Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Int J Ment Health Nurs ; 32(5): 1355-1364, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37231985

ABSTRACT

The provision of community-based space for people experiencing a mental health crisis is regarded as a favourable alternative to the emergency department. However, the only non-emergency department safe spaces in Western Australia are located within hospitals or hospital grounds. This qualitative study asked mental health consumers in Western Australia with experience of presentation at the emergency department during a mental health crisis to describe what a safe space would look and feel like. Data were collected through focus groups and thematically analysed. The findings present the voices of mental health consumers through the framework of health geography and the therapeutic landscape. These participants articulated important physical and social features of a therapeutic safe space and their symbolism as inclusive, accessible places where they would experience a sense of agency and belonging. Participants also expressed a need for trained peer support within the space to complement the skilled professional mental health team. Participants' experiences of the emergency department during mental health crises were described as contrary to their recovery needs. The research reinforces the need for an alternative to the emergency department for adults who experience mental health crises and provides consumer-led evidence to inform the design and development of a recovery-focused safe space.


Subject(s)
Mental Disorders , Mental Health Services , Adult , Humans , Mental Health , Qualitative Research , Focus Groups , Counseling , Mental Disorders/therapy
2.
RSC Med Chem ; 11(6): 707-731, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-33479670

ABSTRACT

Renewed interest in covalent inhibitors of enzymes implicated in disease states has afforded several agents targeted at protein kinases of relevance to cancers. We now report the design, synthesis and biological evaluation of 6-ethynylpurines that act as covalent inhibitors of Nek2 by capturing a cysteine residue (Cys22) close to the catalytic domain of this protein kinase. Examination of the crystal structure of the non-covalent inhibitor 3-((6-cyclohexylmethoxy-7H-purin-2-yl)amino)benzamide in complex with Nek2 indicated that replacing the alkoxy with an ethynyl group places the terminus of the alkyne close to Cys22 and in a position compatible with the stereoelectronic requirements of a Michael addition. A series of 6-ethynylpurines was prepared and a structure activity relationship (SAR) established for inhibition of Nek2. 6-Ethynyl-N-phenyl-7H-purin-2-amine [IC50 0.15 µM (Nek2)] and 4-((6-ethynyl-7H-purin-2-yl)amino)benzenesulfonamide (IC50 0.14 µM) were selected for determination of the mode of inhibition of Nek2, which was shown to be time-dependent, not reversed by addition of ATP and negated by site directed mutagenesis of Cys22 to alanine. Replacement of the ethynyl group by ethyl or cyano abrogated activity. Variation of substituents on the N-phenyl moiety for 6-ethynylpurines gave further SAR data for Nek2 inhibition. The data showed little correlation of activity with the nature of the substituent, indicating that after sufficient initial competitive binding to Nek2 subsequent covalent modification of Cys22 occurs in all cases. A typical activity profile was that for 2-(3-((6-ethynyl-9H-purin-2-yl)amino)phenyl)acetamide [IC50 0.06 µM (Nek2); GI50 (SKBR3) 2.2 µM] which exhibited >5-10-fold selectivity for Nek2 over other kinases; it also showed > 50% growth inhibition at 10 µM concentration against selected breast and leukaemia cell lines. X-ray crystallographic analysis confirmed that binding of the compound to the Nek2 ATP-binding site resulted in covalent modification of Cys22. Further studies confirmed that 2-(3-((6-ethynyl-9H-purin-2-yl)amino)phenyl)acetamide has the attributes of a drug-like compound with good aqueous solubility, no inhibition of hERG at 25 µM and a good stability profile in human liver microsomes. It is concluded that 6-ethynylpurines are promising agents for cancer treatment by virtue of their selective inhibition of Nek2.

4.
Oncotarget ; 8(12): 19089-19124, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-27833088

ABSTRACT

Nek2 (NIMA-related kinase 2) is a cell cycle-dependent serine/threonine protein kinase that regulates centrosome separation at the onset of mitosis. Overexpression of Nek2 is common in human cancers and suppression can restrict tumor cell growth and promote apoptosis. Nek2 inhibition with small molecules, therefore, offers the prospect of a new therapy for cancer. To achieve this goal, a better understanding of the requirements for selective-inhibition of Nek2 is required. 6-Alkoxypurines were identified as ATP-competitive inhibitors of Nek2 and CDK2. Comparison with CDK2-inhibitor structures indicated that judicious modification of the 6-alkoxy and 2-arylamino substituents could achieve discrimination between Nek2 and CDK2. In this study, a library of 6-cyclohexylmethoxy-2-arylaminopurines bearing carboxamide, sulfonamide and urea substituents on the 2-arylamino ring was synthesized. Few of these compounds were selective for Nek2 over CDK2, with the best result being obtained for 3-((6-(cyclohexylmethoxy)-9H-purin-2-yl)amino)-N,N-dimethylbenzamide (CDK2 IC50 = 7.0 µM; Nek2 IC50 = 0.62 µM) with >10-fold selectivity. Deletion of the 6-substituent abrogated activity against both Nek2 and CDK2. Nine compounds containing an (E)-dialkylaminovinyl substituent at C-6, all showed selectivity for Nek2, e.g. (E)-6-(2-(azepan-1-yl)vinyl)-N-phenyl-9H-purin-2-amine (CDK2 IC50 = 2.70 µM; Nek2 IC50 = 0.27 µM). Structural biology of selected compounds enabled a partial rationalization of the observed structure activity relationships and mechanism of Nek2 activation. This showed that carboxamide 11 is the first reported inhibitor of Nek2 in the DFG-in conformation.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , NIMA-Related Kinases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Magnetic Resonance Spectroscopy , Structure-Activity Relationship
5.
Oncotarget ; 7(3): 2329-42, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26295308

ABSTRACT

CCT245737 is the first orally active, clinical development candidate CHK1 inhibitor to be described. The IC50 was 1.4 nM against CHK1 enzyme and it exhibited>1,000-fold selectivity against CHK2 and CDK1. CCT245737 potently inhibited cellular CHK1 activity (IC50 30-220 nM) and enhanced gemcitabine and SN38 cytotoxicity in multiple human tumor cell lines and human tumor xenograft models. Mouse oral bioavailability was complete (100%) with extensive tumor exposure. Genotoxic-induced CHK1 activity (pS296 CHK1) and cell cycle arrest (pY15 CDK1) were inhibited both in vitro and in human tumor xenografts by CCT245737, causing increased DNA damage and apoptosis. Uniquely, we show CCT245737 enhanced gemcitabine antitumor activity to a greater degree than for higher doses of either agent alone, without increasing toxicity, indicating a true therapeutic advantage for this combination. Furthermore, development of a novel ELISA assay for pS296 CHK1 autophosphorylation, allowed the quantitative measurement of target inhibition in a RAS mutant human tumor xenograft of NSCLC at efficacious doses of CCT245737. Finally, CCT245737 also showed significant single-agent activity against a MYC-driven mouse model of B-cell lymphoma. In conclusion, CCT245737 is a new CHK1 inhibitor clinical development candidate scheduled for a first in man Phase I clinical trial, that will use the novel pS296 CHK1 ELISA to monitor target inhibition.


Subject(s)
4-Aminopyridine/analogs & derivatives , Carcinoma, Non-Small-Cell Lung/drug therapy , Checkpoint Kinase 1/drug effects , Lung Neoplasms/drug therapy , Lymphoma, B-Cell/drug therapy , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyrazines/pharmacology , Xenograft Model Antitumor Assays , 4-Aminopyridine/pharmacokinetics , 4-Aminopyridine/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , CDC2 Protein Kinase , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA Damage/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Synergism , HT29 Cells , Humans , Irinotecan , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Pyrazines/pharmacokinetics , Gemcitabine
6.
Cancer Res ; 75(16): 3340-54, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26202014

ABSTRACT

Acquired resistance to therapy is perhaps the greatest challenge to effective clinical management of cancer. With several inhibitors of the mitotic checkpoint kinase MPS1 in preclinical development, we sought to investigate how resistance against these inhibitors may arise so that mitigation or bypass strategies could be addressed as early as possible. Toward this end, we modeled acquired resistance to the MPS1 inhibitors AZ3146, NMS-P715, and CCT251455, identifying five point mutations in the kinase domain of MPS1 that confer resistance against multiple inhibitors. Structural studies showed how the MPS1 mutants conferred resistance by causing steric hindrance to inhibitor binding. Notably, we show that these mutations occur in nontreated cancer cell lines and primary tumor specimens, and that they also preexist in normal lymphoblast and breast tissues. In a parallel piece of work, we also show that the EGFR p.T790M mutation, the most common mutation conferring resistance to the EGFR inhibitor gefitinib, also preexists in cancer cells and normal tissue. Our results therefore suggest that mutations conferring resistance to targeted therapy occur naturally in normal and malignant cells and these mutations do not arise as a result of the increased mutagenic plasticity of cancer cells.


Subject(s)
Cell Cycle Proteins/genetics , Drug Resistance, Neoplasm/genetics , Point Mutation , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Aniline Compounds/chemistry , Aniline Compounds/metabolism , Aniline Compounds/pharmacology , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Dose-Response Relationship, Drug , ErbB Receptors/antagonists & inhibitors , Gefitinib , HCT116 Cells , HEK293 Cells , Heterocyclic Compounds, 2-Ring/chemistry , Heterocyclic Compounds, 2-Ring/metabolism , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Models, Molecular , Molecular Structure , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/metabolism , Pyrazoles/chemistry , Pyrazoles/metabolism , Pyrazoles/pharmacology , Quinazolines/chemistry , Quinazolines/metabolism , Quinazolines/pharmacology
7.
J Med Chem ; 56(24): 10045-65, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24256217

ABSTRACT

The protein kinase MPS1 is a crucial component of the spindle assembly checkpoint signal and is aberrantly overexpressed in many human cancers. MPS1 is one of the top 25 genes overexpressed in tumors with chromosomal instability and aneuploidy. PTEN-deficient breast tumor cells are particularly dependent upon MPS1 for their survival, making it a target of significant interest in oncology. We report the discovery and optimization of potent and selective MPS1 inhibitors based on the 1H-pyrrolo[3,2-c]pyridine scaffold, guided by structure-based design and cellular characterization of MPS1 inhibition, leading to 65 (CCT251455). This potent and selective chemical tool stabilizes an inactive conformation of MPS1 with the activation loop ordered in a manner incompatible with ATP and substrate-peptide binding; it displays a favorable oral pharmacokinetic profile, shows dose-dependent inhibition of MPS1 in an HCT116 human tumor xenograft model, and is an attractive tool compound to elucidate further the therapeutic potential of MPS1 inhibition.


Subject(s)
Aniline Compounds/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Drug Design , Heterocyclic Compounds, 2-Ring/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Administration, Oral , Aniline Compounds/administration & dosage , Aniline Compounds/chemistry , Biological Availability , Cell Cycle Proteins/metabolism , Dose-Response Relationship, Drug , Heterocyclic Compounds, 2-Ring/administration & dosage , Heterocyclic Compounds, 2-Ring/chemistry , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship
8.
Oncotarget ; 4(10): 1647-61, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24072592

ABSTRACT

The ribosomal P70 S6 kinases play a crucial role in PI3K/mTOR regulated signalling pathways and are therefore potential targets for the treatment of a variety of diseases including diabetes and cancer. In this study we describe the identification of three series of chemically distinct S6K1 inhibitors. In addition, we report a novel PKA-S6K1 chimeric protein with five mutations in or near its ATP-binding site, which was used to determine the binding mode of two of the three inhibitor series, and provided a robust system to aid the optimisation of the oxadiazole-substituted benzimidazole inhibitor series. We show that the resulting oxadiazole-substituted aza-benzimidazole is a potent and ligand efficient S6 kinase inhibitor, which blocks the phosphorylation of RPS6 at Ser235/236 in TSC negative HCV29 human bladder cancer cells by inhibiting S6 kinase activity and thus provides a useful tool compound to investigate the function of S6 kinases.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Drug Design , High-Throughput Screening Assays/methods , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Models, Molecular , Phosphorylation , Protein Kinase Inhibitors/chemistry , Recombinant Fusion Proteins/chemistry , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction , Structure-Activity Relationship , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/enzymology
9.
PLoS One ; 8(6): e65689, 2013.
Article in English | MEDLINE | ID: mdl-23776527

ABSTRACT

Checkpoint kinase 2 (CHK2) is an important serine/threonine kinase in the cellular response to DNA damage. A fragment-based screening campaign using a combination of a high-concentration AlphaScreen™ kinase assay and a biophysical thermal shift assay, followed by X-ray crystallography, identified a number of chemically different ligand-efficient CHK2 hinge-binding scaffolds that have not been exploited in known CHK2 inhibitors. In addition, it showed that the use of these orthogonal techniques allowed efficient discrimination between genuine hit matter and false positives from each individual assay technology. Furthermore, the CHK2 crystal structures with a quinoxaline-based fragment and its follow-up compound highlight a hydrophobic area above the hinge region not previously explored in rational CHK2 inhibitor design, but which might be exploited to enhance both potency and selectivity of CHK2 inhibitors.


Subject(s)
Adenosine Triphosphate/metabolism , Checkpoint Kinase 2/chemistry , Checkpoint Kinase 2/metabolism , Models, Molecular , Protein Conformation , Binding Sites/genetics , Checkpoint Kinase 2/antagonists & inhibitors , Crystallography , Crystallography, X-Ray , Molecular Structure , Protein Kinase Inhibitors/chemistry
10.
J Med Chem ; 55(22): 10229-40, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23082860

ABSTRACT

Inhibitors of checkpoint kinase 1 (CHK1) are of current interest as potential antitumor agents, but the most advanced inhibitor series reported to date are not orally bioavailable. A novel series of potent and orally bioavailable 3-alkoxyamino-5-(pyridin-2-ylamino)pyrazine-2-carbonitrile CHK1 inhibitors was generated by hybridization of two lead scaffolds derived from fragment-based drug design and optimized for CHK1 potency and high selectivity using a cell-based assay cascade. Efficient in vivo pharmacokinetic assessment was used to identify compounds with prolonged exposure following oral dosing. The optimized compound (CCT244747) was a potent and highly selective CHK1 inhibitor, which modulated the DNA damage response pathway in human tumor xenografts and showed antitumor activity in combination with genotoxic chemotherapies and as a single agent.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Neuroblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Pyrimidines/pharmacology , Administration, Oral , Aminopyridines/chemical synthesis , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Checkpoint Kinase 1 , Child , Colonic Neoplasms/enzymology , DNA Damage/drug effects , Drug Design , Humans , Mice , Mice, Nude , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neuroblastoma/enzymology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemical synthesis , Protein Kinases/metabolism , Pyrimidines/chemical synthesis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 18(20): 5650-61, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22929806

ABSTRACT

PURPOSE: Many tumors exhibit defective cell-cycle checkpoint control and increased replicative stress. CHK1 is critically involved in the DNA damage response and maintenance of replication fork stability. We have therefore discovered a novel potent, highly selective, orally active ATP-competitive CHK1 inhibitor, CCT244747, and present its preclinical pharmacology and therapeutic activity. EXPERIMENTAL DESIGN: Cellular CHK1 activity was assessed using an ELISA assay, and cytotoxicity a SRB assay. Biomarker modulation was measured using immunoblotting, and cell-cycle effects by flow cytometry analysis. Single-agent oral CCT244747 antitumor activity was evaluated in a MYCN-driven transgenic mouse model of neuroblastoma by MRI and in genotoxic combinations in human tumor xenografts by growth delay. RESULTS: CCT244747 inhibited cellular CHK1 activity (IC(50) 29-170 nmol/L), significantly enhanced the cytotoxicity of several anticancer drugs, and abrogated drug-induced S and G(2) arrest in multiple tumor cell lines. Biomarkers of CHK1 (pS296 CHK1) activity and cell-cycle inactivity (pY15 CDK1) were induced by genotoxics and inhibited by CCT244747 both in vitro and in vivo, producing enhanced DNA damage and apoptosis. Active tumor concentrations of CCT244747 were obtained following oral administration. The antitumor activity of both gemcitabine and irinotecan were significantly enhanced by CCT244747 in several human tumor xenografts, giving concomitant biomarker modulation indicative of CHK1 inhibition. CCT244747 also showed marked antitumor activity as a single agent in a MYCN-driven neuroblastoma. CONCLUSION: CCT244747 represents the first structural disclosure of a highly selective, orally active CHK1 inhibitor and warrants further evaluation alone or combined with genotoxic anticancer therapies.


Subject(s)
Aminopyridines/administration & dosage , Neoplasms, Experimental , Neuroblastoma , Protein Kinases , Pyrimidines/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Checkpoints/drug effects , Checkpoint Kinase 1 , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Transgenic , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinases/genetics , Protein Kinases/metabolism
12.
J Med Chem ; 55(7): 3228-41, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22404346
13.
J Med Chem ; 54(24): 8328-42, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22111927

ABSTRACT

Pyrazolopyridine inhibitors with low micromolar potency for CHK1 and good selectivity against CHK2 were previously identified by fragment-based screening. The optimization of the pyrazolopyridines to a series of potent and CHK1-selective isoquinolines demonstrates how fragment-growing and scaffold morphing strategies arising from a structure-based understanding of CHK1 inhibitor binding can be combined to successfully progress fragment-derived hit matter to compounds with activity in vivo. The challenges of improving CHK1 potency and selectivity, addressing synthetic tractability, and achieving novelty in the crowded kinase inhibitor chemical space were tackled by multiple scaffold morphing steps, which progressed through tricyclic pyrimido[2,3-b]azaindoles to N-(pyrazin-2-yl)pyrimidin-4-amines and ultimately to imidazo[4,5-c]pyridines and isoquinolines. A potent and highly selective isoquinoline CHK1 inhibitor (SAR-020106) was identified, which potentiated the efficacies of irinotecan and gemcitabine in SW620 human colon carcinoma xenografts in nude mice.


Subject(s)
Antineoplastic Agents/chemical synthesis , Isoquinolines/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Protein Kinases/metabolism , Pyrazines/chemical synthesis , Pyridines/chemical synthesis , Adenosine Triphosphate/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Biological Availability , Cell Line, Tumor , Checkpoint Kinase 1 , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Isoquinolines/chemistry , Isoquinolines/pharmacology , Mice , Mice, Nude , Molecular Conformation , Neoplasm Transplantation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazines/chemistry , Pyrazines/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Stereoisomerism , Structure-Activity Relationship , Transplantation, Heterologous
14.
J Med Chem ; 54(6): 1626-39, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21366329

ABSTRACT

We describe herein the structure-activity relationship (SAR) and cocrystal structures of a series of Nek2 inhibitors derived from the published polo-like kinase 1 (Plk1) inhibitor (R)-1. Our studies reveal a nonlinear SAR for Nek2 and our cocrystal structures show that compounds in this series bind to a DFG-out conformation of Nek2 without extending into the enlarged back pocket commonly found in this conformation. These observations were further investigated, and structure-based design led to Nek2 inhibitors derived from (R)-1 with more than a hundred-fold selectivity against Plk1.


Subject(s)
Benzamides/chemical synthesis , Benzimidazoles/chemical synthesis , Models, Molecular , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Benzamides/chemistry , Benzamides/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Binding Sites , Crystallography, X-Ray , Female , Humans , In Vitro Techniques , Male , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , NIMA-Related Kinases , Phosphorylation , Protein Binding , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Stereoisomerism , Structure-Activity Relationship
15.
J Med Chem ; 54(2): 580-90, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21186793

ABSTRACT

Structure-based design was applied to the optimization of a series of 2-(quinazolin-2-yl)phenols to generate potent and selective ATP-competitive inhibitors of the DNA damage response signaling enzyme checkpoint kinase 2 (CHK2). Structure-activity relationships for multiple substituent positions were optimized separately and in combination leading to the 2-(quinazolin-2-yl)phenol 46 (IC(50) 3 nM) with good selectivity for CHK2 against CHK1 and a wider panel of kinases and with promising in vitro ADMET properties. Off-target activity at hERG ion channels shown by the core scaffold was successfully reduced by the addition of peripheral polar substitution. In addition to showing mechanistic inhibition of CHK2 in HT29 human colon cancer cells, a concentration dependent radioprotective effect in mouse thymocytes was demonstrated for the potent inhibitor 46 (CCT241533).


Subject(s)
Phenols/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Quinazolines/chemical synthesis , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line , Checkpoint Kinase 2 , Crystallography, X-Ray , Drug Design , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/metabolism , HT29 Cells , Humans , In Vitro Techniques , Mice , Models, Molecular , Phenols/chemistry , Phenols/pharmacology , Protein Binding , Quinazolines/chemistry , Quinazolines/pharmacology , Radiation-Protective Agents/chemical synthesis , Radiation-Protective Agents/chemistry , Radiation-Protective Agents/pharmacology , Stereoisomerism , Structure-Activity Relationship , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/radiation effects
16.
J Med Chem ; 53(21): 7682-98, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20936789

ABSTRACT

We report herein the first systematic exploration of inhibitors of the mitotic kinase Nek2. Starting from HTS hit aminopyrazine 2, compounds with improved activity were identified using structure-based design. Our structural biology investigations reveal two notable observations. First, 2 and related compounds bind to an unusual, inactive conformation of the kinase which to the best of our knowledge has not been reported for other types of kinase inhibitors. Second, a phenylalanine residue at the center of the ATP pocket strongly affects the ability of the inhibitor to bind to the protein. The implications of these observations are discussed, and the work described here defines key features for potent and selective Nek2 inhibition, which will aid the identification of more advanced inhibitors of Nek2.


Subject(s)
Models, Molecular , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazines/chemical synthesis , Crystallography, X-Ray , Humans , NIMA-Related Kinases , Phosphorylation , Protein Binding , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Pyrazines/chemistry , Stereoisomerism , Structure-Activity Relationship
17.
Bioorg Med Chem Lett ; 20(14): 4045-9, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20561787

ABSTRACT

A range of 3,6-di(hetero)arylimidazo[1,2-a]pyrazine ATP-competitive inhibitors of CHK1 were developed by scaffold hopping from a weakly active screening hit. Efficient synthetic routes for parallel synthesis were developed to prepare analogues with improved potency and ligand efficiency against CHK1. Kinase profiling showed that the imidazo[1,2-a]pyrazines could inhibit other kinases, including CHK2 and ABL, with equivalent or better potency depending on the pendant substitution. These 3,6-di(hetero)aryl imidazo[1,2-a]pyrazines appear to represent a general kinase inhibitor scaffold.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazines/chemistry , Pyrazines/pharmacology , Crystallography, X-Ray , Drug Design , Drug Evaluation, Preclinical
18.
Mol Cancer Ther ; 9(1): 89-100, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20053762

ABSTRACT

Genotoxic antitumor agents continue to be the mainstay of current cancer chemotherapy. These drugs cause DNA damage and activate numerous cell cycle checkpoints facilitating DNA repair and the maintenance of genomic integrity. Most human tumors lack functional p53 and consequently have compromised G(1)-S checkpoint control. This has led to the hypothesis that S and G(2)-M checkpoint abrogation may selectively enhance genotoxic cell killing in a p53-deficient background, as normal cells would be rescued at the G(1)-S checkpoint. CHK1 is a serine/threonine kinase associated with DNA damage-linked S and G(2)-M checkpoint control. SAR-020106 is an ATP-competitive, potent, and selective CHK1 inhibitor with an IC(50) of 13.3 nmol/L on the isolated human enzyme. This compound abrogates an etoposide-induced G(2) arrest with an IC(50) of 55 nmol/L in HT29 cells, and significantly enhances the cell killing of gemcitabine and SN38 by 3.0- to 29-fold in several colon tumor lines in vitro and in a p53-dependent fashion. Biomarker studies have shown that SAR-020106 inhibits cytotoxic drug-induced autophosphorylation of CHK1 at S296 and blocks the phosphorylation of CDK1 at Y15 in a dose-dependent fashion both in vitro and in vivo. Cytotoxic drug combinations were associated with increased gammaH2AX and poly ADP ribose polymerase cleavage consistent with the SAR-020106-enhanced DNA damage and tumor cell death. Irinotecan and gemcitabine antitumor activity was enhanced by SAR-020106 in vivo with minimal toxicity. SAR-020106 represents a novel class of CHK1 inhibitors that can enhance antitumor activity with selected anticancer drugs in vivo and may therefore have clinical utility.


Subject(s)
Isoquinolines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Pyrazines/pharmacology , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/metabolism , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Checkpoint Kinase 1 , DNA Damage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Synergism , G2 Phase/drug effects , Humans , Irinotecan , Isoquinolines/administration & dosage , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Mice , Mice, Nude , Mutagens/toxicity , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrazines/administration & dosage , Pyrazines/chemistry , Pyrazines/pharmacokinetics , Gemcitabine
19.
Bioorg Med Chem ; 18(2): 707-18, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20022510

ABSTRACT

5-(Hetero)aryl-3-(4-carboxamidophenyl)-2-aminopyridine inhibitors of CHK2 were identified from high throughput screening of a kinase-focussed compound library. Rapid exploration of the hits through straightforward chemistry established structure-activity relationships and a proposed ATP-competitive binding mode which was verified by X-ray crystallography of several analogues bound to CHK2. Variation of the 5-(hetero)aryl substituent identified bicyclic dioxolane and dioxane groups which improved the affinity and the selectivity of the compounds for CHK2 versus CHK1. The 3-(4-carboxamidophenyl) substituent could be successfully replaced by acyclic omega-aminoalkylamides, which made additional polar interactions within the binding site and led to more potent inhibitors of CHK2. Compounds from this series showed activity in cell-based mechanistic assays for inhibition of CHK2.


Subject(s)
Aminopyridines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Aminopyridines/chemical synthesis , Aminopyridines/chemistry , Binding Sites , Cell Line , Checkpoint Kinase 2 , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Stereoisomerism , Structure-Activity Relationship
20.
J Med Chem ; 52(15): 4810-9, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19572549

ABSTRACT

Checkpoint kinase 1 (CHK1) is an oncology target of significant current interest. Inhibition of CHK1 abrogates DNA damage-induced cell cycle checkpoints and sensitizes p53 deficient cancer cells to genotoxic therapies. Using template screening, a fragment-based approach to small molecule hit generation, we have identified multiple CHK1 inhibitor scaffolds suitable for further optimization. The sequential combination of in silico low molecular weight template selection, a high concentration biochemical assay and hit validation through protein-ligand X-ray crystallography provided 13 template hits from an initial in silico screening library of ca. 15000 compounds. The use of appropriate counter-screening to rule out nonspecific aggregation by test compounds was essential for optimum performance of the high concentration bioassay. One low molecular weight, weakly active purine template hit was progressed by iterative structure-based design to give submicromolar pyrazolopyridines with good ligand efficiency and appropriate CHK1-mediated cellular activity in HT29 colon cancer cells.


Subject(s)
Protein Kinase Inhibitors/chemical synthesis , Protein Kinases/drug effects , Checkpoint Kinase 1 , Drug Evaluation, Preclinical , HT29 Cells , Humans , Hydrogen Bonding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...