Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
BMC Mol Cell Biol ; 20(1): 37, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31455205

ABSTRACT

BACKGROUND: The EP4 prostanoid receptor is one of four GPCRs that mediate the diverse actions of prostaglandin E2 (PGE2). Novel selective EP4 receptor agonists would assist to further elucidate receptor sub-type function and promote development of therapeutics for bone healing, heart failure, and other receptor associated conditions. The rat EP4 (rEP4) receptor has been used as a surrogate for the human EP4 (hEP4) receptor in multiple SAR studies. To better understand the validity of this traditional approach, homology models were generated by threading for both receptors using the RaptorX server. These models were fit to an implicit membrane using the PPM server and OPM database with refinement of intra and extracellular loops by Prime (Schrödinger). To understand the interaction between the receptors and known agonists, induced-fit docking experiments were performed using Glide and Prime (Schrödinger), with both endogenous agonists and receptor sub-type selective, small-molecule agonists. The docking scores and observed interactions were compared with radioligand displacement experiments and receptor (rat & human) activation assays monitoring cAMP. RESULTS: Rank-ordering of in silico compound docking scores aligned well with in vitro activity assay EC50 and radioligand binding Ki. We observed variations between rat and human EP4 binding pockets that have implications in future small-molecule receptor-modulator design and SAR, specifically a S103G mutation within the rEP4 receptor. Additionally, these models helped identify key interactions between the EP4 receptor and ligands including PGE2 and several known sub-type selective agonists while serving as a marked improvement over the previously reported models. CONCLUSIONS: This work has generated a set of novel homology models of the rEP4 and hEP4 receptors. The homology models provide an improvement upon the previously reported model, largely due to improved solvation. The hEP4 docking scores correlates best with the cAMP activation data, where both data sets rank order Rivenprost>CAY10684 > PGE1 ≈ PGE2 > 11-deoxy-PGE1 ≈ 11-dexoy-PGE2 > 8-aza-11-deoxy-PGE1. This rank-ordering matches closely with the rEP4 receptor as well. Species-specific differences were noted for the weak agonists Sulprostone and Misoprostol, which appear to dock more readily within human receptor versus rat receptor.


Subject(s)
Models, Molecular , Receptors, Prostaglandin E, EP4 Subtype/chemistry , Structural Homology, Protein , Amino Acid Sequence , Animals , Decapodiformes , Dinoprostone/analogs & derivatives , Dinoprostone/chemistry , Humans , Ligands , Molecular Docking Simulation , Rats , Receptors, Prostaglandin E, EP4 Subtype/agonists , Rhodopsin/chemistry
2.
ChemMedChem ; 14(16): 1560-1572, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31283109

ABSTRACT

UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase (LpxC) is a Zn2+ deacetylase that is essential for the survival of most pathogenic Gram-negative bacteria. ACHN-975 (N-((S)-3-amino-1-(hydroxyamino)-3-methyl-1-oxobutan-2-yl)-4-(((1R,2R)-2-(hydroxymethyl)cyclopropyl)buta-1,3-diyn-1-yl)benzamide) was the first LpxC inhibitor to reach human clinical testing and was discovered to have a dose-limiting cardiovascular toxicity of transient hypotension without compensatory tachycardia. Herein we report the effort beyond ACHN-975 to discover LpxC inhibitors optimized for enzyme potency, antibacterial activity, pharmacokinetics, and cardiovascular safety. Based on its overall profile, compound 26 (LPXC-516, (S)-N-(2-(hydroxyamino)-1-(3-methoxy-1,1-dioxidothietan-3-yl)-2-oxoethyl)-4-(6-hydroxyhexa-1,3-diyn-1-yl)benzamide) was chosen for further development. A phosphate prodrug of 26 was developed that provided a solubility of >30 mg mL-1 for parenteral administration and conversion into the active drug with a t1/2 of approximately two minutes. Unexpectedly, and despite our optimization efforts, the prodrug of 26 still possesses a therapeutic window insufficient to support further clinical development.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Diynes/pharmacology , Enzyme Inhibitors/pharmacology , Heart/drug effects , Hydroxamic Acids/pharmacology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/toxicity , Bacterial Proteins/antagonists & inhibitors , Cardiotoxicity , Diynes/chemical synthesis , Diynes/pharmacokinetics , Diynes/toxicity , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/toxicity , Hydroxamic Acids/chemical synthesis , Hydroxamic Acids/pharmacokinetics , Hydroxamic Acids/toxicity , Male , Molecular Structure , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/toxicity , Pseudomonas aeruginosa/drug effects , Rats, Sprague-Dawley , Structure-Activity Relationship
3.
J Med Chem ; 62(9): 4731-4741, 2019 05 09.
Article in English | MEDLINE | ID: mdl-30964292

ABSTRACT

A series of small-molecule full agonists of the prostaglandin E2 type 4 (EP4) receptor have been generated and evaluated for binding affinity and cellular potency. KMN-80 and its gem-difluoro analog KMN-159 possess high selectivity relative to other prostanoid receptors. Difluoro substitution is positioned alpha to the lactam ring carbonyl and results in KMN-159's fivefold increase in potency versus KMN-80. The two analogs exhibit electronic and conformational variations, including altered nitrogen hybridization and lactam ring puckering, that may drive the observed difluoro-associated increased potency within this four-compound series.


Subject(s)
Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Heptanoic Acids/pharmacology , Lactams/pharmacology , Pyrrolidines/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/agonists , Alprostadil/metabolism , Animals , Binding Sites , CHO Cells , Caco-2 Cells , Cricetulus , Humans , Lactams/chemical synthesis , Lactams/metabolism , Models, Chemical , Molecular Docking Simulation , Molecular Structure , Quantum Theory , Receptors, Prostaglandin E, EP3 Subtype/chemistry , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/chemistry , Receptors, Prostaglandin E, EP4 Subtype/metabolism
4.
Biochemistry ; 57(47): 6604-6614, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30365304

ABSTRACT

Pancreatic cancer cells are characterized by deregulated metabolic programs that facilitate growth and resistance to oxidative stress. Among these programs, pancreatic cancers preferentially utilize a metabolic pathway through the enzyme aspartate aminotransferase 1 [also known as glutamate oxaloacetate transaminase 1 (GOT1)] to support cellular redox homeostasis. As such, small molecule inhibitors that target GOT1 could serve as starting points for the development of new therapies for pancreatic cancer. We ran a high-throughput screen for inhibitors of GOT1 and identified a small molecule, iGOT1-01, with in vitro GOT1 inhibitor activity. Application in pancreatic cancer cells revealed metabolic and growth inhibitory activity reflecting a promiscuous inhibitory profile. We then performed an in silico docking analysis to study inhibitor-GOT1 interactions with iGOT1-01 analogues that possess improved solubility and potency properties. These results suggested that the GOT1 inhibitor competed for binding to the pyridoxal 5-phosphate (PLP) cofactor site of GOT1. To analyze how the GOT1 inhibitor bound to GOT1, a series of GOT1 mutant enzymes that abolished PLP binding were generated. Application of the mutants in X-ray crystallography and thermal shift assays again suggested but were unable to formally conclude that the GOT1 inhibitor bound to the PLP site. Mutational studies revealed the relationship between PLP binding and the thermal stability of GOT1 while highlighting the essential nature of several residues for GOT1 catalytic activity. Insight into the mode of action of GOT1 inhibitors may provide leads to the development of drugs that target redox balance in pancreatic cancer.


Subject(s)
Aspartate Aminotransferase, Cytoplasmic/antagonists & inhibitors , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Mutation , Pancreatic Neoplasms/pathology , Aspartate Aminotransferase, Cytoplasmic/genetics , Aspartate Aminotransferase, Cytoplasmic/metabolism , Binding Sites , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Humans , Metabolomics , Models, Molecular , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Protein Conformation , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Sci Transl Med ; 10(431)2018 03 07.
Article in English | MEDLINE | ID: mdl-29515000

ABSTRACT

Glycogen synthase kinase 3 (GSK3), a key regulatory kinase in the wingless-type MMTV integration site family (WNT) pathway, is a therapeutic target of interest in many diseases. Although dual GSK3α/ß inhibitors have entered clinical trials, none has successfully translated to clinical application. Mechanism-based toxicities, driven in part by the inhibition of both GSK3 paralogs and subsequent ß-catenin stabilization, are a concern in the translation of this target class because mutations and overexpression of ß-catenin are associated with many cancers. Knockdown of GSK3α or GSK3ß individually does not increase ß-catenin and offers a conceptual resolution to targeting GSK3: paralog-selective inhibition. However, inadequate chemical tools exist. The design of selective adenosine triphosphate (ATP)-competitive inhibitors poses a drug discovery challenge due to the high homology (95% identity and 100% similarity) in this binding domain. Taking advantage of an Asp133→Glu196 "switch" in their kinase hinge, we present a rational design strategy toward the discovery of paralog-selective GSK3 inhibitors. These GSK3α- and GSK3ß-selective inhibitors provide insights into GSK3 targeting in acute myeloid leukemia (AML), where GSK3α was identified as a therapeutic target using genetic approaches. The GSK3α-selective compound BRD0705 inhibits kinase function and does not stabilize ß-catenin, mitigating potential neoplastic concerns. BRD0705 induces myeloid differentiation and impairs colony formation in AML cells, with no apparent effect on normal hematopoietic cells. Moreover, BRD0705 impairs leukemia initiation and prolongs survival in AML mouse models. These studies demonstrate feasibility of paralog-selective GSK3α inhibition, offering a promising therapeutic approach in AML.


Subject(s)
Enzyme Inhibitors/therapeutic use , Glycogen Synthase Kinase 3/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Dipeptides/chemistry , Dipeptides/metabolism , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3/metabolism , Humans , Mutagenesis, Site-Directed , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , U937 Cells , beta Catenin/genetics , beta Catenin/metabolism
6.
Bioorg Med Chem Lett ; 27(3): 413-419, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28041831

ABSTRACT

As a follow-up to the discovery of our spirocyclic proline-based TPH1 inhibitor lead, we describe the optimization of this scaffold. Through a combination of X-ray co-crystal structure guided design and an in vivo screen, new substitutions in the lipophilic region of the inhibitors were identified. This effort led to new TPH1 inhibitors with in vivo efficacy when dosed as their corresponding ethyl ester prodrugs. In particular, 15b (KAR5585), the prodrug of the potent TPH1 inhibitor 15a (KAR5417), showed robust reduction of intestinal serotonin (5-HT) levels in mice. Furthermore, oral administration of 15b generated high and sustained systemic exposure of the active parent 15a in rats and dogs. KAR5585 was selected for further pharmacological evaluation in disease models associated with a dysfunctional peripheral 5-HT system.


Subject(s)
Prodrugs/chemistry , Proline/analogs & derivatives , Pyrimidines/chemistry , Spiro Compounds/chemistry , Tryptophan Hydroxylase/antagonists & inhibitors , Tryptophan Hydroxylase/metabolism , Animals , Binding Sites , Dogs , Half-Life , Humans , Inhibitory Concentration 50 , Intestinal Mucosa/metabolism , Intestines/drug effects , Mice , Molecular Docking Simulation , Prodrugs/metabolism , Prodrugs/pharmacology , Proline/metabolism , Proline/pharmacology , Protein Structure, Tertiary , Pyrimidines/metabolism , Pyrimidines/pharmacology , Rats , Serotonin/metabolism , Spiro Compounds/metabolism , Spiro Compounds/pharmacology , Structure-Activity Relationship
7.
J Pharmacol Exp Ther ; 360(1): 1-13, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27754931

ABSTRACT

Autotaxin (ATX) is a secreted glycoprotein that converts lysophosphatidylcholine (LPC) to the bioactive phospholipid lysophosphatidic acid (LPA) and is the major enzyme generating circulating LPA. Inhibition of LPA signaling has profound antifibrotic effects in multiple organ systems, including lung, kidney, skin, and peritoneum. However, other LPA-generating pathways exist, and the role of ATX in localized tissue LPA production and fibrosis remains unclear and controversial. In this study, we describe the preclinical pharmacologic, pharmacokinetic, and pharmacodynamic properties of a novel small-molecule ATX inhibitor, PAT-505 [3-((6-chloro-2-cyclopropyl-1-(1-ethyl-1H-pyrazol-4-yl)-7-fluoro-1H-indol-3-yl) thio)-2-fluorobenzoic acid sodium salt]. PAT-505 is a potent, selective, noncompetitive inhibitor that displays significant inhibition of ATX activity in plasma and liver tissue after oral administration. When dosed therapeutically in a Stelic Mouse Animal Model of nonalcoholic steatohepatitis (NASH), PAT-505 treatment resulted in a small but significant improvement in fibrosis with only minor improvements in hepatocellular ballooning and hepatic inflammation. In a choline-deficient, high-fat diet model of NASH, therapeutic treatment with PAT-505 robustly reduced liver fibrosis with no significant effect on steatosis, hepatocellular ballooning, or inflammation. These data demonstrate that inhibiting autotaxin is antifibrotic and may represent a novel therapeutic approach for the treatment of multiple fibrotic liver diseases, including NASH.


Subject(s)
Enzyme Inhibitors/pharmacology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/enzymology , Phosphoric Diester Hydrolases/metabolism , Piperazines/pharmacology , Animals , Disease Models, Animal , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Female , Humans , Male , Mice , Piperazines/pharmacokinetics , Piperazines/therapeutic use
8.
Bioorg Med Chem Lett ; 26(12): 2855-2860, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27146606

ABSTRACT

An increasing number of diseases have been linked to a dysfunctional peripheral serotonin system. Given that tryptophan hydroxylase 1 (TPH1) is the rate limiting enzyme in the biosynthesis off serotonin, it represents an attractive target to regulate peripheral serotonin. Following up to our first disclosure, we report a new chemotype of TPH1 inhibitors where-by the more common central planar heterocycle has been replaced with an open-chain, acyl guanidine surrogate. Through our work, we found that compounds of this nature provide highly potent TPH1 inhibitors with favorable physicochemical properties that were effective in reducing murine intestinal 5-HT in vivo. Furthermore, we obtained a high resolution (1.90Å) X-ray structure crystal structure of one of these inhibitors (compound 51) that elucidated the active conformation along with revealing a dimeric form of TPH1 for the first time.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Guanidine/pharmacology , Tryptophan Hydroxylase/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Guanidine/chemical synthesis , Guanidine/chemistry , Humans , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Tryptophan Hydroxylase/metabolism
9.
Mol Pharmacol ; 88(6): 982-92, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26371182

ABSTRACT

Autotaxin (ATX) is a secreted enzyme that hydrolyzes lysophosphatidylcholine to lysophosphatidic acid (LPA). LPA is a bioactive phospholipid that regulates diverse biological processes, including cell proliferation, migration, and survival/apoptosis, through the activation of a family of G protein-coupled receptors. The ATX-LPA pathway has been implicated in many pathologic conditions, including cancer, fibrosis, inflammation, cholestatic pruritus, and pain. Therefore, ATX inhibitors represent an attractive strategy for the development of therapeutics to treat a variety of diseases. Mouse and rat ATX have been crystallized previously with LPA or small-molecule inhibitors bound. Here, we present the crystal structures of human ATX in complex with four previously unpublished, structurally distinct ATX inhibitors. We demonstrate that the mechanism of inhibition of each compound reflects its unique interactions with human ATX. Our studies may provide a basis for the rational design of novel ATX inhibitors.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/metabolism , Animals , Cell Line, Tumor , Crystallization , HEK293 Cells , Humans , Mice , Protein Binding/physiology , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
10.
Biochemistry ; 50(38): 8103-6, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21870819

ABSTRACT

Isocitrate dehydrogenase kinase/phosphatase (AceK) regulates entry into the glyoxylate bypass by reversibly phosphorylating isocitrate dehydrogenase (ICDH). On the basis of the recently determined structure of the AceK-ICDH complex from Escherichia coli, we have classified the structures of homodimeric NADP(+)-ICDHs to rationalize and predict which organisms likely contain substrates for AceK. One example is Burkholderia pseudomallei (Bp). Here we report a crystal structure of Bp-ICDH that exhibits the necessary structural elements required for AceK recognition. Kinetic analyses provided further confirmation that Bp-ICDH is a substrate for AceK. We conclude that the highly stringent AceK binding sites on ICDH are maintained only in Gram-negative bacteria.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Burkholderia pseudomallei/enzymology , Catalytic Domain , Dimerization , Escherichia coli/enzymology , Gram-Negative Bacteria/enzymology , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/classification , Isocitrate Dehydrogenase/metabolism , Kinetics , Models, Molecular , Protein Conformation , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Substrate Specificity
11.
Nat Struct Mol Biol ; 13(11): 1002-9, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17041599

ABSTRACT

The Ro autoantigen is ring-shaped, binds misfolded noncoding RNAs and is proposed to function in quality control. Here we determine how Ro interacts with misfolded RNAs. Binding of Ro to misfolded precursor (pre)-5S ribosomal RNA requires a single-stranded 3' end and helical elements. As mutating most sequences of the helices and tail results in modest decreases in binding, Ro may be able to associate with a range of RNAs. Ro binds several other RNAs that contain single-stranded tails. A crystal structure of Ro bound to a misfolded pre-5S rRNA fragment reveals that the tail inserts into the cavity, while a helix binds on the surface. Most contacts of Ro with the helix are to the backbone. Mutagenesis reveals that Ro has an extensive RNA-binding surface. We propose that Ro uses this surface to scavenge RNAs that fail to bind their specific RNA-binding proteins.


Subject(s)
Nucleic Acid Conformation , RNA Precursors/chemistry , RNA, Ribosomal, 5S/chemistry , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Animals , Autoantigens , Base Sequence , Binding Sites , Crystallography, X-Ray , Models, Molecular , Molecular Sequence Data , Mutagenesis , Nuclease Protection Assays/methods , Oocytes/chemistry , Oocytes/metabolism , Protein Binding , Protein Structure, Tertiary , RNA 3' End Processing , RNA Precursors/genetics , RNA Precursors/metabolism , RNA, Ribosomal, 5S/genetics , RNA, Ribosomal, 5S/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus laevis/metabolism
12.
Cell ; 121(4): 529-539, 2005 May 20.
Article in English | MEDLINE | ID: mdl-15907467

ABSTRACT

The Ro 60 kDa autoantigen is a major target of the immune response in patients with systemic lupus erythematosus. In vertebrate cells, Ro binds misfolded small RNAs and likely functions in RNA quality control. In eukaryotes and bacteria, Ro also associates with small RNAs called Y RNAs. We present structures of unliganded Ro and Ro complexed with two RNAs at 1.95 and 2.2 A resolution, respectively. Ro consists of a von Willebrand factor A domain and a doughnut-shaped domain composed of HEAT repeats. In the complex, a fragment of Y RNA binds on the outer surface of the HEAT-repeat ring, and single-stranded RNA binds in the toroid hole. Mutagenesis supports a binding site for misfolded RNAs that encompasses both sites, with a single-stranded end inserted into the toroid cavity. Our experiments suggest that one role of Y RNAs may be to regulate access of other RNAs to Ro.


Subject(s)
RNA Processing, Post-Transcriptional/physiology , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , RNA/metabolism , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , Amino Acid Motifs/physiology , Amino Acid Sequence/physiology , Animals , Binding Sites/physiology , Cells, Cultured , Crystallography, X-Ray , Insecta , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding/physiology , Protein Structure, Tertiary/physiology , RNA/chemistry , RNA, Small Cytoplasmic/chemistry , RNA, Small Cytoplasmic/metabolism , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/genetics , Ribonucleoproteins/genetics , Xenopus laevis
13.
J Biol Chem ; 277(11): 9484-91, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11779862

ABSTRACT

1-l-myo-Inositol-1-phosphate synthase catalyzes the conversion of d-glucose 6-phosphate to 1-l-myo-inositol-1-phosphate (MIP), the first and rate-limiting step in the biosynthesis of all inositol-containing compounds. It involves an oxidation, intramolecular aldol cyclization, and reduction. We have determined the first crystal structure of MIP synthase. We present structures of both the NAD-bound enzyme and the enzyme bound to an inhibitor, 2-deoxy-glucitol-6-phosphate. While 58 amino acids are disordered in the unbound form of the enzyme in the vicinity of the active site, the inhibitor nucleates the folding of this domain in a striking example of induced fit, serving to completely encapsulate it within the enzyme. Three helices and a long beta-strand are formed in this process. We postulate a mechanism for the conversion based on the structure of the inhibitor-bound complex.


Subject(s)
Myo-Inositol-1-Phosphate Synthase/chemistry , Sorbitol/analogs & derivatives , Crystallization , Crystallography , Myo-Inositol-1-Phosphate Synthase/antagonists & inhibitors , Myo-Inositol-1-Phosphate Synthase/metabolism , NAD/metabolism , Protein Conformation , Sorbitol/metabolism , Sugar Phosphates/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...