exceeding the potency of 2f. Concurrent with these studies, the effect of substitution on the pyridine ring of 2f was also examined. A series of small, nonaromatic C5-substituents was also explored and revealed that the K(i) follows a well-defined correlation with the Hammett sigma(p) constant (rho = 3.01, R2 = 0.91) in which electron-withdrawing substituents enhance potency, leading to inhibitors with K(i)s as low as 400 pM (20n). Proteomic-wide screening of the inhibitors revealed that most are exquisitely selective for FAAH over all other mammalian proteases, reversing the 100-fold preference of 20a (C5 substituent = H) for the enzyme TGH.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Arachidonic Acids/metabolism , Benzene Derivatives/chemical synthesis , Oleic Acids/metabolism , Oxazoles/chemical synthesis , Polyunsaturated Alkamides/metabolism , Amidohydrolases/chemistry , Animals , Benzene Derivatives/chemistry , Benzene Derivatives/pharmacology , COS Cells , Chlorocebus aethiops , Endocannabinoids , Humans , Oxazoles/chemistry , Oxazoles/pharmacology , Proteomics , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Structure-Activity Relationship
9.
Biochemistry ; 45(15): 4720-6, 2006 Apr 18.
Article in English | MEDLINE | ID: mdl-16605240

ABSTRACT

N-Acyl ethanolamines (NAEs) constitute a large and diverse class of signaling lipids that includes the endogenous cannabinoid anandamide. Like other lipid transmitters, NAEs are thought to be biosynthesized and degraded on-demand rather than being stored in vesicles prior to signaling. The identification of enzymes involved in NAE metabolism is therefore imperative to achieve a complete understanding of this lipid signaling system and control it for potential therapeutic gain. Recently, an N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD) was identified as a candidate enzyme involved in the biosynthesis of NAEs. Here, we describe the generation and characterization of mice with a targeted disruption in the NAPE-PLD gene [NAPE-PLD(-/-) mice]. Brain tissue from NAPE-PLD(-/-) mice showed more than a 5-fold reduction in the calcium-dependent conversion of NAPEs to NAEs bearing both saturated and polyunsaturated N-acyl chains. However, only the former group of NAEs was decreased in level in NAPE-PLD(-/-) brains, and these reductions were most dramatic for NAEs bearing very long acyl chains (>or=C20). Further studies identified a calcium-independent PLD activity in brains from NAPE-PLD(-/-) mice that accepted multiple NAPEs as substrates, including the anandamide precursor C20:4 NAPE. The illumination of distinct enzymatic pathways for the biosynthesis of long chain saturated and polyunsaturated NAEs suggests a strategy to control the activity of specific subsets of these lipids without globally affecting the function of the NAE family as a whole.


Subject(s)
Cannabinoid Receptor Modulators/biosynthesis , Endocannabinoids , Phospholipase D/metabolism , Animals , Brain/cytology , Brain/metabolism , Ethanolamines/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Models, Genetic , Phosphatidylethanolamines/metabolism , Phospholipase D/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 102(17): 6195-200, 2005 Apr 26.
Article in English | MEDLINE | ID: mdl-15840715

ABSTRACT

Organophosphorus (OP) insecticides and chemical warfare agents act primarily by inhibiting acetylcholinesterase. There are many secondary targets for OP toxicants as observed for example with the major insecticide chlorpyrifos and its bioactivated metabolite chlorpyrifos oxon (CPO). Therefore, it was surprising that the predominant mouse brain protein labeled in vitro by [(3)H-ethyl]CPO (1 nM) (designated CPO-binding protein or CPO-BP) is not one of these known OP toxicant targets. CPO-BP is a 50-kDa membrane-bound serine hydrolase measured by derivatization with [(3)H]CPO and SDS/PAGE or filtration binding assay. It appears to undergo rapid diethylphosphorylation by [(3)H]CPO followed by either dephosphorylation and reactivation or aging on loss of an ethyl group. CPO and several other OP toxicants potently inhibit CPO-BP in vivo (i.p., 2 h) (50% inhibition at 2-25 mg/kg) and in vitro (50% inhibition at 8-68 nM). Using three chemical labeling reagents, i.e., [(3)H]CPO and the activity-based proteomic probes fluorophosphonate-biotin and fluorophosphonate-rhodamine, mouse brain CPO-BP is identified as serine hydrolase KIAA1363 of unknown function. Brains from KIAA1363(-/-) mice show greatly reduced levels of CPO labeling and hydrolytic metabolism compared to brains from wild-type mice. KIAA1363 therefore is the principal enzyme for metabolizing low levels of CPO in brain and may play a more general role in detoxification of OP nerve poisons.


Subject(s)
Brain/enzymology , Chlorpyrifos/analogs & derivatives , Chlorpyrifos/pharmacokinetics , Chlorpyrifos/toxicity , Neurotoxins/pharmacokinetics , Animals , Cell Membrane/metabolism , Hydrolysis , Inactivation, Metabolic , Kinetics , Male , Mice , Serine Endopeptidases , Serine Proteases , Sterol Esterase , Tritium
11.
J Med Chem ; 48(6): 1849-56, 2005 Mar 24.
Article in English | MEDLINE | ID: mdl-15771430

ABSTRACT

Fatty acid amide hydrolase (FAAH) degrades neuromodulating fatty acid amides including anandamide (endogenous cannabinoid agonist) and oleamide (sleep-inducing lipid) at their sites of action and is intimately involved in their regulation. Herein we report the discovery of a potent, selective, and efficacious class of reversible FAAH inhibitors that produce analgesia in animal models validating a new therapeutic target for pain intervention. Key to the useful inhibitor discovery was the routine implementation of a proteomics-wide selectivity screen against the serine hydrolase superfamily ensuring selectivity for FAAH coupled with systematic in vivo examinations of candidate inhibitors.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesics/chemical synthesis , Ketones/chemical synthesis , Oxazoles/chemical synthesis , Pyridines/chemical synthesis , Amidohydrolases/chemistry , Analgesics/chemistry , Analgesics/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Humans , Ketones/chemistry , Ketones/pharmacology , Models, Molecular , Oxazoles/chemistry , Oxazoles/pharmacology , Proteomics , Pyridines/chemistry , Pyridines/pharmacology , Rats , Recombinant Proteins/antagonists & inhibitors , Serine Endopeptidases/chemistry , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 15(5): 1423-8, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15713400

ABSTRACT

The concurrent implementation of a proteome-wide serine hydrolase selectivity screen with traditional efforts to optimize fatty acid amide hydrolase (FAAH) inhibition potency led to the expedited discovery of a new class of exceptionally potent (Ki < 300 pM) and unusually selective (> 100-fold selective) inhibitors. The iterative inhibitor design and evaluation with assistance of the selectivity screen served to differentiate otherwise indistinguishable inhibitors permitting the simultaneous optimization of potency and selectivity. Significantly, the simultaneous assessment of all potential competitive enzymes with the selectivity screen does not require the use of expressed or purified enzymes or a competitive substrate, no modification of the inhibitors is required, and the relative potency for competitive enzymes can be quantified (IC50's) including those that lack known substrates or function.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Proteome/analysis , Proteomics/methods , Drug Evaluation, Preclinical , Molecular Structure , Structure-Activity Relationship , Substrate Specificity
13.
J Pharmacol Exp Ther ; 311(2): 441-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15229230

ABSTRACT

Fatty acid amide hydrolase (FAAH) is the primary catabolic regulator of several bioactive lipid amides in vivo, including the endogenous cannabinoid anandamide and the sleep-inducing substance oleamide. Inhibitors of FAAH are considered a potential therapeutic approach for the treatment of several nervous system disorders, including pain, anxiety, and insomnia. However, for FAAH inhibitors to achieve clinical utility, they must not only display efficacy in vivo but also selectivity for this enzyme relative to the numerous other serine hydrolases present in mammalian proteomes. Here, we report a general strategy for evaluating the pharmacological activity and target specificity of FAAH inhibitors and its implementation to develop the first class of selective reversible inhibitors of this enzyme that are highly efficacious in vivo. Using a series of functional proteomics, analytical chemistry, and behavioral pharmacology assays, we have identified a class of alpha-keto-heterocycles that show unprecedented selectivity for FAAH relative to other mammalian hydrolases, and, when administered to rodents, raise central nervous system levels of anandamide and promote cannabinoid receptor 1-dependent analgesia in several assays of pain sensation. These studies provide further evidence that FAAH may represent an attractive therapeutic target and describe a general route by which inhibitors of this enzyme can be optimized to achieve exceptional potency, selectivity, and efficacy in vivo.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Analgesia , Arachidonic Acids , Enzyme Inhibitors/pharmacology , Pyridines , Animals , Drug Synergism , Endocannabinoids , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Polyunsaturated Alkamides , Proteomics , Receptor, Cannabinoid, CB1/physiology
14.
Nat Biotechnol ; 21(6): 687-91, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12740587

ABSTRACT

To realize the promise of genomics-based therapeutics, new methods are needed to accelerate the discovery of small molecules that selectively modulate protein activity. Toward this end, advances in combinatorial synthesis have provided unprecedented access to large compound libraries of considerable structural complexity and diversity, shifting the bottleneck in drug discovery to the development of efficient screens for protein targets. Screening for reversible enzyme inhibitors typically requires extensive target-specific work, including protein expression and purification, as well as the development of specific substrate assays. Here we report a proteomic method for the discovery of reversible enzyme inhibitors that avoids these steps. We show that competitive profiling of a library of candidate serine hydrolase inhibitors in complex proteomes with activity-based chemical probes identifies nanomolar reversible inhibitors of several enzymes simultaneously, including the endocannabinoid-degrading enzyme fatty acid amide hydrolase (FAAH), triacylglycerol hydrolase (TGH) and an uncharacterized membrane-associated hydrolase that lacks known substrates. The strategy tests inhibitors against numerous enzymes in parallel, assigning both potency and selectivity factors to each agent. In this way, promiscuous inhibitors were readily rejected in favor of equally potent compounds with 500-fold or greater selectivity for their targets.


Subject(s)
Enzyme Inhibitors/analysis , Enzyme Inhibitors/chemistry , Peptide Library , Protein Array Analysis/methods , Proteome , Proteomics/methods , Amidohydrolases/antagonists & inhibitors , Amidohydrolases/chemistry , Animals , Binding, Competitive , Cannabinoid Receptor Modulators , Enzyme Activation , Enzyme Inhibitors/isolation & purification , Hydrolases/antagonists & inhibitors , Hydrolases/chemistry , Lipase/antagonists & inhibitors , Lipase/chemistry , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...