Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Haematologica ; 106(7): 1979-1987, 2021 07 01.
Article in English | MEDLINE | ID: mdl-32586904

ABSTRACT

Pharmacological induction of fetal hemoglobin (HbF) expression is an effective therapeutic strategy for the management of beta-hemoglobinopathies such as sickle cell disease. DNA methyltransferase (DNMT) inhibitors 5-azacytidine (5-aza) and 5-aza-2'-deoxycytidine (decitabine) have been shown to induce fetal hemoglobin expression in both preclinical models and clinical studies, but are not currently approved for the management of hemoglobinopathies. We report here the discovery of a novel class of orally bioavailable DNMT1-selective inhibitors as exemplified by GSK3482364. This molecule potently inhibits the methyltransferase activity of DNMT1, but not DNMT family members DNMT3A or DNMT3B. In contrast with cytidine analog DNMT inhibitors, the DNMT1 inhibitory mechanism of GSK3482364 does not require DNA incorporation and is reversible. In cultured human erythroid progenitor cells (EPCs), GSK3482364 decreased overall DNA methylation resulting in de-repression of the gamma globin genes HBG1 and HBG2 and increased HbF expression. In a transgenic mouse model of sickle cell disease, orally administered GSK3482364 caused significant increases in both HbF levels and in the percentage HbF-expressing erythrocytes, with good overall tolerability. We conclude that in these preclinical models, selective, reversible inhibition of DNMT1 is sufficient for the induction of HbF, and is well-tolerated. We anticipate that GSK3482364 will be a useful tool molecule for the further study of selective DNMT1 inhibition both in vitro and in vivo.


Subject(s)
Anemia, Sickle Cell , Fetal Hemoglobin , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/genetics , Animals , Azacitidine/pharmacology , DNA Methylation , Fetal Hemoglobin/genetics , Mice , gamma-Globins/genetics
2.
Nat Struct Mol Biol ; 27(12): 1152-1164, 2020 12.
Article in English | MEDLINE | ID: mdl-33046907

ABSTRACT

The synthesis of poly(ADP-ribose) (PAR) reconfigures the local chromatin environment and recruits DNA-repair complexes to damaged chromatin. PAR degradation by poly(ADP-ribose) glycohydrolase (PARG) is essential for progression and completion of DNA repair. Here, we show that inhibition of PARG disrupts homology-directed repair (HDR) mechanisms that underpin alternative lengthening of telomeres (ALT). Proteomic analyses uncover a new role for poly(ADP-ribosyl)ation (PARylation) in regulating the chromatin-assembly factor HIRA in ALT cancer cells. We show that HIRA is enriched at telomeres during the G2 phase and is required for histone H3.3 deposition and telomere DNA synthesis. Depletion of HIRA elicits systemic death of ALT cancer cells that is mitigated by re-expression of ATRX, a protein that is frequently inactivated in ALT tumors. We propose that PARylation enables HIRA to fulfill its essential role in the adaptive response to ATRX deficiency that pervades ALT cancers.


Subject(s)
DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Glycoside Hydrolases/genetics , Poly(ADP-ribose) Polymerases/genetics , Protein Processing, Post-Translational , Recombinational DNA Repair , Telomere/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin/metabolism , Chromatin/ultrastructure , DNA Damage , DNA, Neoplasm/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , G2 Phase , Glycoside Hydrolases/metabolism , HeLa Cells , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/genetics , Histone Chaperones/metabolism , Histones/genetics , Histones/metabolism , Humans , Poly ADP Ribosylation , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Telomere/ultrastructure , Telomere Homeostasis , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
3.
ACS Med Chem Lett ; 11(4): 497-505, 2020 Apr 09.
Article in English | MEDLINE | ID: mdl-32292556

ABSTRACT

A combination of focused library and virtual screening, hit expansion, and rational design has resulted in the development of a series of inhibitors of RETV804M kinase, the anticipated drug-resistant mutant of RET kinase. These agents do not inhibit the wild type (wt) isoforms of RET or KDR and therefore offer a potential adjunct to RET inhibitors currently undergoing clinical evaluation.

4.
Sci Rep ; 10(1): 3836, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32123273

ABSTRACT

There is mounting evidence of androgen receptor signaling inducing genome instability and changing DNA repair capacity in prostate cancer cells. Expression of genes associated with base excision repair (BER) is increased with prostate cancer progression and correlates with poor prognosis. Poly(ADP-ribose) polymerase (PARP) and poly(ADP-ribose) glycohydrolase (PARG) are key enzymes in BER that elongate and degrade PAR polymers on target proteins. While PARP inhibitors have been tested in clinical trials and are a promising therapy for prostate cancer patients with TMPRSS2-ERG fusions and mutations in DNA repair genes, PARG inhibitors have not been evaluated. We show that PARG is a direct androgen receptor (AR) target gene. AR is recruited to the PARG locus and induces PARG expression. Androgen ablation combined with PARG inhibition synergistically reduces BER capacity in independently derived LNCaP and LAPC4 prostate cancer cell lines. A combination of PARG inhibition with androgen ablation or with the DNA damaging drug, temozolomide, significantly reduces cellular proliferation and increases DNA damage. PARG inhibition alters AR transcriptional output without changing AR protein levels. Thus, AR and PARG are engaged in reciprocal regulation suggesting that the success of androgen ablation therapy can be enhanced by PARG inhibition in prostate cancer patients.


Subject(s)
Glycoside Hydrolase Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Repair/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Glycoside Hydrolases/metabolism , Humans , Male , Molecular Targeted Therapy
5.
Cancer Res ; 79(17): 4491-4502, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31273064

ABSTRACT

Patients with metastatic pancreatic ductal adenocarcinoma (PDAC) have an average survival of less than 1 year, underscoring the importance of evaluating novel targets with matched targeted agents. We recently identified that poly (ADP) ribose glycohydrolase (PARG) is a strong candidate target due to its dependence on the pro-oncogenic mRNA stability factor HuR (ELAVL1). Here, we evaluated PARG as a target in PDAC models using both genetic silencing of PARG and established small-molecule PARG inhibitors (PARGi), PDDX-01/04. Homologous repair-deficient cells compared with homologous repair-proficient cells were more sensitive to PARGi in vitro. In vivo, silencing of PARG significantly decreased tumor growth. PARGi synergized with DNA-damaging agents (i.e., oxaliplatin and 5-fluorouracil), but not with PARPi therapy. Mechanistically, combined PARGi and oxaliplatin treatment led to persistence of detrimental PARylation, increased expression of cleaved caspase-3, and increased γH2AX foci. In summary, these data validate PARG as a relevant target in PDAC and establish current therapies that synergize with PARGi. SIGNIFICANCE: PARG is a potential target in pancreatic cancer as a single-agent anticancer therapy or in combination with current standard of care.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Glycoside Hydrolases/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , DNA Damage , Enzyme Inhibitors/pharmacology , Female , Gene Silencing , Glycoside Hydrolases/genetics , Humans , Mice, Nude , Molecular Targeted Therapy , Oxaliplatin/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Recombinational DNA Repair , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays
7.
Cancer Cell ; 35(3): 519-533.e8, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30889383

ABSTRACT

Inhibitors of poly(ADP-ribose) polymerase (PARP) have demonstrated efficacy in women with BRCA-mutant ovarian cancer. However, only 15%-20% of ovarian cancers harbor BRCA mutations, therefore additional therapies are required. Here, we show that a subset of ovarian cancer cell lines and ex vivo models derived from patient biopsies are sensitive to a poly(ADP-ribose) glycohydrolase (PARG) inhibitor. Sensitivity is due to underlying DNA replication vulnerabilities that cause persistent fork stalling and replication catastrophe. PARG inhibition is synthetic lethal with inhibition of DNA replication factors, allowing additional models to be sensitized by CHK1 inhibitors. Because PARG and PARP inhibitor sensitivity are mutually exclusive, our observations demonstrate that PARG inhibitors have therapeutic potential to complement PARP inhibitor strategies in the treatment of ovarian cancer.


Subject(s)
DNA Replication/drug effects , Enzyme Inhibitors/pharmacology , Ovarian Neoplasms/genetics , Cell Line, Tumor , Checkpoint Kinase 1 , Female , Glycoside Hydrolases/antagonists & inhibitors , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Quinazolinones/pharmacology
8.
Bioorg Med Chem Lett ; 29(4): 560-562, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30616904

ABSTRACT

Fluorination of metabolic hotspots in a molecule is a common medicinal chemistry strategy to improve in vivo half-life and exposure and, generally, this strategy offers significant benefits. Here, we report the application of this strategy to a series of poly-ADP ribose glycohydrolase (PARG) inhibitors, resulting in unexpected in vivo toxicity which was attributed to this single-atom modification.


Subject(s)
Cyclopropanes/pharmacology , Glycoside Hydrolases/toxicity , Microsomes, Liver/drug effects , Administration, Oral , Animals , Cyclopropanes/administration & dosage , Cyclopropanes/chemistry , Cyclopropanes/pharmacokinetics , Glycoside Hydrolases/administration & dosage , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/pharmacokinetics , Half-Life , Humans , Mice , Microsomes, Liver/metabolism
9.
J Med Chem ; 61(23): 10767-10792, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30403352

ABSTRACT

DNA damage repair enzymes are promising targets in the development of new therapeutic agents for a wide range of cancers and potentially other diseases. The enzyme poly(ADP-ribose) glycohydrolase (PARG) plays a pivotal role in the regulation of DNA repair mechanisms; however, the lack of potent drug-like inhibitors for use in cellular and in vivo models has limited the investigation of its potential as a novel therapeutic target. Using the crystal structure of human PARG in complex with the weakly active and cytotoxic anthraquinone 8a, novel quinazolinedione sulfonamides PARG inhibitors have been identified by means of structure-based virtual screening and library design. 1-Oxetan-3-ylmethyl derivatives 33d and 35d were selected for preliminary investigations in vivo. X-ray crystal structures help rationalize the observed structure-activity relationships of these novel inhibitors.


Subject(s)
DNA Repair , Drug Design , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Quinazolinones/chemistry , Quinazolinones/pharmacology , Administration, Oral , Animals , Biological Availability , Catalytic Domain , Glycoside Hydrolase Inhibitors/administration & dosage , Glycoside Hydrolase Inhibitors/pharmacokinetics , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/metabolism , HeLa Cells , Humans , Male , Mice , Models, Molecular , Quinazolinones/administration & dosage , Quinazolinones/pharmacokinetics , Structure-Activity Relationship
10.
Cancer Cell ; 33(6): 1078-1093.e12, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29894693

ABSTRACT

Inhibitors of poly(ADP-ribose) (PAR) polymerase (PARPi) have recently entered the clinic for the treatment of homologous recombination (HR)-deficient cancers. Despite the success of this approach, drug resistance is a clinical hurdle, and we poorly understand how cancer cells escape the deadly effects of PARPi without restoring the HR pathway. By combining genetic screens with multi-omics analysis of matched PARPi-sensitive and -resistant Brca2-mutated mouse mammary tumors, we identified loss of PAR glycohydrolase (PARG) as a major resistance mechanism. We also found the presence of PARG-negative clones in a subset of human serous ovarian and triple-negative breast cancers. PARG depletion restores PAR formation and partially rescues PARP1 signaling. Importantly, PARG inactivation exposes vulnerabilities that can be exploited therapeutically.


Subject(s)
Glycoside Hydrolases/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Synthetic Lethal Mutations , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/metabolism , Homologous Recombination/drug effects , Homologous Recombination/genetics , Humans , Mice, 129 Strain , Mice, Knockout , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly ADP Ribosylation/drug effects
11.
Bioorg Med Chem Lett ; 27(20): 4755-4759, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28927796

ABSTRACT

As part of our ongoing efforts to develop reversible inhibitors of LSD1, we identified a series of 4-(pyrrolidin-3-yl)benzonitrile derivatives that act as successful scaffold-hops of the literature inhibitor GSK-690. The most active compound, 21g, demonstrated a Kd value of 22nM and a biochemical IC50 of 57nM. In addition, this compound displayed improved selectivity over the hERG ion channel compared to GSK-690, and no activity against the related enzymes MAO-A and B. In human THP-1 acute myeloid leukaemia cells, 21g was found to increase the expression of the surrogate cellular biomarker CD86. This work further demonstrates the versatility of scaffold-hopping asa method to develop structurally diverse, potent inhibitors of LSD1.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Nitriles/chemistry , Nitriles/pharmacology , Binding Sites , Cell Line, Tumor , Drug Design , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Histone Demethylases/metabolism , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Nitriles/chemical synthesis , Protein Structure, Tertiary , Pyrrolidines/chemistry , Stereoisomerism , Structure-Activity Relationship
12.
J Med Chem ; 60(19): 7984-7999, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28892629

ABSTRACT

Inhibition of lysine specific demethylase 1 (LSD1) has been shown to induce the differentiation of leukemia stem cells in acute myeloid leukemia (AML). Irreversible inhibitors developed from the nonspecific inhibitor tranylcypromine have entered clinical trials; however, the development of effective reversible inhibitors has proved more challenging. Herein, we describe our efforts to identify reversible inhibitors of LSD1 from a high throughput screen and subsequent in silico modeling approaches. From a single hit (12) validated by biochemical and biophysical assays, we describe our efforts to develop acyclic scaffold-hops from GSK-690 (1). A further scaffold modification to a (4-cyanophenyl)glycinamide (e.g., 29a) led to the development of compound 32, with a Kd value of 32 nM and an EC50 value of 0.67 µM in a surrogate cellular biomarker assay. Moreover, this derivative does not display the same level of hERG liability as observed with 1 and represents a promising lead for further development.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glycine/analogs & derivatives , Histone Demethylases/antagonists & inhibitors , Leukemia/drug therapy , Spiro Compounds/pharmacology , Biomarkers , Cell Line, Tumor , Computer Simulation , Drug Design , Drug Discovery , Ether-A-Go-Go Potassium Channels/drug effects , Glycine/chemical synthesis , Glycine/pharmacology , High-Throughput Screening Assays , Humans , Models, Molecular , Molecular Docking Simulation , Spiro Compounds/chemical synthesis , Structure-Activity Relationship , Tranylcypromine/analogs & derivatives , Tranylcypromine/chemistry , Tranylcypromine/pharmacology
13.
Bioorg Med Chem Lett ; 27(14): 3190-3195, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28545974

ABSTRACT

A series of reversible inhibitors of lysine specific demethylase 1 (LSD1) with a 5-hydroxypyrazole scaffold have been developed from compound 7, which was identified from the patent literature. Surface plasmon resonance (SPR) and biochemical analysis showed it to be a reversible LSD1 inhibitor with an IC50 value of 0.23µM. Optimisation of this compound by rational design afforded compounds with Kd values of <10nM. In human THP-1 cells, these compounds were found to upregulate the expression of the surrogate cellular biomarker CD86. Compound 11p was found to have moderate oral bioavailability in mice suggesting its potential for use as an in vivo tool compound.


Subject(s)
Histone Demethylases/antagonists & inhibitors , Pyrazoles/chemistry , Animals , B7-2 Antigen/metabolism , Binding Sites , Catalytic Domain , Cell Differentiation/drug effects , Cell Line , Half-Life , Histone Demethylases/metabolism , Humans , Inhibitory Concentration 50 , Mice , Molecular Docking Simulation , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Structure-Activity Relationship , Surface Plasmon Resonance
14.
J Med Chem ; 59(24): 11120-11137, 2016 12 22.
Article in English | MEDLINE | ID: mdl-28002956

ABSTRACT

A collaborative high throughput screen of 1.35 million compounds against mutant (R132H) isocitrate dehydrogenase IDH1 led to the identification of a novel series of inhibitors. Elucidation of the bound ligand crystal structure showed that the inhibitors exhibited a novel binding mode in a previously identified allosteric site of IDH1 (R132H). This information guided the optimization of the series yielding submicromolar enzyme inhibitors with promising cellular activity. Encouragingly, one compound from this series was found to induce myeloid differentiation in primary human IDH1 R132H AML cells in vitro.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Isocitrate Dehydrogenase/antagonists & inhibitors , Leukemia, Myeloid, Acute/enzymology , Allosteric Regulation/drug effects , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Isocitrate Dehydrogenase/isolation & purification , Isocitrate Dehydrogenase/metabolism , Leukemia, Myeloid, Acute/pathology , Models, Molecular , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured
15.
F1000Res ; 5: 962, 2016.
Article in English | MEDLINE | ID: mdl-27703665

ABSTRACT

We present IncucyteDRC, an R package for the analysis of data from live cell imaging cell proliferation experiments carried out on the Essen Biosciences IncuCyte ZOOM instrument. The package provides a simple workflow for summarising data into a form that can be used to calculate dose response curves and EC50 values for small molecule inhibitors. Data from different cell lines, or cell lines grown under different conditions, can be normalised as to their doubling time. A simple graphical web interface, implemented using shiny, is provided for the benefit of non-R users. The software is potentially useful to any research group studying the impact of small molecule inhibitors on cell proliferation using the IncuCyte ZOOM.

16.
ACS Chem Biol ; 11(11): 3179-3190, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27689388

ABSTRACT

The enzyme poly(ADP-ribose) glycohydrolase (PARG) performs a critical role in the repair of DNA single strand breaks (SSBs). However, a detailed understanding of its mechanism of action has been hampered by a lack of credible, cell-active chemical probes. Herein, we demonstrate inhibition of PARG with a small molecule, leading to poly(ADP-ribose) (PAR) chain persistence in intact cells. Moreover, we describe two advanced, and chemically distinct, cell-active tool compounds with convincing on-target pharmacology and selectivity. Using one of these tool compounds, we demonstrate pharmacology consistent with PARG inhibition. Further, while the roles of PARG and poly(ADP-ribose) polymerase (PARP) are closely intertwined, we demonstrate that the pharmacology of a PARG inhibitor differs from that observed with the more thoroughly studied PARP inhibitor olaparib. We believe that these tools will facilitate a wider understanding of this important component of DNA repair and may enable the development of novel therapeutic agents exploiting the critical dependence of tumors on the DNA damage response (DDR).


Subject(s)
DNA Repair , Glycoside Hydrolases/chemistry , Molecular Probes/chemistry , Phthalazines/pharmacology , Piperazines/pharmacology , Enzyme Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , HeLa Cells , Humans , Surface Plasmon Resonance
17.
F1000Res ; 5: 736, 2016.
Article in English | MEDLINE | ID: mdl-27610220

ABSTRACT

After a DNA damage signal multiple polymers of ADP ribose attached to poly(ADP) ribose (PAR) polymerases (PARPs) are broken down by the enzyme poly(ADP) ribose glycohydrolase (PARG). Inhibition of PARG leads to a failure of DNA repair and small molecule inhibition of PARG has been a goal for many years. To determine whether biochemical inhibitors of PARG are active in cells we have designed an immunofluorescence assay to detect nuclear PAR after DNA damage. This 384-well assay is suitable for medium throughput high-content screening and can detect cell-permeable inhibitors of PARG from nM to µM potency. In addition, the assay has been shown to work in murine cells and in a variety of human cancer cells. Furthermore, the assay is suitable for detecting the DNA damage response induced by treatment with temozolomide and methylmethane sulfonate (MMS). Lastly, the assay has been shown to be robust over a period of several years.

18.
F1000Res ; 5: 1005, 2016.
Article in English | MEDLINE | ID: mdl-27429741

ABSTRACT

RET (REarranged during Transfection) is a receptor tyrosine kinase, which plays pivotal roles in regulating cell survival, differentiation, proliferation, migration and chemotaxis. Activation of RET is a mechanism of oncogenesis in medullary thyroid carcinomas where both germline and sporadic activating somatic mutations are prevalent. At present, there are no known specific RET inhibitors in clinical development, although many potent inhibitors of RET have been opportunistically identified through selectivity profiling of compounds initially designed to target other tyrosine kinases. Vandetanib and cabozantinib, both multi-kinase inhibitors with RET activity, are approved for use in medullary thyroid carcinoma, but additional pharmacological activities, most notably inhibition of vascular endothelial growth factor - VEGFR2 (KDR), lead to dose-limiting toxicity. The recent identification of RET fusions present in ~1% of lung adenocarcinoma patients has renewed interest in the identification and development of more selective RET inhibitors lacking the toxicities associated with the current treatments. In an earlier publication [Newton et al, 2016; 1] we reported the discovery of a series of 2-substituted phenol quinazolines as potent and selective RET kinase inhibitors. Here we describe the development of the robust screening cascade which allowed the identification and advancement of this chemical series.  Furthermore we have profiled a panel of RET-active clinical compounds both to validate the cascade and to confirm that none display a RET-selective target profile.

19.
Anal Biochem ; 503: 58-64, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27036617

ABSTRACT

Poly(ADP-ribose) (PAR) polymers are transient post-translational modifications, and their formation is catalyzed by poly(ADP-ribose) polymerase (PARP) enzymes. A number of PARP inhibitors are in advanced clinical development for BRCA-mutated breast cancer, and olaparib has recently been approved for BRCA-mutant ovarian cancer; however, there has already been evidence of developed resistance mechanisms. Poly(ADP-ribose) glycohydrolase (PARG) catalyzes the hydrolysis of the endo- and exo-glycosidic bonds within the PAR polymers. As an alternative strategy, PARG is a potentially attractive therapeutic target. There is only one PARG gene, compared with 17 known PARP family members, and therefore a PARG inhibitor may have wider application with fewer compensatory mechanisms. Prior to the initiation of this project, there were no known existing cell-permeable small molecule PARG inhibitors for use as tool compounds to assess these hypotheses and no suitable high-throughput screening (HTS)-compatible biochemical assays available to identify start points for a drug discovery project. The development of this newly described high-throughput homogeneous time-resolved fluorescence (HTRF) assay has allowed HTS to proceed and, from this, the identification and advancement of multiple validated series of tool compounds for PARG inhibition.


Subject(s)
Fluorescence , Glycoside Hydrolases/metabolism , High-Throughput Screening Assays/methods , Luminescent Measurements/methods , Cell Line , Enzyme Inhibitors/pharmacology , Glycoside Hydrolases/analysis , Glycoside Hydrolases/antagonists & inhibitors , Humans , Structure-Activity Relationship , Time Factors
20.
Biochem J ; 473(13): 1869-79, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27099339

ABSTRACT

Tyrosyl-DNA phosphodiesterase 2 (TDP2) is a 5'-tyrosyl DNA phosphodiesterase important for the repair of DNA adducts generated by non-productive (abortive) activity of topoisomerase II (TOP2). TDP2 facilitates therapeutic resistance to topoisomerase poisons, which are widely used in the treatment of a range of cancer types. Consequently, TDP2 is an interesting target for the development of small molecule inhibitors that could restore sensitivity to topoisomerase-directed therapies. Previous studies identified a class of deazaflavin-based molecules that showed inhibitory activity against TDP2 at therapeutically useful concentrations, but their mode of action was uncertain. We have confirmed that the deazaflavin series inhibits TDP2 enzyme activity in a fluorescence-based assay, suitable for high-throughput screen (HTS)-screening. We have gone on to determine crystal structures of these compounds bound to a 'humanized' form of murine TDP2. The structures reveal their novel mode of action as competitive ligands for the binding site of an incoming DNA substrate, and point the way to generating novel and potent inhibitors of TDP2.


Subject(s)
Phosphoric Diester Hydrolases/metabolism , Riboflavin/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Enzyme Activation/drug effects , Humans , Mice , Phosphoric Diester Hydrolases/chemistry , Protein Binding , Protein Denaturation , Protein Structure, Secondary , Protein Structure, Tertiary , Riboflavin/analogs & derivatives , Riboflavin/pharmacology , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...