Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Med Chem ; 65(21): 14366-14390, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36261130

ABSTRACT

The branched-chain amino acid transaminases (BCATs) are enzymes that catalyze the first reaction of catabolism of the essential branched-chain amino acids to branched-chain keto acids to form glutamate. They are known to play a key role in different cancer types. Here, we report a new structural class of BCAT1/2 inhibitors, (trifluoromethyl)pyrimidinediones, identified by a high-throughput screening campaign and subsequent optimization guided by a series of X-ray crystal structures. Our potent dual BCAT1/2 inhibitor BAY-069 displays high cellular activity and very good selectivity. Along with a negative control (BAY-771), BAY-069 was donated as a chemical probe to the Structural Genomics Consortium.


Subject(s)
Amino Acids, Branched-Chain , Transaminases , Transaminases/metabolism , Amino Acids, Branched-Chain/metabolism , Keto Acids/metabolism
2.
Bioorg Med Chem Lett ; 30(16): 127298, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32631518

ABSTRACT

We report on the discovery of the new clinical candidate BAY 1003803 as glucocorticoid receptor agonist for the topical treatment of psoriasis or severe atopic dermatitis. In the course of optimizing the amino alcohol series as a highly potent new non-steroidal lead structure, considerations were made as to how physicochemical properties and safety concerns relate to structural motifs. BAY 1003803 demonstrates strong anti-inflammatory activity in vitro paired with a pharmacokinetic profile suitable for topical application.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Discovery , Receptors, Glucocorticoid/agonists , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Dogs , Dose-Response Relationship, Drug , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Male , Molecular Structure , Rats , Receptors, Glucocorticoid/metabolism , Structure-Activity Relationship
3.
Nat Chem Biol ; 16(5): 497-506, 2020 05.
Article in English | MEDLINE | ID: mdl-32231343

ABSTRACT

We recently described glutathione peroxidase 4 (GPX4) as a promising target for killing therapy-resistant cancer cells via ferroptosis. The onset of therapy resistance by multiple types of treatment results in a stable cell state marked by high levels of polyunsaturated lipids and an acquired dependency on GPX4. Unfortunately, all existing inhibitors of GPX4 act covalently via a reactive alkyl chloride moiety that confers poor selectivity and pharmacokinetic properties. Here, we report our discovery that masked nitrile-oxide electrophiles, which have not been explored previously as covalent cellular probes, undergo remarkable chemical transformations in cells and provide an effective strategy for selective targeting of GPX4. The new GPX4-inhibiting compounds we describe exhibit unexpected proteome-wide selectivity and, in some instances, vastly improved physiochemical and pharmacokinetic properties compared to existing chloroacetamide-based GPX4 inhibitors. These features make them superior tool compounds for biological interrogation of ferroptosis and constitute starting points for development of improved inhibitors of GPX4.


Subject(s)
Enzyme Inhibitors/pharmacology , Nitriles/chemistry , Nitriles/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Animals , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Ferroptosis/drug effects , Humans , Lipid Peroxidation/drug effects , Mice, SCID , Molecular Probes/chemistry , Molecular Targeted Therapy , Oxides/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/chemistry , Prodrugs/chemistry , Rats, Wistar , Selenocysteine/chemistry , Selenocysteine/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
4.
J Med Chem ; 63(15): 8025-8042, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32338514

ABSTRACT

Inhibition of monopolar spindle 1 (MPS1) kinase represents a novel approach to cancer treatment: instead of arresting the cell cycle in tumor cells, cells are driven into mitosis irrespective of DNA damage and unattached/misattached chromosomes, resulting in aneuploidy and cell death. Starting points for our optimization efforts with the goal to identify MPS1 inhibitors were two HTS hits from the distinct chemical series "triazolopyridines" and "imidazopyrazines". The major initial issue of the triazolopyridine series was the moderate potency of the HTS hits. The imidazopyrazine series displayed more than 10-fold higher potencies; however, in the early project phase, this series suffered from poor metabolic stability. Here, we outline the evolution of the two hit series to clinical candidates BAY 1161909 and BAY 1217389 and reveal how both clinical candidates bind to the ATP site of MPS1 kinase, while addressing different pockets utilizing different binding interactions, along with their synthesis and preclinical characterization in selected in vivo efficacy models.


Subject(s)
Antineoplastic Agents/metabolism , Cell Cycle Proteins/metabolism , Drug Delivery Systems/methods , Drug Discovery/methods , M Phase Cell Cycle Checkpoints/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Spindle Apparatus/drug effects , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Dogs , Female , HT29 Cells , HeLa Cells , Humans , M Phase Cell Cycle Checkpoints/physiology , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Structure, Tertiary , Protein-Tyrosine Kinases/antagonists & inhibitors , Rats , Rats, Wistar , Spindle Apparatus/metabolism , Treatment Outcome
5.
Mol Cancer Ther ; 17(11): 2285-2296, 2018 11.
Article in English | MEDLINE | ID: mdl-30115664

ABSTRACT

The lactate transporter SLC16A1/monocarboxylate transporter 1 (MCT1) plays a central role in tumor cell energy homeostasis. In a cell-based screen, we identified a novel class of MCT1 inhibitors, including BAY-8002, which potently suppress bidirectional lactate transport. We investigated the antiproliferative activity of BAY-8002 in a panel of 246 cancer cell lines and show that hematopoietic tumor cells, in particular diffuse large B-cell lymphoma cell lines, and subsets of solid tumor models are particularly sensitive to MCT1 inhibition. Associated markers of sensitivity were, among others, lack of MCT4 expression, low pleckstrin homology like domain family A member 2, and high pellino E3 ubiquitin protein ligase 1 expression. The antitumor effect of MCT1 inhibition was less pronounced on tumor xenografts, with tumor stasis being the maximal response. BAY-8002 significantly increased intratumor lactate levels and transiently modulated pyruvate levels. In order to address potential acquired resistance mechanisms to MCT1 inhibition, we generated MCT1 inhibitor-resistant cell lines and show that resistance can occur by upregulation of MCT4 even in the presence of sufficient oxygen, as well as by shifting energy generation toward oxidative phosphorylation. These findings provide insight into novel aspects of tumor response to MCT1 modulation and offer further rationale for patient selection in the clinical development of MCT1 inhibitors. Mol Cancer Ther; 17(11); 2285-96. ©2018 AACR.


Subject(s)
Aminobenzoates/pharmacology , Benzoates/pharmacology , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , Monocarboxylic Acid Transporters/antagonists & inhibitors , Sulfones/pharmacology , Symporters/antagonists & inhibitors , Aminobenzoates/chemistry , Animals , Benzoates/chemistry , Biological Transport/drug effects , Carbon Radioisotopes , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Fluorescence , Humans , Hydrogen-Ion Concentration , Lactic Acid/metabolism , Mice, SCID , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Pyrimidinones/pharmacology , Pyruvic Acid/metabolism , Sulfones/chemistry , Symporters/metabolism , Thiophenes/pharmacology , Treatment Outcome , Xenopus laevis
6.
ACS Chem Biol ; 12(8): 1986-1992, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28679043

ABSTRACT

MTH1 is a hydrolase responsible for sanitization of oxidized purine nucleoside triphosphates to prevent their incorporation into replicating DNA. Early tool compounds published in the literature inhibited the enzymatic activity of MTH1 and subsequently induced cancer cell death; however recent studies have questioned the reported link between these two events. Therefore, it is important to validate MTH1 as a cancer dependency with high quality chemical probes. Here, we present BAY-707, a substrate-competitive, highly potent and selective inhibitor of MTH1, chemically distinct compared to those previously published. Despite superior cellular target engagement and pharmacokinetic properties, inhibition of MTH1 with BAY-707 resulted in a clear lack of in vitro or in vivo anticancer efficacy either in mono- or in combination therapies. Therefore, we conclude that MTH1 is dispensable for cancer cell survival.


Subject(s)
DNA Repair Enzymes/metabolism , Drug Delivery Systems , Morpholines/pharmacology , Neoplasms/drug therapy , Neoplasms/enzymology , Phosphoric Monoester Hydrolases/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Caco-2 Cells , Cells, Cultured , DNA Repair Enzymes/antagonists & inhibitors , Enzyme Activation/drug effects , HeLa Cells , Hepatocytes/drug effects , Humans , MCF-7 Cells , Mice , Mice, Nude , Microsomes, Liver/drug effects , Models, Molecular , Morpholines/chemistry , Neoplasms/physiopathology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats
7.
Cell Death Dis ; 8(3): e2709, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28358364

ABSTRACT

Owing to lagging or insufficient neo-angiogenesis, hypoxia is a feature of most solid tumors. Hypoxic tumor regions contribute to resistance against antiproliferative chemotherapeutics, radiotherapy and immunotherapy. Targeting cells in hypoxic tumor areas is therefore an important strategy for cancer treatment. Most approaches for targeting hypoxic cells focus on the inhibition of hypoxia adaption pathways but only a limited number of compounds with the potential to specifically target hypoxic tumor regions have been identified. By using tumor spheroids in hypoxic conditions as screening system, we identified a set of compounds, including the phenothiazine antipsychotic Fluphenazine, as hits with novel mode of action. Fluphenazine functionally inhibits acid sphingomyelinase and causes cellular sphingomyelin accumulation, which induces cancer cell death specifically in hypoxic tumor spheroids. Moreover, we found that functional inhibition of acid sphingomyelinase leads to overactivation of hypoxia stress-response pathways and that hypoxia-specific cell death is mediated by the stress-responsive transcription factor ATF4. Taken together, the here presented data suggest a novel, yet unexplored mechanism in which induction of sphingolipid stress leads to the overactivation of hypoxia stress-response pathways and thereby promotes their pro-apoptotic tumor-suppressor functions to specifically kill cells in hypoxic tumor areas.


Subject(s)
Colonic Neoplasms/enzymology , Fluphenazine/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Cell Death/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Sphingomyelin Phosphodiesterase/genetics
8.
Acta Neuropathol ; 133(4): 629-644, 2017 04.
Article in English | MEDLINE | ID: mdl-28124097

ABSTRACT

Mutations in codon 132 of isocitrate dehydrogenase (IDH) 1 are frequent in diffuse glioma, acute myeloid leukemia, chondrosarcoma and intrahepatic cholangiocarcinoma. These mutations result in a neomorphic enzyme specificity which leads to a dramatic increase of intracellular D-2-hydroxyglutarate (2-HG) in tumor cells. Therefore, mutant IDH1 protein is a highly attractive target for inhibitory drugs. Here, we describe the development and properties of BAY 1436032, a pan-inhibitor of IDH1 protein with different codon 132 mutations. BAY 1436032 strongly reduces 2-HG levels in cells carrying IDH1-R132H, -R132C, -R132G, -R132S and -R132L mutations. Cells not carrying IDH mutations were unaffected. BAY 1436032 did not exhibit toxicity in vitro or in vivo. The pharmacokinetic properties of BAY 1436032 allow for oral administration. In two independent experiments, BAY 1436032 has been shown to significantly prolong survival of mice intracerebrally transplanted with human astrocytoma carrying the IDH1R132H mutation. In conclusion, we developed a pan-inhibitor targeting tumors with different IDH1R132 mutations.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Astrocytoma/drug therapy , Benzimidazoles/pharmacology , Brain Neoplasms/drug therapy , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Aniline Compounds/chemistry , Aniline Compounds/pharmacokinetics , Aniline Compounds/toxicity , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Astrocytoma/enzymology , Astrocytoma/genetics , Benzimidazoles/chemistry , Benzimidazoles/pharmacokinetics , Benzimidazoles/toxicity , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/toxicity , Escherichia coli , Female , Glutarates/metabolism , HEK293 Cells , Humans , Isocitrate Dehydrogenase/metabolism , Mice, Inbred BALB C , Mice, Nude , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sarcoma/drug therapy , Sarcoma/enzymology , Sarcoma/genetics , Sf9 Cells , Xenograft Model Antitumor Assays
9.
Int J Cancer ; 140(2): 449-459, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27699769

ABSTRACT

The PI3K-AKT-mTOR signaling cascade is activated in the majority of human cancers, and its activation also plays a key role in resistance to chemo and targeted therapeutics. In particular, in both breast and prostate cancer, increased AKT pathway activity is associated with cancer progression, treatment resistance and poor disease outcome. Here, we evaluated the activity of a novel allosteric AKT1/2 inhibitor, BAY 1125976, in biochemical, cellular mechanistic, functional and in vivo efficacy studies in a variety of tumor models. In in vitro kinase activity assays, BAY 1125976 potently and selectively inhibited the activity of full-length AKT1 and AKT2 by binding into an allosteric binding pocket formed by kinase and PH domain. In accordance with this proposed allosteric binding mode, BAY 1125976 bound to inactive AKT1 and inhibited T308 phosphorylation by PDK1, while the activity of truncated AKT proteins lacking the pleckstrin homology domain was not inhibited. In vitro, BAY 1125976 inhibited cell proliferation in a broad panel of human cancer cell lines. Particularly high activity was observed in breast and prostate cancer cell lines expressing estrogen or androgen receptors. Furthermore, BAY 1125976 exhibited strong in vivo efficacy in both cell line and patient-derived xenograft models such as the KPL4 breast cancer model (PIK3CAH1074R mutant), the MCF7 and HBCx-2 breast cancer models and the AKTE17K mutant driven prostate cancer (LAPC-4) and anal cancer (AXF 984) models. These findings indicate that BAY 1125976 is a potent and highly selective allosteric AKT1/2 inhibitor that targets tumors displaying PI3K/AKT/mTOR pathway activation, providing opportunities for the clinical development of new, effective treatments.


Subject(s)
Nitriles/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , Sulfones/pharmacology , Animals , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Female , HeLa Cells , Humans , MCF-7 Cells , Male , Mice , Mice, Nude , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays/methods
10.
ChemMedChem ; 11(20): 2261-2271, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27552707

ABSTRACT

Despite the long-known fact that the facilitative glucose transporter GLUT1 is one of the key players safeguarding the increase in glucose consumption of many tumor entities even under conditions of normal oxygen supply (known as the Warburg effect), only few endeavors have been undertaken to find a GLUT1-selective small-molecule inhibitor. Because other transporters of the GLUT1 family are involved in crucial processes, these transporters should not be addressed by such an inhibitor. A high-throughput screen against a library of ∼3 million compounds was performed to find a small molecule with this challenging potency and selectivity profile. The N-(1H-pyrazol-4-yl)quinoline-4-carboxamides were identified as an excellent starting point for further compound optimization. After extensive structure-activity relationship explorations, single-digit nanomolar inhibitors with a selectivity factor of >100 against GLUT2, GLUT3, and GLUT4 were obtained. The most promising compound, BAY-876 [N4 -[1-(4-cyanobenzyl)-5-methyl-3-(trifluoromethyl)-1H-pyrazol-4-yl]-7-fluoroquinoline-2,4-dicarboxamide], showed good metabolic stability in vitro and high oral bioavailability in vivo.


Subject(s)
Glucose Transporter Type 1/antagonists & inhibitors , Pyrazoles/pharmacology , Quinolines/pharmacology , Administration, Oral , Biological Availability , Glucose Transporter Type 1/metabolism , High-Throughput Screening Assays , Humans , Molecular Structure , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Quinolines/administration & dosage , Quinolines/chemistry , Structure-Activity Relationship
12.
Bioorg Med Chem Lett ; 26(7): 1732-7, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26949183

ABSTRACT

The compound class of 1H-pyrazolo[3,4-d]pyrimidines was identified using HTS as very potent inhibitors of facilitated glucose transporter 1 (GLUT1). Extensive structure-activity relationship studies (SAR) of each ring system of the molecular framework was established revealing essential structural motives (i.e., ortho-methoxy substituted benzene, piperazine and pyrimidine). The selectivity against GLUT2 was excellent and initial in vitro and in vivo pharmacokinetic (PK) studies are encouraging.


Subject(s)
Glucose Transporter Type 1/antagonists & inhibitors , Pyrimidines/chemistry , Pyrimidines/pharmacology , Animals , Cell Line , Drug Discovery , Glucose Transporter Type 1/metabolism , Humans , Male , Pyrimidines/pharmacokinetics , Rats, Wistar , Structure-Activity Relationship
13.
Mol Cancer Ther ; 15(4): 583-92, 2016 04.
Article in English | MEDLINE | ID: mdl-26832791

ABSTRACT

Monopolar spindle 1 (Mps1) has been shown to function as the key kinase that activates the spindle assembly checkpoint (SAC) to secure proper distribution of chromosomes to daughter cells. Here, we report the structure and functional characterization of two novel selective Mps1 inhibitors, BAY 1161909 and BAY 1217389, derived from structurally distinct chemical classes. BAY 1161909 and BAY 1217389 inhibited Mps1 kinase activity with IC50 values below 10 nmol/L while showing an excellent selectivity profile. In cellular mechanistic assays, both Mps1 inhibitors abrogated nocodazole-induced SAC activity and induced premature exit from mitosis ("mitotic breakthrough"), resulting in multinuclearity and tumor cell death. Both compounds efficiently inhibited tumor cell proliferation in vitro (IC50 nmol/L range). In vivo, BAY 1161909 and BAY 1217389 achieved moderate efficacy in monotherapy in tumor xenograft studies. However, in line with its unique mode of action, when combined with paclitaxel, low doses of Mps1 inhibitor reduced paclitaxel-induced mitotic arrest by the weakening of SAC activity. As a result, combination therapy strongly improved efficacy over paclitaxel or Mps1 inhibitor monotreatment at the respective MTDs in a broad range of xenograft models, including those showing acquired or intrinsic paclitaxel resistance. Both Mps1 inhibitors showed good tolerability without adding toxicity to paclitaxel monotherapy. These preclinical findings validate the innovative concept of SAC abrogation for cancer therapy and justify clinical proof-of-concept studies evaluating the Mps1 inhibitors BAY 1161909 and BAY 1217389 in combination with antimitotic cancer drugs to enhance their efficacy and potentially overcome resistance. Mol Cancer Ther; 15(4); 583-92. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Discovery , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Female , Humans , Male , Mice , Mitosis/drug effects , Protein Kinase Inhibitors/chemistry , Rats , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
Oncotarget ; 7(5): 6015-28, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26760770

ABSTRACT

Androgen receptor (AR) mutations arise in patients developing resistance to hormone deprivation therapies. Here we describe BAY 1024767, a thiohydantoin derivative with strong antagonistic activity against nine AR variants with mutations located in the AR ligand-binding domain (LBD), and against wild-type AR. Antagonism was maintained, though reduced, at increased androgen levels. Anti-tumor efficacy was evidenced in vivo in the KuCaP-1 prostate cancer model which bears the W741C bicalutamide resistance mutation and in the syngeneic prostate cancer rat model Dunning R3327-G. The prevalence of six selected AR mutations was determined in plasma DNA originating from 100 resistant patients and found to be at least 12%. Altogether the results show BAY 1024767 to be a strong antagonist for several AR mutants linked to therapy resistance, which opens the door for next-generation compounds that can benefit patients based on their mutation profile.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/genetics , Thiohydantoins/pharmacology , Animals , COS Cells , Caco-2 Cells , Cell Line, Tumor , Chlorocebus aethiops , Down-Regulation , Humans , Male , Mice , Mice, SCID , Mutation , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Random Allocation , Rats , Receptors, Androgen/metabolism , Transfection , Xenograft Model Antitumor Assays
15.
Bioorg Med Chem Lett ; 26(1): 186-93, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26611920

ABSTRACT

Recently, we had identified an unexplored pocket adjacent to the known binding site of allosteric MEK inhibitors which allowed us to design highly potent and in vivo efficacious novel inhibitors. We now report that our initial preclinical candidate, featuring a phenoxy side chain with a sulfamide capping group, displayed human carbonic anhydrase off-target activity and species-dependent blood cell accumulation, which prevented us from advancing this candidate further. Since this sulfamide MEK inhibitor displayed an exceptionally favorable PK profile with low brain penetration potential despite being highly oral bioavailable, we elected to keep the sulfamide capping group intact while taming its unwanted off-target activity by optimizing the structural surroundings. Introduction of a neighboring fluorine atom or installation of a methylene linker reduced hCA potency sufficiently, at the cost of MEK target potency. Switching to a higher fluorinated central core reinstated high MEK potency, leading to two new preclinical candidates with long half-lives, high bioavailabilities, low brain penetration potential and convincing efficacy in a K-Ras-mutated A549 xenograft model.


Subject(s)
Antineoplastic Agents/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Sulfonamides/pharmacology , Allosteric Regulation/drug effects , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Biological Availability , Brain/drug effects , Brain/metabolism , Carbonic Anhydrases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Half-Life , Humans , Mice , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry , Sulfonamides/pharmacokinetics , Xenograft Model Antitumor Assays
16.
ChemMedChem ; 10(12): 2004-13, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26541480

ABSTRACT

Having recently identified a so-far unexplored area adjacent to the known binding site of allosteric mitogen-activated protein kinase kinase (MEK) inhibitors, we now report an extension of these studies by combining our new side chains with different MEK inhibitor cores in a modular manner. Replacement of the amide headgroup with inverse sulfonamides resulted in the identification of new MEK inhibitors with at least 10-fold higher cellular potency against K-Ras-mutated tumor cells. A selected inhibitor from this new series retained the favorable pharmacokinetic profile of its predecessor in rodent and non-rodent species and displayed significant in vivo efficacy at once-daily oral doses of 0.25-1 mg kg(-1) in a K-Ras-mutated xenograft model. The brain penetration potential of this analogue was significantly attenuated relative to PD325901. In a second series, the central fluorophenyl core was replaced by a pyridine moiety which gave rise to a similar boost in cellular potency. Most notably, analogues from this second series do not show MEK feedback phosphorylation in K-Ras-mutated A549 cells. Our results complement recent reports on the structural intricacies of MEK-Raf feedback interactions.


Subject(s)
MAP Kinase Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Allosteric Regulation , Animals , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Brain/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Diphenylamine/analogs & derivatives , Diphenylamine/chemistry , Diphenylamine/metabolism , Diphenylamine/pharmacology , Female , Half-Life , Humans , MAP Kinase Kinase 1/metabolism , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Rats , Signal Transduction/drug effects , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Transplantation, Heterologous
17.
Neoplasia ; 15(10): 1161-71, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24204195

ABSTRACT

OBJECTIVE: The objectives of the study were to evaluate the allosteric mitogen-activated protein kinase kinase (MEK) inhibitor BAY 86-9766 in monotherapy and in combination with sorafenib in orthotopic and subcutaneous hepatocellular carcinoma (HCC) models with different underlying etiologies in two species. DESIGN: Antiproliferative potential of BAY 86-9766 and synergistic effects with sorafenib were studied in several HCC cell lines. Relevant pathway signaling was studied in MH3924a cells. For in vivo testing, the HCC cells were implanted subcutaneously or orthotopically. Survival and mode of action (MoA) were analyzed. RESULTS: BAY 86-9766 exhibited potent antiproliferative activity in HCC cell lines with half-maximal inhibitory concentration values ranging from 33 to 762 nM. BAY 86-9766 was strongly synergistic with sorafenib in suppressing tumor cell proliferation and inhibiting phosphorylation of the extracellular signal-regulated kinase (ERK). BAY 86-9766 prolonged survival in Hep3B xenografts, murine Hepa129 allografts, and MH3924A rat allografts. Additionally, tumor growth, ascites formation, and serum alpha-fetoprotein levels were reduced. Synergistic effects in combination with sorafenib were shown in Huh-7, Hep3B xenografts, and MH3924A allografts. On the signaling pathway level, the combination of BAY 86-9766 and sorafenib led to inhibition of the upregulatory feedback loop toward MEK phosphorylation observed after BAY 86-9766 monotreatment. With regard to the underlying MoA, inhibition of ERK phosphorylation, tumor cell proliferation, and microvessel density was observed in vivo. CONCLUSION: BAY 86-9766 shows potent single-agent antitumor activity and acts synergistically in combination with sorafenib in preclinical HCC models. These results support the ongoing clinical development of BAY 86-9766 and sorafenib in advanced HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Diphenylamine/analogs & derivatives , Liver Neoplasms/drug therapy , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Sulfonamides/pharmacology , Allografts , Allosteric Regulation , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drug Synergism , Female , Heterografts , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Rats , Sorafenib , Sulfonamides/therapeutic use
18.
Bioorg Med Chem Lett ; 23(8): 2384-90, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23474388

ABSTRACT

Using PD325901 as a starting point for identifying novel allosteric MEK inhibitors with high cell potency and long-lasting target inhibition in vivo, truncation of its hydroxamic ester headgroup was combined with incorporation of alkyl and aryl ethers at the neighboring ring position. Whereas alkoxy side chains did not yield sufficient levels of cell potency, specifically substituted aryloxy groups allowed for high enzymatic and cellular potencies. Sulfamide 28 was identified as a highly potent MEK inhibitor with nanomolar cell potency against B-RAF (V600E) as well as Ras-mutated cell lines, high metabolic stability and resulting long half-lives. It was efficacious against B-RAF as well as K-Ras driven xenograft models and showed-despite being orally bioavailable and not a P-glycoprotein substrate-much lower brain/plasma exposure ratios than PD325901.


Subject(s)
MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/chemistry , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Allosteric Regulation , Animals , Benzamides/chemistry , Benzamides/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/chemistry , Diphenylamine/pharmacology , Drug Design , Mice , Structure-Activity Relationship , Xenograft Model Antitumor Assays
19.
J Biomol Screen ; 14(8): 913-23, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19675314

ABSTRACT

The potential of enzyme inhibition of a drug is frequently quantified in terms of IC(50) values. Although this is a suitable quantity for reversible inhibitors, concerns arise when dealing with irreversible or mechanism-based inhibitors (MBIs). IC(50) values of MBIs are time dependent, causing serious problems when aiming at ranking different compounds with respect to their inhibitory potential. As a consequence, most studies and ranking schemes related to MBIs rely on the inhibition constant (K(I)) and the rate of enzyme inactivation (k(inact)) rather than on IC(50) values. In this article, the authors derive a novel relation between potentially time-dependent IC(50) values and K(I), k(inact) parameters for different types of inhibition. This allows for direct estimation of K(I) and k(inact) values from time-dependent IC(50) values, even without the need of additional preincubation experiments. The application of this approach is illustrated using a fluorimetric assay to access the drug-drug interaction potential associated with new chemical entities. The approach can easily be implemented using standard software tools (e.g., XLfit) and may also be suitable for applications where mechanism-based inhibition is a desired mode of action (e.g., at particular pharmacological drug targets).


Subject(s)
Drug Antagonism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacokinetics , Inhibitory Concentration 50 , Binding, Competitive , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1A2 Inhibitors , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors , Humans , Models, Chemical , Models, Theoretical , Time Factors
20.
J Med Chem ; 48(14): 4618-27, 2005 Jul 14.
Article in English | MEDLINE | ID: mdl-15999999

ABSTRACT

Under pathophysiological conditions, alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate (AMPA) receptor activation is considered to play a key role in several disorders of the central nervous system. In the search for AMPA receptor antagonists, the synthesis and pharmacological characterization of a series of novel compounds that are structurally related to GYKI 52466 (1), a well-known selective noncompetitive AMPA receptor antagonist, was performed. In vitro, 2,3-dimethyl-6-phenyl-12H-[1,3]dioxolo[4,5-h]imidazo[1,2-c][2,3]benzodiazepine (ZK 187638, 14a) antagonized the kainate-induced currents in cultured hippocampal neurons with an IC(50) of 3.4 microM in a noncompetitive fashion. When tested in a clinically predictive rat model of acute ischemic stroke, this noncompetitive AMPA receptor antagonist significantly reduced brain infarction, indicating that it is neuroprotective after permanent focal cerebral ischemia.


Subject(s)
Benzodiazepines/chemical synthesis , Dioxoles/chemical synthesis , Nerve Degeneration/prevention & control , Neuroprotective Agents/chemical synthesis , Receptors, AMPA/antagonists & inhibitors , Acute Disease , Animals , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Binding, Competitive , Brain Infarction/etiology , Brain Infarction/pathology , Brain Infarction/prevention & control , Cells, Cultured , Dioxoles/chemistry , Dioxoles/pharmacology , Hippocampus/cytology , In Vitro Techniques , Ischemic Attack, Transient/complications , Kainic Acid/pharmacology , Male , Mice , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacology , Patch-Clamp Techniques , Prosencephalon/metabolism , Radioligand Assay , Rats , Rats, Inbred F344 , Receptors, AMPA/agonists , Receptors, AMPA/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...