Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Cell Death Dis ; 10(10): 749, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31582741

ABSTRACT

Lysosomal sequestration of anti-cancer compounds reduces drug availability at intracellular target sites, thereby limiting drug-sensitivity and inducing chemoresistance. For hepatocellular carcinoma (HCC), sorafenib (SF) is the first line systemic treatment, as well as a simultaneous activator of autophagy-induced drug resistance. The purpose of this study is to elucidate how combination therapy with the FDA-approved photosensitizer verteporfin (VP) can potentiate the antitumor effect of SF, overcoming its acquired resistance mechanisms. HCC cell lines and patient-derived in vitro and in vivo preclinical models were used to identify the molecular mechanism of action of VP alone and in combination with SF. We demonstrate that SF is lysosomotropic and increases the total number of lysosomes in HCC cells and patient-derived xenograft model. Contrary to the effect on lysosomal stability by SF, VP is not only sequestered in lysosomes, but induces lysosomal pH alkalinization, lysosomal membrane permeabilization (LMP) and tumor-selective proteotoxicity. In combination, VP-induced LMP potentiates the antitumor effect of SF, further decreasing tumor proliferation and progression in HCC cell lines and patient-derived samples in vitro and in vivo. Our data suggest that combination of lysosome-targeting compounds, such as VP, in combination with already approved chemotherapeutic agents could open a new avenue to overcome chemo-insensitivity caused by passive lysosomal sequestration of anti-cancer drugs in the context of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Lysosomes/metabolism , Sorafenib/pharmacology , Verteporfin/pharmacology , Alkalies/chemistry , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hydrogen-Ion Concentration , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Lysosomes/drug effects , Male , Mice , Models, Biological , Neoplasm Proteins/toxicity , Permeability , ras Proteins/metabolism
2.
Chembiochem ; 20(5): 644-649, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30462880

ABSTRACT

Selective and specific inhibitors of Plasmodium falciparum lysyl-tRNA synthetase represent promising therapeutic antimalarial avenues. Cladosporin was identified as a potent P. falciparum lysyl-tRNA synthetase inhibitor, with an activity against parasite lysyl-tRNA synthetase >100-fold more potent than that of the activity registered against the human enzyme. Despite its compelling activity, cladosporin exhibits poor oral bioavailability; a critical requirement for antimalarial drugs. Thus, the quest to develop metabolically stable cladosporin-derived analogues, while retaining similar selectivity and potency to that of the natural compound, has begun. Chemogenomic profiling of a designed library allowed an entirely innovative structure-activity relationship study to be initiated; this shed light on structural evidence of a privileged scaffold with a unique activity against tRNA synthetases.


Subject(s)
Antimalarials/chemical synthesis , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Isocoumarins/chemical synthesis , Lysine-tRNA Ligase/antagonists & inhibitors , Malaria, Falciparum/drug therapy , Humans , Plasmodium falciparum/enzymology , Structure-Activity Relationship
3.
ACS Chem Biol ; 13(12): 3354-3360, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30451487

ABSTRACT

Hypoxia, a condition of reduced oxygen, occurs in a wide variety of biological contexts, including solid tumors and bacterial biofilms, which are relevant to human health. Consequently, the development of chemical tools to study hypoxia is vital. Here we report a hypoxia-activated, small-molecule-mediated gene expression system using a bioreductive prodrug of the inducer isopropyl 1-thio-ß-d-galactopyranoside. As a proof-of-concept we have placed the production of a green fluorescent protein under the control of hypoxia. Our system has the potential to be extended to regulate the production of any given protein of choice.


Subject(s)
Gene Expression/drug effects , Green Fluorescent Proteins/metabolism , Isopropyl Thiogalactoside/analogs & derivatives , Isopropyl Thiogalactoside/pharmacology , Prodrugs/pharmacology , Anaerobiosis/physiology , Cell Line, Tumor , Escherichia coli/genetics , Escherichia coli/metabolism , Green Fluorescent Proteins/genetics , Humans , Isopropyl Thiogalactoside/chemical synthesis , Isopropyl Thiogalactoside/metabolism , Nitrofurans/chemical synthesis , Nitrofurans/metabolism , Oxazines/chemical synthesis , Oxazines/metabolism , Prodrugs/chemical synthesis , Prodrugs/metabolism
4.
SLAS Discov ; 22(5): 571-582, 2017 06.
Article in English | MEDLINE | ID: mdl-28345372

ABSTRACT

Oral and intestinal mucositis is a debilitating side effect of radiation treatment. A mouse model of radiation-induced mucositis leads to weight loss and tissue damage, reflecting the human ailment as it responds to keratinocyte growth factor (KGF), the standard-of-care treatment. Cultured intestinal crypt organoids allowed the development of an assay monitoring the effect of treatments of intestinal epithelium to radiation-induced damage. This in vitro assay resembles the mouse model as KGF and roof plate-specific spondin-1 (RSPO1) enhanced crypt organoid recovery following radiation. Screening identified compounds that increased the survival of organoids postradiation. Testing of these compounds revealed that the organoids changed their responses over time. Unbiased transcriptome analysis was performed on crypt organoid cultures at various time points in culture to investigate this adaptive behavior. A number of genes and pathways were found to be modulated over time, providing a rationale for the altered sensitivity of the organoid cultures. This report describes an in vitro assay that reflects aspects of human disease. The assay was used to identify bioactive compounds, which served as probes to interrogate the biology of crypt organoids over prolonged culture. The pathways that are changing over time may offer potential targets for treatment of mucositis.


Subject(s)
Drug Screening Assays, Antitumor/methods , Intestines/drug effects , Organoids/drug effects , Animals , Cell Culture Techniques/methods , Fibroblast Growth Factor 7/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Organoids/metabolism , Thrombospondins/metabolism , Transcriptome/physiology
5.
Stem Cells ; 34(2): 299-310, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26577380

ABSTRACT

The multilineage differentiation capacity of mouse and human embryonic stem (ES) cells offers a testing platform for small molecules that mediate mammalian lineage determination and cellular specialization. Here we report the identification of two small molecules which drives mouse 129 ES cell differentiation to skeletal muscle with high efficiency without any genetic modification. Mouse embryoid bodies (EBs) were used to screen a library of 1,000 small molecules to identify compounds capable of inducing high levels of Pax3 mRNA. Stimulation of EBs with SMIs (skeletal muscle inducer, SMI1 and SMI2) from the screen resulted in a high percentage of intensively twitching skeletal muscle fibers 3 weeks after induction. Gene expression profiling studies that were carried out for mode of actions analysis showed that SMIs activated genes regulated by the Wnt pathway and inhibited expression of Smad2/3 and Sonic Hedgehog (Shh) target genes. A combination of three small molecules known to modulate these three pathways acted similarly to the SMIs found here, driving ES cells from 129 as well as Balb/c and C57Bl/6 to skeletal muscle. Taken together, these data demonstrate that the SMI drives ES cells to skeletal muscle via concerted activation of the Wnt pathway, and inhibition of Smad2/3 signaling and Shh pathways. This provides important developmental biological information about skeletal muscle differentiation from embryonic stem cells and may lead to the development of new therapeutics for muscle disease.


Subject(s)
Cell Differentiation , Hedgehog Proteins/metabolism , Mouse Embryonic Stem Cells/metabolism , Muscle Fibers, Fast-Twitch/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Wnt Signaling Pathway , Animals , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Mice , Mouse Embryonic Stem Cells/cytology , Muscle Fibers, Fast-Twitch/cytology
6.
Int J Radiat Biol ; 90(8): 687-99, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24650104

ABSTRACT

PURPOSE: Radioresistance of cancer cells remains a fundamental barrier for maximum efficient radiotherapy. Tumor heterogeneity and the existence of distinct cell subpopulations exhibiting different genotypes and biological behaviors raise difficulties to eradicate all tumorigenic cells. Recent evidence indicates that a distinct population of tumor cells, called cancer stem cells (CSC), is involved in tumor initiation and recurrence and is a putative cause of tumor radioresistance. There is an urgent need to identify the intrinsic molecular mechanisms regulating the generation and maintenance of resistance to radiotherapy, especially within the CSC subset. The chemokine C-X-C motif receptor 4 (CXCR4) has been found to be a prognostic marker in various types of cancer, being involved in chemotaxis, stemness and drug resistance. The interaction of CXCR4 with its ligand, the chemokine C-X-C motif ligand 12 (CXCL12), plays an important role in modulating the tumor microenvironment, angiogenesis and CSC niche. Moreover, the therapeutic inhibition of the CXCR4/CXCL12 signaling pathway is sensitizing the malignant cells to conventional anti-cancer therapy. CONTENT: Within this review we are summarizing the role of the CXCR4/CXCL12 axis in the modulation of CSC properties, the regulation of the tumor microenvironment in response to irradiation, therapy resistance and tumor relapse. CONCLUSION: In light of recent findings, the inhibition of the CXCR4/CXCL12 signaling pathway is a promising therapeutic option to refine radiotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Radiation Tolerance , Receptors, CXCR4/metabolism , Animals , Humans , Neoplastic Stem Cells/drug effects , Radiation Tolerance/drug effects , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/radiation effects
7.
PLoS One ; 7(7): e40976, 2012.
Article in English | MEDLINE | ID: mdl-22815884

ABSTRACT

The Wnt/ß-catenin signaling pathbway controls many important biological processes. R-Spondin (RSPO) proteins are a family of secreted molecules that strongly potentiate Wnt/ß-catenin signaling, however, the molecular mechanism of RSPO action is not yet fully understood. We performed an unbiased siRNA screen to identify molecules specifically required for RSPO, but not Wnt, induced ß-catenin signaling. From this screen, we identified LGR4, then an orphan G protein-coupled receptor (GPCR), as the cognate receptor of RSPO. Depletion of LGR4 completely abolished RSPO-induced ß-catenin signaling. The loss of LGR4 could be compensated by overexpression of LGR5, suggesting that LGR4 and LGR5 are functional homologs. We further demonstrated that RSPO binds to the extracellular domain of LGR4 and LGR5, and that overexpression of LGR4 strongly sensitizes cells to RSPO-activated ß-catenin signaling. Supporting the physiological significance of RSPO-LGR4 interaction, Lgr4-/- crypt cultures failed to grow in RSPO-containing intestinal crypt culture medium. No coupling between LGR4 and heterotrimeric G proteins could be detected in RSPO-treated cells, suggesting that LGR4 mediates RSPO signaling through a novel mechanism. Identification of LGR4 and its relative LGR5, an adult stem cell marker, as the receptors of RSPO will facilitate the further characterization of these receptor/ligand pairs in regenerative medicine applications.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Thrombospondins/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , DNA, Complementary/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Intestinal Mucosa/metabolism , Ligands , Models, Biological , Open Reading Frames , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , Stem Cells/cytology
8.
Science ; 336(6082): 717-21, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22491093

ABSTRACT

Osteoarthritis (OA) is a degenerative joint disease that involves the destruction of articular cartilage and eventually leads to disability. Molecules that promote the selective differentiation of multipotent mesenchymal stem cells (MSCs) into chondrocytes may stimulate the repair of damaged cartilage. Using an image-based high-throughput screen, we identified the small molecule kartogenin, which promotes chondrocyte differentiation (median effective concentration = 100 nM), shows chondroprotective effects in vitro, and is efficacious in two OA animal models. Kartogenin binds filamin A, disrupts its interaction with the transcription factor core-binding factor ß subunit (CBFß), and induces chondrogenesis by regulating the CBFß-RUNX1 transcriptional program. This work provides new insights into the control of chondrogenesis that may ultimately lead to a stem cell-based therapy for osteoarthritis.


Subject(s)
Anilides/pharmacology , Cartilage, Articular/cytology , Chondrocytes/drug effects , Chondrogenesis , Mesenchymal Stem Cells/drug effects , Osteoarthritis/drug therapy , Phthalic Acids/pharmacology , Anilides/administration & dosage , Anilides/chemistry , Anilides/therapeutic use , Animals , Cattle , Cell Nucleus/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrocytes/physiology , Contractile Proteins/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/metabolism , Disease Models, Animal , Filamins , High-Throughput Screening Assays , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Microfilament Proteins/metabolism , Osteoarthritis/pathology , Osteoarthritis/physiopathology , Phthalic Acids/administration & dosage , Phthalic Acids/chemistry , Phthalic Acids/therapeutic use , Regeneration , Small Molecule Libraries , Structure-Activity Relationship
9.
PLoS One ; 7(2): e31226, 2012.
Article in English | MEDLINE | ID: mdl-22359577

ABSTRACT

Tumor progenitor cells represent a population of drug-resistant cells that can survive conventional chemotherapy and lead to tumor relapse. However, little is known of the role of tumor progenitors in prostate cancer metastasis. The studies reported herein show that the CXCR4/CXCL12 axis, a key regulator of tumor dissemination, plays a role in the maintenance of prostate cancer stem-like cells. The CXCL4/CXCR12 pathway is activated in the CD44(+)/CD133(+) prostate progenitor population and affects differentiation potential, cell adhesion, clonal growth and tumorigenicity. Furthermore, prostate tumor xenograft studies in mice showed that a combination of the CXCR4 receptor antagonist AMD3100, which targets prostate cancer stem-like cells, and the conventional chemotherapeutic drug Taxotere, which targets the bulk tumor, is significantly more effective in eradicating tumors as compared to monotherapy.


Subject(s)
Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/metabolism , Receptors, CXCR4/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols , Benzylamines , Cell Adhesion , Cell Proliferation , Chemokine CXCL12/metabolism , Cyclams , Docetaxel , Heterocyclic Compounds/pharmacology , Humans , Male , Mice , Neoplasm Metastasis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Taxoids/pharmacology
12.
Science ; 329(5997): 1345-8, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20688981

ABSTRACT

Although practiced clinically for more than 40 years, the use of hematopoietic stem cell (HSC) transplants remains limited by the ability to expand these cells ex vivo. An unbiased screen with primary human HSCs identified a purine derivative, StemRegenin 1 (SR1), that promotes the ex vivo expansion of CD34+ cells. Culture of HSCs with SR1 led to a 50-fold increase in cells expressing CD34 and a 17-fold increase in cells that retain the ability to engraft immunodeficient mice. Mechanistic studies show that SR1 acts by antagonizing the aryl hydrocarbon receptor (AHR). The identification of SR1 and AHR modulation as a means to induce ex vivo HSC expansion should facilitate the clinical use of HSC therapy.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Purines/metabolism , Purines/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , AC133 Antigen , Animals , Antigens, CD/analysis , Antigens, CD34/analysis , Aryl Hydrocarbon Hydroxylases/genetics , Aryl Hydrocarbon Hydroxylases/metabolism , Cell Count , Cell Lineage , Cell Proliferation , Cells, Cultured , Cytochrome P-450 CYP1B1 , Cytokines/pharmacology , Glycoproteins/analysis , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/physiology , Peptides/analysis , Polychlorinated Dibenzodioxins/pharmacology , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Small Molecule Libraries , Species Specificity
13.
Proc Natl Acad Sci U S A ; 106(22): 8912-7, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19447925

ABSTRACT

Ectopic expression of defined transcription factors can reprogram somatic cells to induced pluripotent stem (iPS) cells, but the utility of iPS cells is hampered by the use of viral delivery systems. Small molecules offer an alternative to replace virally transduced transcription factors with chemical signaling cues responsible for reprogramming. In this report we describe a small-molecule screening platform applied to identify compounds that functionally replace the reprogramming factor Klf4. A series of small-molecule scaffolds were identified that activate Nanog expression in mouse fibroblasts transduced with a subset of reprogramming factors lacking Klf4. Application of one such molecule, kenpaullone, in lieu of Klf4 gave rise to iPS cells that are indistinguishable from murine embryonic stem cells. This experimental platform can be used to screen large chemical libraries in search of novel compounds to replace the reprogramming factors that induce pluripotency. Ultimately, such compounds may provide mechanistic insight into the reprogramming process.


Subject(s)
Benzazepines/pharmacology , Cell Differentiation , Epigenesis, Genetic/drug effects , Fibroblasts/drug effects , Indoles/pharmacology , Pluripotent Stem Cells/cytology , Small Molecule Libraries/pharmacology , Animals , Fibroblasts/cytology , Genes, Reporter , Homeodomain Proteins/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Luciferases/genetics , Mice , Nanog Homeobox Protein
14.
Chemistry ; 15(16): 3983-4010, 2009.
Article in English | MEDLINE | ID: mdl-19241433

ABSTRACT

Nature is a pretty unselective "chemist" when it comes to making the highly cytotoxic amphidinolide macrolides of the B/G/H series. To date, 16 different such compounds have been isolated, all of which could now be approached by a highly convergent and largely catalysis-based route (see figure). This notion is exemplified by the total synthesis of five prototype members of this family.Dinoflagellates of the genus Amphidinium produce a "library" of closely related secondary metabolites of mixed polyketide origin, which are extremely scarce but highly promising owing to the exceptional cytotoxicity against various cancer cell lines. Because of the dense array of sensitive functionalities on their largely conserved macrocyclic frame, however, these amphidinolides of the B, D, G and H types elapsed many previous attempts at their synthesis. Described herein is a robust, convergent and hence general blueprint which allowed not only to conquest five prototype members of these series, but also holds the promise of making "non-natural" analogues available by diverted total synthesis. This notion transpires for a synthesis-driven structure revision of amphidinolide H2. The successful route hinges upon a highly productive Stille-Migita cross-coupling reaction at the congested and chemically labile 1,3-diene site present in all such targets, which required the development of a modified chloride- and fluoride-free protocol. The macrocyclic ring could be formed with high efficiency and selectivity by ring-closing metathesis (RCM) engaging a vinyl epoxide unit as one of the reaction partners. Because of the sensitivity of the targets to oxidizing and reducing conditions as well as to pH changes, the proper adjustment of the protecting group pattern for the peripheral -OH functions also constitutes a critical aspect, which has to converge to silyl groups only once the diene is in place. Tris(dimethylamino)sulfonium difluorotrimethylsilicate (TASF) turned out to be a sufficiently mild fluoride source to allow for the final deprotection without damaging the precious macrolides.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Macrolides/chemistry , Macrolides/chemical synthesis , Marine Toxins/chemistry , Marine Toxins/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Catalysis , Combinatorial Chemistry Techniques , Dinoflagellida/chemistry , Macrolides/pharmacology , Marine Toxins/pharmacology , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Structure-Activity Relationship
15.
Chem Commun (Camb) ; (25): 2873-5, 2008 Jul 07.
Article in English | MEDLINE | ID: mdl-18566709

ABSTRACT

The combination of catalytic amounts of [Pd(PPh3)4], copper thiophene-2-carboxylate (CuTC) and [Ph2PO2][NBu4] allowed a series of exigent Stille-Migita reactions to be performed with high yields; as the protocol is fluoride free, a variety of O-silyl and C-silyl groups remained intact.


Subject(s)
Cross-Linking Reagents/chemistry , Catalysis , Copper/chemistry , Molecular Structure , Palladium/chemistry
17.
Chemistry ; 11(16): 4609-20, 2005 Aug 05.
Article in English | MEDLINE | ID: mdl-15954151

ABSTRACT

A new sulfur dioxide-based organic chemistry has been developed as a novel approach for the stereoselective synthesis of polyene fragments based on our one-pot, four-component synthesis of polyfunctional epsilon-alkanesulfonyl-gamma,delta-unsaturated ketones. The flexibility and efficiency of this methodology are illustrated by the preparation of (+)-methyl (2E,4E,6E,8R,9S)-9-{[(tert-butyl)dimethylsilyl]oxy}-2,4,6,8-tetramethyl-11-(triethylsilyl)undeca-2,4,6-trien-10-ynoate, a synthetic intermediate of Nicolaou and co-workers, that corresponds to the C(1)-C(11) fragment of apoptolidin, which was used by the authors in their total synthesis of this promising anticancer agent.


Subject(s)
Antineoplastic Agents/chemical synthesis , Macrolides/chemical synthesis , Sulfur Dioxide/chemistry , Actinobacteria/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis , Macrolides/chemistry , Molecular Conformation , Molecular Structure , Neoplasms/therapy , Rats
18.
Org Lett ; 7(5): 897-900, 2005 Mar 03.
Article in English | MEDLINE | ID: mdl-15727469

ABSTRACT

Efficient methods for the stereoselective synthesis of polyfunctional (E)- and (Z)-alkenes and conjugated (E,E)- and (E,Z)-dienones are presented. They rely upon one-pot, four-component processes that condense 1-oxy-1,3-dienes, silyl enol ethers, SO2, and carbon electrophiles. [structure: see text]

19.
J Org Chem ; 69(19): 6413-8, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15357602

ABSTRACT

The ene reaction of sulfur dioxide with enoxysilanes or with allylsilanes generates silyl sulfinates that can be brominated (Br(2) or NBS) or chlorinated (NCS or Cl(2)) to produce the corresponding sulfonyl halides. They react with primary and secondary amines or alcohols to give the corresponding sulfonamides and sulfonic esters, respectively. The hetero-Diels-Alder addition of sulfur dioxide to 1-oxy- or 1,3-dioxy-1,3-dienes generates zwitterions that add to enoxysilanes or allylsilanes giving silyl sulfinates that can be converted in situ into polyfunctional sulfonamides or sulfonic esters. This realizes quick access to libraries of complicated sulfonamides and sulfonic esters applying one-pot, three- and four-component methods.

SELECTION OF CITATIONS
SEARCH DETAIL