Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters











Publication year range
1.
SLAS Discov ; 29(2): 100146, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38311110

ABSTRACT

Here we offer perspectives on phenotypic screening based on a wide-ranging discussion entitled "Phenotypic screening, target ID, and multi-omics: enabling more disease relevance in early discovery?" at the Screen Design and Assay Technology Special Interest Group Meeting at the 2023 SLAS Conference. During the session, the authors shared their own experience from within their respective organizations, followed by an open discussion with the audience. It was recognized that while substantial progress has been made towards translating disease-relevant phenotypic early discovery into clinical success, there remain significant operational and scientific challenges to implementing phenotypic screening efforts, and improving translation of screening hits comes with substantial resource demands and organizational commitment. This Perspective assesses progress, highlights pitfalls, and offers possible solutions to help unlock the therapeutic potential of phenotypic drug discovery. Areas explored comprise screening and hit validation strategy, choice of cellular model, moving beyond 2D cell culture into three dimensions, and leveraging high-dimensional data sets downstream of phenotypic screens.


Subject(s)
Drug Discovery , Public Opinion , Drug Discovery/methods , Phenotype
2.
SLAS Discov ; 24(7): 714-723, 2019 08.
Article in English | MEDLINE | ID: mdl-31039326

ABSTRACT

Cell-based high-throughput drug screening (HTS) is a common starting point for the drug discovery and development process. Currently, there is a push to combine complex cell culture systems with HTS to provide more clinically applicable results. However, there are mechanistic requirements inherent to HTS as well as material limitations that make this integration challenging. Here, we used the peptide-based shear-thinning hydrogel MAX8 tagged with the RGDS sequence to create a synthetic extracellular scaffold to culture cells in three dimensions and showed a preliminary implementation of the scaffold within an automated HTS setup using a pilot drug screen targeting medulloblastoma, a pediatric brain cancer. A total of 2202 compounds were screened in the 384-well format against cells encapsulated in the hydrogel as well as cells growing on traditional two-dimensional (2D) plastic. Eighty-two compounds passed the first round of screening at a single point of concentration. Sixteen-point dose-response was done on those 82 compounds, of which 17 compounds were validated. Three-dimensional (3D) cell-based HTS could be a powerful screening tool that allows researchers to finely tune the cell microenvironment, getting more clinically applicable data as a result. Here, we have shown the successful integration of a peptide-based hydrogel into the high-throughput format.


Subject(s)
Cell Culture Techniques , Drug Discovery/methods , High-Throughput Screening Assays/methods , Hydrogels , Peptides , Amino Acid Sequence , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Hydrogels/chemistry , Peptides/chemistry , Reproducibility of Results , Small Molecule Libraries
3.
Clin Transl Sci ; 11(5): 461-470, 2018 09.
Article in English | MEDLINE | ID: mdl-29877628

ABSTRACT

The Assay Guidance Manual (AGM) is an eBook of best practices for the design, development, and implementation of robust assays for early drug discovery. Initiated by pharmaceutical company scientists, the manual provides guidance for designing a "testing funnel" of assays to identify genuine hits using high-throughput screening (HTS) and advancing them through preclinical development. Combined with a workshop/tutorial component, the overall goal of the AGM is to provide a valuable resource for training translational scientists.


Subject(s)
Biological Assay/methods , Drug Discovery , Geography , High-Throughput Screening Assays , Humans , Translational Research, Biomedical
4.
Anal Biochem ; 535: 25-34, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28757092

ABSTRACT

Automated cell-based high-throughput screening (HTS) is a powerful tool in drug discovery, and it is increasingly being recognized that three-dimensional (3D) models, which more closely mimic in vivo-like conditions, are desirable screening platforms. One limitation hampering the development of 3D HTS is the lack of suitable 3D culture scaffolds that can readily be incorporated into existing HTS infrastructure. We now show that ß-hairpin peptide hydrogels can serve as a 3D cell culture platform that is compatible with HTS. MAX8 ß-hairpin peptides can physically assemble into a hydrogel with defined porosity, permeability and mechanical stability with encapsulated cells. Most importantly, the hydrogels can then be injected under shear-flow and immediately reheal into a hydrogel with the same properties exhibited prior to injection. The post-injection hydrogels are cell culture compatible at physiological conditions. Using standard HTS equipment and medulloblastoma pediatric brain tumor cells as a model system, we show that automatic distribution of cell-peptide mixtures into 384-well assay plates results in evenly dispensed, viable MAX8-cell constructs suitable for commercially available cell viability assays. Since MAX8 peptides can be functionalized to mimic the microenvironment of cells from a variety of origins, MAX8 peptide gels should have broad applicability for 3D HTS drug discovery.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Culture Techniques/methods , Drug Discovery , High-Throughput Screening Assays , Hydrogels/chemical synthesis , Peptides/chemistry , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Hydrogels/chemistry , Peptides/chemical synthesis , Rheology , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Front Oncol ; 7: 42, 2017.
Article in English | MEDLINE | ID: mdl-28421158

ABSTRACT

Costello syndrome (CS) patients suffer from a very high 10% incidence of embryonal rhabdomyosarcoma (ERMS). As tools to discover targeted therapeutic leads, we used a CS patient-derived ERMS cell line (CS242 ERMS) harboring a homozygous p.G12A mutation in HRAS, and a control cell line derived from the same patient comprising non-malignant CS242 fibroblasts with a heterozygous p.G12A HRAS mutation. A library of 2,000 compounds with known pharmacological activities was screened for their effect on CS242 ERMS cell viability. Follow-up testing in a panel of cell lines revealed that various compounds originally developed for other indications were remarkably selective; notably, the phosphodiesterase (PDE) inhibitor zardaverine was at least 1,000-fold more potent in CS242 ERMS than in the patient-matched non-malignant CS242 fibroblasts, other ERMS, or normal fibroblasts. Chronic treatment with zardaverine led to the emergence of resistant cells, consistent with CS242 ERMS comprising a mixed population of cells. Many PDE inhibitors in addition to zardaverine were tested on CS242 ERMS, but almost all had no effect. Interestingly, zardaverine and analogs showed a similar cytotoxicity profile in CS242 ERMS and cervical carcinoma-derived HeLa cells, suggesting a mechanism of action common to both cell types that does not require the presence of an HRAS mutation (HeLa contains wild type HRAS). Two recent studies presented possible mechanistic explanations for the cytotoxicity of zardaverine in HeLa cells. One revealed that zardaverine inhibited a HeLa cell-based screen measuring glucocorticoid receptor (GR) activation; however, using engineered HeLa cells, we ruled out a specific effect of zardaverine on signaling through the GR. The second attributed zardaverine toxicity in HeLa cells to promotion of the interaction of phosphodiesterase 3A and the growth regulatory protein Schlafen 12. We speculate that this work may provide a possible mechanism for zardaverine action in CS242 ERMS, although we have not yet tested this hypothesis. In conclusion, we have identified zardaverine as a potent cytotoxic agent in a CS-derived ERMS cell line and in HeLa. Although we have ruled out some possibilities, the mechanism of action of zardaverine in CS242 ERMS remains to be determined.

8.
PLoS One ; 10(10): e0140564, 2015.
Article in English | MEDLINE | ID: mdl-26465153

ABSTRACT

The bone marrow microenvironment plays an important role in acute lymphoblastic leukemia (ALL) cell proliferation, maintenance, and resistance to chemotherapy. Annexin II (ANX2) is abundantly expressed on bone marrow cells and complexes with p11 to form ANX2/p11-hetero-tetramer (ANX2T). We present evidence that p11 is upregulated in refractory ALL cell lines and patient samples. A small molecule inhibitor that disrupts ANX2/p11 interaction (ANX2T inhibitor), an anti-ANX2 antibody, and knockdown of p11, abrogated ALL cell adhesion to osteoblasts, indicating that ANX2/p11 interaction facilitates binding and retention of ALL cells in the bone marrow. Furthermore, ANX2T inhibitor increased the sensitivity of primary ALL cells co-cultured with osteoblasts to dexamethasone and vincristine induced cell death. Finally, in an orthotopic leukemia xenograft mouse model, the number of ALL cells homing to the bone marrow was reduced by 40-50% in mice injected with anti-ANX2 antibody, anti-p11 antibody or ANX2T inhibitor compared to respective controls. In a long-term engraftment assay, the percentage of ALL cells in mouse blood, bone marrow and spleen was reduced in mice treated with agents that disrupt ANX2/p11 interaction. These data show that disruption of ANX2/p11 interaction results in reduced ALL cell adhesion to osteoblasts, increased ALL cell sensitization to chemotherapy, and suppression of ALL cell homing and engraftment.


Subject(s)
Annexin A2/metabolism , Cell Proliferation/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , S100 Proteins/metabolism , Tumor Microenvironment/genetics , Animals , Annexin A2/antagonists & inhibitors , Annexin A2/genetics , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Cell Line, Tumor , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Leukemic/drug effects , Humans , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Interaction Maps/drug effects , Protein Multimerization/drug effects , S100 Proteins/genetics , Small Molecule Libraries , Vincristine/administration & dosage , Xenograft Model Antitumor Assays
14.
Assay Drug Dev Technol ; 12(8): 443-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25383720

ABSTRACT

Has the impact of irreproducibility on the discovery and development of drugs, as with global warming, metaphorically speaking, crept up on us as we slept? Or is the problem more an issue of heightened awareness? We currently find ourselves in a time when the impact of irreproducibility can easily be amplified by the combinatorial effect of our increasing reliance on advanced technologies and unrealistic expectations of how scientific truths unfold. How and why we got here is a topic that has been written on extensively (1-3) and is probably as complex as any other problem, given the dependence of science today on so many external forces. Through a series of questions, we asked members of our editorial board their opinions on scientific irreproducibility. They chose to answer the same questions from different levels, indicating the depth of the problem and perhaps where they each believe change for the better needs to begin. My thanks to the participants.


Subject(s)
Drug Discovery/standards , High-Throughput Screening Assays/standards , Reproducibility of Results , Animals , Humans
15.
Assay Drug Dev Technol ; 12(5): 258-71, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24927133

ABSTRACT

A major focus of our pediatric cancer research is the discovery of chemical probes to further our understanding of the biology of leukemia harboring fusion proteins arising from chromosomal rearrangements, and to develop novel specifically targeted therapies. The NUP98-NSD1 fusion protein occurs in a highly aggressive subtype of acute myeloid leukemia after rearrangement of the genes NUP98 and NSD1. The methyltransferase activity of NSD1 is retained in the fusion, and it gives rise to abnormally high levels of methylation at lysine 36 on histone 3, enforcing oncogene activation. Therefore, inhibition of the methyltransferase activity of NUP98-NSD1 may be considered a viable therapeutic strategy. Here, we report the development and validation of a highly sensitive and robust luminescence-based assay for NSD1 and other methyltransferases that use S-adenosylmethionine (SAM) as a methyl donor. The assay quantifies S-adenosylhomocysteine (SAH), which is produced during methyl transfer from SAM. SAH is converted enzymatically to adenosine monophosphate (AMP); in the process, adenosine triphosphate (ATP) is consumed and the amount of ATP remaining is measured using a luminescent assay kit. The assay was validated by pilot high-throughput screening (HTS), dose-response confirmation of hits, and elimination of artifacts through counterscreening against SAH detection in the absence of NSD1. The known methyltransferase inhibitor suramin was identified, and profiled for selectivity against the histone methyltransferases EZH2, SETD7, and PRMT1. HTS using the luminescent NSD1 assay described here has the potential to deliver selective NSD1 inhibitors that may serve as leads in the development of targeted therapies for NUP98-NSD1-driven leukemias.


Subject(s)
Enzyme Assays/methods , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Luminescent Measurements/methods , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , S-Adenosylmethionine/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Histone Methyltransferases , Histone-Lysine N-Methyltransferase , Humans , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , Structure-Activity Relationship
16.
Front Oncol ; 3: 170, 2013.
Article in English | MEDLINE | ID: mdl-23847761

ABSTRACT

Despite dramatic advances in the treatment of pediatric leukemia over the past 50 years, there remain subsets of patients who respond poorly to treatment. Many of the high-risk cases of childhood leukemia with the poorest prognosis have been found to harbor specific genetic signatures, often resulting from chromosomal rearrangements. With increased understanding of the genetic and epigenetic makeup of high-risk pediatric leukemia has come the opportunity to develop targeted therapies that promise to be both more effective and less toxic than current chemotherapy. Of particular importance is an understanding of the interconnections between different targets within the same cancer, and observations of synergy between two different targeted therapies or between a targeted drug and conventional chemotherapy. It has become clear that many cancers are able to circumvent a single specific blockade, and pediatric leukemias are no exception in this regard. This review highlights the most promising approaches to new drugs and drug combinations for high-risk pediatric leukemia. Key biological evidence supporting selection of molecular targets is presented, together with a critical survey of recent progress toward the discovery, pre-clinical development, and clinical study of novel molecular therapeutics.

17.
Assay Drug Dev Technol ; 11(4): 253-68, 2013 May.
Article in English | MEDLINE | ID: mdl-23679849

ABSTRACT

Rearrangements of the mixed-lineage leukemia (MLL) gene occur predominately in pediatric leukemia cases and are generally predictors of a poor prognosis. These chromosomal rearrangements result in fusion of the protein MLL to one of more than 60 protein partners. MLL fusions are potent inducers of leukemia through activation of oncogene expression; therefore, targeting this transcriptional activation function may arrest MLL-rearranged (MLL-R) leukemia. Leukemic cell lines harboring the most common fusion protein, MLL-AF4, require the direct interaction of AF4 with the transcription factor AF9 to survive and self-renew; disrupting this interaction with a cell-penetrating AF4-derived peptide results in cell death, suggesting that the AF4-AF9 interaction could be a viable target for a novel MLL-R leukemia therapy. Here we describe the use of AlphaScreen technology to develop a high-throughput screening (HTS) assay to detect nonpeptidic inhibitors of AF4-AF9 binding. The assay is economical, requiring only low nanomolar concentrations of biotinylated AF4-derived peptide and FLAG-tagged AF9 in low-volume 384-well plates. A Z'-factor of 0.71 and a signal-to-background ratio of 21.3 showed the assay to be robust, and sensitivity to inhibition was demonstrated with competing AF4-derived peptides. Two pilot screens comprising 5,680 compounds served as validation for HTS at Nemours and the Broad Institute. Assay artifacts were excluded using a counterscreen comprising a biotinylated FLAG peptide. This is the first reported HTS-compatible assay to identify compounds that inhibit a key binding interaction of an MLL fusion partner, and the results presented here demonstrate suitability for screening large chemical libraries in high-density, low-volume plate formats.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Drug Discovery/methods , High-Throughput Screening Assays/methods , Myeloid-Lymphoid Leukemia Protein/antagonists & inhibitors , Nuclear Proteins/antagonists & inhibitors , Oncogene Proteins, Fusion/antagonists & inhibitors , Protein Interaction Mapping/methods , DNA-Binding Proteins/analysis , Myeloid-Lymphoid Leukemia Protein/analysis , Nuclear Proteins/analysis , Oncogene Proteins, Fusion/analysis , Protein Binding/drug effects , Transcriptional Elongation Factors
18.
Mol Cancer Res ; 11(8): 901-11, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23696131

ABSTRACT

UNLABELLED: Pancreatic ductal adenocarcinoma (PDA) is the fourth leading cause of cancer-related death in the United States, with a 95% five-year mortality rate. For over a decade, gemcitabine (GEM) has been the established first-line treatment for this disease despite suboptimal response rates. The development of PARP inhibitors that target the DNA damage repair (DDR) system in PDA cells has generated encouraging results. Ubiquitin-specific peptidase 11 (USP11), an enzyme that interacts with the DDR protein BRCA2, was recently discovered to play a key role in DNA double-strand break repair and may be a novel therapeutic target. A systematic high-throughput approach was used to biochemically screen 2,000 U.S. Food and Drug Administration (FDA)-approved compounds for inhibition of USP11 enzymatic activity. Six pharmacologically active small molecules that inhibit USP11 enzymatic activity were identified. An in vitro drug sensitivity assay demonstrated that one of these USP11 inhibitors, mitoxantrone, impacted PDA cell survival with an IC50 of less than 10 nM. Importantly, across six different PDA cell lines, two with defects in the Fanconi anemia/BRCA2 pathway (Hs766T and Capan-1), mitoxantrone is 40- to 20,000-fold more potent than GEM, with increased endogenous USP11 mRNA levels associated with increased sensitivity to mitoxantrone. Interestingly, USP11 silencing in PDA cells also enhanced sensitivity to GEM. These findings establish a preclinical model for the rapid discovery of FDA-approved compounds and identify USP11 as a target of mitoxantrone in PDA. IMPLICATIONS: This high-throughput approach provides a strong rationale to study mitoxantrone in an early-phase clinical setting for the treatment of PDA.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Mitoxantrone/therapeutic use , Pancreatic Neoplasms/drug therapy , Thiolester Hydrolases/antagonists & inhibitors , BRCA2 Protein/genetics , Benzimidazoles/therapeutic use , Carcinoma, Pancreatic Ductal/enzymology , Cell Line, Tumor , DNA Damage/genetics , Deoxycytidine/therapeutic use , Drug Screening Assays, Antitumor , Gene Silencing , High-Throughput Screening Assays , Humans , Pancreatic Neoplasms/enzymology , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Thiolester Hydrolases/metabolism , Gemcitabine
19.
PLoS One ; 6(10): e26169, 2011.
Article in English | MEDLINE | ID: mdl-22028826

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is a devastating muscle wasting disease caused by mutations in dystrophin, a muscle cytoskeletal protein. Utrophin is a homologue of dystrophin that can functionally compensate for its absence when expressed at increased levels in the myofibre, as shown by studies in dystrophin-deficient mice. Utrophin upregulation is therefore a promising therapeutic approach for DMD. The use of a small, drug-like molecule to achieve utrophin upregulation offers obvious advantages in terms of delivery and bioavailability. Furthermore, much of the time and expense involved in the development of a new drug can be eliminated by screening molecules that are already approved for clinical use. METHODOLOGY/PRINCIPAL FINDINGS: We developed and validated a cell-based, high-throughput screening assay for utrophin promoter activation, and used it to screen the Prestwick Chemical Library of marketed drugs and natural compounds. Initial screening produced 20 hit molecules, 14 of which exhibited dose-dependent activation of the utrophin promoter and were confirmed as hits. Independent validation demonstrated that one of these compounds, nabumetone, is able to upregulate endogenous utrophin mRNA and protein, in C2C12 muscle cells. CONCLUSIONS/SIGNIFICANCE: We have developed a cell-based, high-throughput screening utrophin promoter assay. Using this assay, we identified and validated a utrophin promoter-activating drug, nabumetone, for which pharmacokinetics and safety in humans are already well described, and which represents a lead compound for utrophin upregulation as a therapy for DMD.


Subject(s)
Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Utrophin/genetics , Biological Availability , Cell Line , Humans , Reproducibility of Results , Small Molecule Libraries/adverse effects , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Up-Regulation/drug effects , Up-Regulation/genetics
20.
ACS Chem Biol ; 6(6): 628-35, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21428443

ABSTRACT

RAD51 is a key protein of homologous recombination that plays a critical role in the repair of DNA double-strand breaks (DSB) and interstrand cross-links (ICL). To better understand the cellular function(s) of human RAD51, we propose to develop specific RAD51 inhibitors. RAD51 inhibitors may also help to increase the potency of anticancer drugs that act by inducing DSBs or ICLs, e.g., cisplatin or ionizing radiation. In vitro, RAD51 promotes DNA strand exchange between homologous ss- and dsDNA. Here, we developed a DNA strand exchange assay based on fluorescence resonance energy transfer and used this assay to identify RAD51 inhibitors by high-throughput screening of the NIH Small Molecule Repository (>200,000 compounds). Seventeen RAD51 inhibitors were identified and analyzed for selectivity using additional nonfluorescent DNA-based assays. As a result, we identified a compound (B02) that specifically inhibited human RAD51 (IC(50) = 27.4 µM) but not its E. coli homologue RecA (IC(50) > 250 µM). Two other compounds (A03 and A10) were identified that inhibited both RAD51 and RecA but not the structurally unrelated RAD54 protein. The structure-activity relationship (SAR) analysis allowed us to identify the structural components of B02 that are critical for RAD51 inhibition. The described approach can be used for identification of specific inhibitors of other human proteins that play an important role in DNA repair, e.g., RAD54 or Bloom's syndrome helicase.


Subject(s)
Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Quinazolinones/pharmacology , Rad51 Recombinase/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Fluorescence Resonance Energy Transfer , Humans , Molecular Structure , Quinazolinones/chemistry , Rad51 Recombinase/metabolism , Small Molecule Libraries , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL