Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Nat Chem Biol ; 12(11): 931-936, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27595327

ABSTRACT

Targeted covalent inhibition of disease-associated proteins has become a powerful methodology in the field of drug discovery, leading to the approval of new therapeutics. Nevertheless, current approaches are often limited owing to their reliance on a cysteine residue to generate the covalent linkage. Here we used aryl boronic acid carbonyl warheads to covalently target a noncatalytic lysine side chain, and generated to our knowledge the first reversible covalent inhibitors for Mcl-1, a protein-protein interaction (PPI) target that has proven difficult to inhibit via traditional medicinal chemistry strategies. These covalent binders exhibited improved potency in comparison to noncovalent congeners, as demonstrated in biochemical and cell-based assays. We identified Lys234 as the residue involved in covalent modification, via point mutation. The covalent binders discovered in this study will serve as useful starting points for the development of Mcl-1 therapeutics and probes to interrogate Mcl-1-dependent biological phenomena.


Subject(s)
Boronic Acids/chemistry , Boronic Acids/pharmacology , Lysine/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Boronic Acids/chemical synthesis , Dose-Response Relationship, Drug , Humans , Hydrogen-Ion Concentration , Lysine/metabolism , Molecular Structure , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Structure-Activity Relationship
2.
Bioorg Med Chem Lett ; 28(8): 1336-1341, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29559278

ABSTRACT

The design and synthesis of a novel series of 2,6-disubstituted pyrazine derivatives as CK2 kinase inhibitors is described. Structure-guided optimization of a 5-substituted-3-thiophene carboxylic acid screening hit (3a) led to the development of a lead compound (12b), which shows inhibition in both enzymatic and cellular assays. Subsequent design and hybridization efforts also led to the unexpected identification of analogs with potent PIM kinase activity (14f).


Subject(s)
Casein Kinase II/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Pyrazines/pharmacology , Cell Line, Tumor , Drug Design , Humans , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Pyrazines/chemical synthesis , Pyrazines/chemistry , Pyrazines/pharmacokinetics , Structure-Activity Relationship
3.
Blood ; 123(6): 905-13, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24363397

ABSTRACT

Upregulation of Pim kinases is observed in several types of leukemias and lymphomas. Pim-1, -2, and -3 promote cell proliferation and survival downstream of cytokine and growth factor signaling pathways. AZD1208 is a potent, highly selective, and orally available Pim kinase inhibitor that effectively inhibits all three isoforms at <5 nM or <150 nM in enzyme and cell assays, respectively. AZD1208 inhibited the growth of 5 of 14 acute myeloid leukemia (AML) cell lines tested, and sensitivity correlates with Pim-1 expression and STAT5 activation. AZD1208 causes cell cycle arrest and apoptosis in MOLM-16 cells, accompanied by a dose-dependent reduction in phosphorylation of Bcl-2 antagonist of cell death, 4EBP1, p70S6K, and S6, as well as increases in cleaved caspase 3 and p27. Inhibition of p4EBP1 and p-p70S6K and suppression of translation are the most representative effects of Pim inhibition in sensitive AML cell lines. AZD1208 inhibits the growth of MOLM-16 and KG-1a xenograft tumors in vivo with a clear pharmacodynamic-pharmacokinetic relationship. AZD1208 also potently inhibits colony growth and Pim signaling substrates in primary AML cells from bone marrow that are Flt3 wild-type or Flt3 internal tandem duplication mutant. These results underscore the therapeutic potential of Pim kinase inhibition for the treatment of AML.


Subject(s)
Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Cell Proliferation/drug effects , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Thiazolidines/pharmacology , Animals , Biphenyl Compounds/pharmacokinetics , Blotting, Western , Cell Cycle , Enzyme-Linked Immunosorbent Assay , Female , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Mice , Mice, SCID , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-pim-1/metabolism , Thiazolidines/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured
4.
Bioorg Med Chem Lett ; 26(19): 4775-4780, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27578247

ABSTRACT

During the lead generation and optimization of PARP inhibitors blocking centrosome clustering, it was discovered that increasing hydrogen bond acceptor (HBA) strength improved cellular potency but led to elevated Caco2 and MDR1 efflux and thus poor oral bioavailability. Conversely, compounds with lower efflux had reduced potency. The project team was able to improve the bioavailability by reducing efflux through systematic modifications to the strength of the HBA by changing the electronic properties of neighboring groups, whilst maintaining sufficient acceptor strength for potency. Additionally, it was observed that enantiomers with different potency showed similar efflux, which is consistent with the promiscuity of efflux transporters. Eventually, a balance between potency and low efflux was achieved for a set of lead compounds with good bioavailability which allowed the project to progress towards establishing in vivo pharmacokinetic/pharmacodynamic relationships.


Subject(s)
Centrosome/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Dogs , Humans , Hydrogen Bonding , Madin Darby Canine Kidney Cells , Mice , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Rats
5.
Bioorg Med Chem Lett ; 25(24): 5743-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26546219

ABSTRACT

The propensity for cancer cells to accumulate additional centrosomes relative to normal cells could be exploited for therapeutic benefit in oncology. Following literature reports that suggested TNKS1 (tankyrase 1) and PARP16 may be involved with spindle structure and function and may play a role in suppressing multi-polar spindle formation in cells with supernumerary centrosomes, we initiated a phenotypic screen to look for small molecule poly (ADP-ribose) polymerase (PARP) enzyme family inhibitors that could produce a multi-polar spindle phenotype via declustering of centrosomes. Screening of AstraZeneca's collection of phthalazinone PARP inhibitors in HeLa cells using high-content screening techniques identified several compounds that produced a multi-polar spindle phenotype at low nanomolar concentrations. Characterization of these compounds across a broad panel of PARP family enzyme assays indicated that they had activity against several PARP family enzymes, including PARP1, 2, 3, 5a, 5b, and 6. Further optimization of these initial hits for improved declustering potency, solubility, permeability, and oral bioavailability resulted in AZ0108, a PARP1, 2, 6 inhibitor that potently inhibits centrosome clustering and is suitable for in vivo efficacy and tolerability studies.


Subject(s)
Centrosome/metabolism , Phthalazines/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Administration, Oral , Animals , Binding Sites , Caco-2 Cells , Centrosome/drug effects , Crystallography, X-Ray , Drug Evaluation, Preclinical , HeLa Cells , Humans , Microsomes/metabolism , Molecular Conformation , Molecular Dynamics Simulation , Phthalazines/administration & dosage , Phthalazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Protein Structure, Tertiary , Rats , Tankyrases/antagonists & inhibitors , Tankyrases/metabolism
6.
J Chem Inf Model ; 55(8): 1552-65, 2015 Aug 24.
Article in English | MEDLINE | ID: mdl-26176600

ABSTRACT

The SZMAP method computes binding free energies and the corresponding thermodynamic components for water molecules in the binding site of a protein structure [ SZMAP, 1.0.0 ; OpenEye Scientific Software Inc. : Santa Fe, NM, USA , 2011 ]. In this work, the ability of SZMAP to predict water structure and thermodynamic stability is examined for the X-ray crystal structures of a series of protein-ligand complexes. SZMAP results correlate with higher-level replica exchange thermodynamic integration double decoupling calculations of the absolute free energy of bound waters in the test set complexes. In addition, SZMAP calculations show good agreement with experimental data in terms of water conservation (across multiple crystal structures) and B-factors over a subset of the test set. In particular, the SZMAP neutral entropy difference term calculated at crystallographic water positions within each of the complex structures correlates well with whether that crystallographic water is conserved or displaceable. Furthermore, the calculated entropy of the water probe relative to the continuum shows a significant degree of correlation with the B-factors associated with the oxygen atoms of the water molecules. Taken together, these results indicate that SZMAP is capable of quantitatively predicting water positions and their energetics and is potentially a useful tool for determining which waters to attempt to displace, maintain, or build in through water-mediated interactions when evolving a lead series during a drug discovery program.


Subject(s)
Proteins/chemistry , Software , Thermodynamics , Water/chemistry , Bacterial Proteins/chemistry , Binding Sites , Carrier Proteins/chemistry , Crystallography, X-Ray , Databases, Protein , HIV Protease/chemistry , HIV-1/chemistry , Ligands , Lipoproteins/chemistry , Models, Molecular , Protein Binding , Salmonella enterica/chemistry
7.
J Chem Inf Model ; 54(7): 2127-38, 2014 Jul 28.
Article in English | MEDLINE | ID: mdl-24881672

ABSTRACT

Proteins are dynamic molecules, and understanding their movements, especially as they relate to molecular recognition and protein-ligand interactions, poses a significant challenge to structure-based drug discovery. In most instances, protein flexibility is underrepresented in computer-aided drug design due to uncertainties on how it should be accurately modeled as well as the computational cost associated with attempting to incorporate flexibility in the calculations. One approach that aims to address these issues is ensemble-based docking. With this technique, ligands are docked to an ensemble of rigid protein conformations. Molecular dynamics (MD) simulations can be used to generate the ensemble of protein conformations for the subsequent docking. Here we present a novel approach that uses biased-MD simulations to generate the docking ensemble. The MD simulations are biased toward an initial protein-ligand X-ray complex structure. The biasing maintains some of the original crystallographic pocket-ligand information and thereby enhances sampling of the more relevant conformational space of the protein. Resulting trajectories are clustered to select a representative set of protein conformations, and ligands are docked to that reduced set of conformations. Cross-docking to this ensemble and then selecting the lowest scoring pose enables reliable identification of the correct binding mode. Various levels of biasing are investigated, and the method is validated for cyclin-dependent kinase 2 and factor Xa.


Subject(s)
Molecular Docking Simulation , Molecular Dynamics Simulation , Cyclin-Dependent Kinase 2/chemistry , Cyclin-Dependent Kinase 2/metabolism , Drug Discovery , Factor Xa/chemistry , Factor Xa/metabolism , Ligands , Protein Conformation , Temperature
8.
J Med Chem ; 67(13): 11209-11225, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38916990

ABSTRACT

Covalent hit identification is a viable approach to identify chemical starting points against difficult-to-drug targets. While most researchers screen libraries of <2k electrophilic fragments, focusing on lead-like compounds can be advantageous in terms of finding hits with improved affinity and with a better chance of identifying cryptic pockets. However, due to the increased molecular complexity, larger numbers of compounds (>10k) are desirable to ensure adequate coverage of chemical space. Herein, the approach taken to build a library of 12k covalent lead-like compounds is reported, utilizing legacy compounds, robust library chemistry, and acquisitions. The lead-like covalent library was screened against the antiapoptotic protein Bfl-1, and six promising hits that displaced the BIM peptide from the PPI interface were identified. Intriguingly, X-ray crystallography of lead-like compound 8 showed that it binds to a previously unobserved conformation of the Bfl-1 protein and is an ideal starting point for the optimization of Bfl-1 inhibitors.


Subject(s)
Cysteine , Drug Design , Small Molecule Libraries , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Crystallography, X-Ray , Cysteine/chemistry , Humans , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , Structure-Activity Relationship , Models, Molecular , Minor Histocompatibility Antigens
9.
J Med Chem ; 67(2): 1500-1512, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38227216

ABSTRACT

Casitas B-lymphoma proto-oncogene-b (Cbl-b), a member of the Cbl family of RING finger E3 ubiquitin ligases, has been demonstrated to play a central role in regulating effector T-cell function. Multiple studies using gene-targeting approaches have provided direct evidence that Cbl-b negatively regulates T, B, and NK cell activation via a ubiquitin-mediated protein modulation. Thus, inhibition of Cbl-b ligase activity can lead to immune activation and has therapeutic potential in immuno-oncology. Herein, we describe the discovery and optimization of an arylpyridone series as Cbl-b inhibitors by structure-based drug discovery to afford compound 31. This compound binds to Cbl-b with an IC50 value of 30 nM and induces IL-2 production in T-cells with an EC50 value of 230 nM. Compound 31 also shows robust intracellular target engagement demonstrated through inhibition of Cbl-b autoubiquitination, inhibition of ubiquitin transfer to ZAP70, and the cellular modulation of phosphorylation of a downstream signal within the TCR axis.


Subject(s)
Proto-Oncogene Proteins c-cbl , Ubiquitin-Protein Ligases , Proto-Oncogene Proteins c-cbl/metabolism , Ubiquitin-Protein Ligases/metabolism , T-Lymphocytes/metabolism , Phosphorylation , Ubiquitin/metabolism
10.
ACS Med Chem Lett ; 15(6): 791-797, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38894895

ABSTRACT

Bfl-1 is overexpressed in both hematological and solid tumors; therefore, inhibitors of Bfl-1 are highly desirable. A DNA-encoded chemical library (DEL) screen against Bfl-1 identified the first known reversible covalent small-molecule ligand for Bfl-1. The binding was validated through biophysical and biochemical techniques, which confirmed the reversible covalent mechanism of action and pointed to binding through Cys55. This represented the first identification of a cyano-acrylamide reversible covalent compound from a DEL screen and highlights further opportunities for covalent drug discovery through DEL screening. A 10-fold improvement in potency was achieved through a systematic SAR exploration of the hit. The more potent analogue compound 13 was successfully cocrystallized in Bfl-1, revealing the binding mode and providing further evidence of a covalent interaction with Cys55.

11.
Bioorg Med Chem Lett ; 23(10): 3105-10, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23562594

ABSTRACT

The discovery of the activating mutation V617F in the JH2 domain of Jak2 and the modulation of oncogenic Stat3 by Jak2 inhibitors have spurred a great interest in the inhibition of the Jak2/Stat pathway in oncology. In this Letter, we communicate the discovery of novel inhibitors of the Jak2/Stat5 axis, the N-(1H-pyrazol-3-yl)pyrimidin-2-amino derivatives. The rationale, synthesis and biological evaluation of these derivatives are reported. Two lead analogs from this series, 6 and 9, displayed prolonged residence time on Jak2, at enzymatic level. Although 6 and 9 exhibited moderate selectivity in a selected kinase panel, we chose to test these inhibitors in vivo as a consequence to their long residence time. However, extended inhibition of Jak2 due to the long residence time, in the form of inhibiting phosphorylation of downstream Stat5, was not recapitulated in an in vivo setting.


Subject(s)
Drug Discovery , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , STAT5 Transcription Factor/antagonists & inhibitors , Animals , Cell Line, Transformed , Cell Proliferation/drug effects , Dogs , Dose-Response Relationship, Drug , Female , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , Mice , Mice, Inbred Strains , Models, Molecular , Molecular Conformation , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Rats , Rats, Wistar , STAT5 Transcription Factor/metabolism , Structure-Activity Relationship , Substrate Specificity , Time Factors
12.
J Med Chem ; 66(13): 8782-8807, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37343272

ABSTRACT

Recent clinical reports have highlighted the need for wild-type (WT) and mutant dual inhibitors of c-MET kinase for the treatment of cancer. We report herein a novel chemical series of ATP competitive type-III inhibitors of WT and D1228V mutant c-MET. Using a combination of structure-based drug design and computational analyses, ligand 2 was optimized to a highly selective chemical series with nanomolar activities in biochemical and cellular settings. Representatives of the series demonstrate excellent pharmacokinetic profiles in rat in vivo studies with promising free-brain exposures, paving the way for the design of brain permeable drugs for the treatment of c-MET driven cancers.


Subject(s)
Antineoplastic Agents , Neoplasms , Rats , Animals , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met , Drug Design , Adenosine Triphosphate , Antineoplastic Agents/pharmacology
13.
ACS Med Chem Lett ; 14(12): 1848-1856, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38116444

ABSTRACT

Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b) is a RING finger E3 ligase that is responsible for repressing T-cell, natural killer (NK) cell, and B-cell activation. The robust antitumor activity observed in Cbl-b deficient mice arising from elevated T-cell and NK-cell activity justified our discovery effort toward Cbl-b inhibitors that might show therapeutic promise in immuno-oncology, where activation of the immune system can drive the recognition and killing of cancer cells. We undertook a high-throughput screening campaign followed by structure-enabled optimization to develop a novel benzodiazepine series of potent Cbl-b inhibitors. This series displayed nanomolar levels of biochemical potency, as well as potent T-cell activation. The functional activity of this class of Cbl-b inhibitors was further corroborated with ubiquitin-based cellular assays.

15.
SLAS Technol ; 27(3): 209-213, 2022 06.
Article in English | MEDLINE | ID: mdl-35058193

ABSTRACT

In laboratories with multiple identical analytical instruments and consistent sample workflows, analysts frequently perform repetitive software control steps, yet automation options in vendor-supplied instrument software are generally limited and may not support the desired laboratory workflow. Scripts that automate tasks to monitor systems, streamline worklist creation, or minimize downtime can save valuable personnel time and reduce errors. AutoHotkey is a free, open-source scripting language for Windows that allows users with no programming experience to easily create scripts automating a wide variety of activities. The scripts automate the tasks that a user performs while interacting with the instrument control software, such as mouse clicks and keyboard entries and closing software windows, rather than modify the underlying instrument software, and thus these scripts are compatible with multiple vendor software packages and Windows OS versions. The scripts can be triggered manually from a desktop icon or automatically through Windows Task Scheduler.


Subject(s)
Laboratories , Software , Automation , Workflow
16.
ACS Chem Biol ; 17(1): 54-67, 2022 01 21.
Article in English | MEDLINE | ID: mdl-34955012

ABSTRACT

Cyclin-dependent-kinases (CDKs) are members of the serine/threonine kinase family and are highly regulated by cyclins, a family of regulatory subunits that bind to CDKs. CDK9 represents one of the most studied examples of these transcriptional CDKs. CDK9 forms a heterodimeric complex with its regulatory subunit cyclins T1, T2 and K to form the positive transcription elongation factor b (P-TEFb). This complex regulates transcription via the phosphorylation of RNA polymerase II (RNAPolII) on Ser-2, facilitating promoter clearance and transcription elongation and thus remains an attractive therapeutic target. Herein, we have utilized classical affinity purification chemical proteomics, kinobeads assay, compressed CEllular Thermal Shift Assay (CETSA)-MS and Limited Proteolysis (LiP) to study the selectivity, target engagement and downstream mechanistic insights of a CDK9 tool compound. The above experiments highlight the value of quantitative mass spectrometry approaches to drug discovery, specifically proteome wide target identification and selectivity profiling. The approaches utilized in this study unanimously indicated that the CDK family of kinases are the main target of the compound of interest, with CDK9, showing the highest target affinity with remarkable consistency across approaches. We aim to provide guidance to the scientific community on the available chemical biology/proteomic tools to study advanced lead molecules and to highlight pros and cons of each technology while describing our findings in the context of the CDKs biology.


Subject(s)
Cyclin-Dependent Kinase 9/antagonists & inhibitors , Proteomics , Cell Line, Tumor , Chemical Fractionation , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mass Spectrometry
17.
Bioorg Med Chem Lett ; 21(10): 2958-61, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21493067

ABSTRACT

Synthesis and biological evaluation of a series of 6-aminopyrazolyl-pyridine-3-carbonitriles as JAK2 kinase inhibitors was reported. Biochemical screening, followed by profile optimization, resulted in JAK2 inhibitors exhibiting good kinase selectivity, pharmacokinetic properties, physical properties and pharmacodynamic effects.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Nitriles/chemical synthesis , Nitriles/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemistry , Pyridines/chemistry , Animals , Cell Line , Cell Proliferation/drug effects , Inhibitory Concentration 50 , Mice , Molecular Structure , Nitriles/chemistry , Nitriles/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Rats , Structure-Activity Relationship
18.
J Med Chem ; 64(20): 15189-15213, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34647738

ABSTRACT

Optimization of a series of azabenzimidazoles identified from screening hit 2 and the information gained from a co-crystal structure of the azabenzimidazole-based lead 6 bound to CDK9 led to the discovery of azaindoles as highly potent and selective CDK9 inhibitors. With the goal of discovering a highly selective and potent CDK9 inhibitor administrated intravenously that would enable transient target engagement of CDK9 for the treatment of hematological malignancies, further optimization focusing on physicochemical and pharmacokinetic properties led to azaindoles 38 and 39. These compounds are highly potent and selective CDK9 inhibitors having short half-lives in rodents, suitable physical properties for intravenous administration, and the potential to achieve profound but transient inhibition of CDK9 in vivo.


Subject(s)
Cyclin-Dependent Kinase 9/antagonists & inhibitors , Drug Discovery , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Cyclin-Dependent Kinase 9/metabolism , Dose-Response Relationship, Drug , Humans , Indoles/chemical synthesis , Indoles/chemistry , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
20.
J Med Chem ; 63(24): 15564-15590, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33306391

ABSTRACT

A CDK9 inhibitor having short target engagement would enable a reduction of Mcl-1 activity, resulting in apoptosis in cancer cells dependent on Mcl-1 for survival. We report the optimization of a series of amidopyridines (from compound 2), focusing on properties suitable for achieving short target engagement after intravenous administration. By increasing potency and human metabolic clearance, we identified compound 24, a potent and selective CDK9 inhibitor with suitable predicted human pharmacokinetic properties to deliver transient inhibition of CDK9. Furthermore, the solubility of 24 was considered adequate to allow i.v. formulation at the anticipated effective dose. Short-term treatment with compound 24 led to a rapid dose- and time-dependent decrease of pSer2-RNAP2 and Mcl-1, resulting in cell apoptosis in multiple hematological cancer cell lines. Intermittent dosing of compound 24 demonstrated efficacy in xenograft models derived from multiple hematological tumors. Compound 24 is currently in clinical trials for the treatment of hematological malignancies.


Subject(s)
Cyclin-Dependent Kinase 9/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Pyridines/chemistry , Animals , Apoptosis/drug effects , Binding Sites , Cell Line, Tumor , Cyclin-Dependent Kinase 9/metabolism , Dogs , Drug Evaluation, Preclinical , Half-Life , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Humans , Mice , Molecular Docking Simulation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridines/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Rats , Solubility , Structure-Activity Relationship , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL