Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
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.
Bioconjug Chem ; 33(6): 1210-1221, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35658441

ABSTRACT

Inhibition of intracellular nicotinamide phosphoribosyltransferase (NAMPT) represents a new mode of action for cancer-targeting antibody-drug conjugates (ADCs) with activity also in slowly proliferating cells. To extend the repertoire of available effector chemistries, we have developed a novel structural class of NAMPT inhibitors as ADC payloads. A structure-activity relationship-driven approach supported by protein structural information was pursued to identify a suitable attachment point for the linker to connect the NAMPT inhibitor with the antibody. Optimization of scaffolds and linker structures led to highly potent effector chemistries which were conjugated to antibodies targeting C4.4a (LYPD3), HER2 (c-erbB2), or B7H3 (CD276) and tested on antigen-positive and -negative cancer cell lines. Pharmacokinetic studies, including metabolite profiling, were performed to optimize the stability and selectivity of the ADCs and to evaluate potential bystander effects. Optimized NAMPTi-ADCs demonstrated potent in vivo antitumor efficacy in target antigen-expressing xenograft mouse models. This led to the development of highly potent NAMPT inhibitor ADCs with a very good selectivity profile compared with the corresponding isotype control ADCs. Moreover, we demonstrate─to our knowledge for the first time─the generation of NAMPTi payload metabolites from the NAMPTi-ADCs in vitro and in vivo. In conclusion, NAMPTi-ADCs represent an attractive new payload class designed for use in ADCs for the treatment of solid and hematological cancers.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Nicotinamide Phosphoribosyltransferase , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B7 Antigens , Cell Line, Tumor , Humans , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Mice , Neoplasms/drug therapy , Neoplasms/enzymology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/chemistry , Structure-Activity Relationship , Xenograft Model Antitumor Assays
3.
ACS Med Chem Lett ; 13(3): 348-357, 2022 Mar 10.
Article in English | MEDLINE | ID: mdl-35300083

ABSTRACT

Mitochondria are key regulators of energy supply and cell death. Generation of ATP within mitochondria occurs through oxidative phosphorylation (OXPHOS), a process which utilizes the four complexes (complex I-IV) of the electron transport chain and ATP synthase. Certain oncogenic mutations (e.g., LKB1 or mIDH) can further enhance the reliance of cancer cells on OXPHOS for their energetic requirements, rendering cells sensitive to complex I inhibition and highlighting the potential value of complex I as a therapeutic target. Herein, we describe the discovery of a potent, selective, and species cross-reactive complex I inhibitor. A high-throughput screen of the Bayer compound library followed by hit triaging and initial hit-to-lead activities led to a lead structure which was further optimized in a comprehensive lead optimization campaign. Focusing on balancing potency and metabolic stability, this program resulted in the identification of BAY-179, an excellent in vivo suitable tool with which to probe the biological relevance of complex I inhibition in cancer indications.

4.
Acta Crystallogr D Struct Biol ; 77(Pt 2): 237-248, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33559612

ABSTRACT

Wild-type human glutathione peroxidase 4 (GPX4) was co-expressed with SBP2 (selenocysteine insertion sequence-binding protein 2) in human HEK cells to achieve efficient production of this selenocysteine-containing enzyme on a preparative scale for structural biology. The protein was purified and crystallized, and the crystal structure of the wild-type form of GPX4 was determined at 1.0 Šresolution. The overall fold and the active site are conserved compared with previously determined crystal structures of mutated forms of GPX4. A mass-spectrometry-based approach was developed to monitor the reaction of the active-site selenocysteine Sec46 with covalent inhibitors. This, together with the introduction of a surface mutant (Cys66Ser), enabled the crystal structure determination of GPX4 in complex with the covalent inhibitor ML162 [(S)-enantiomer]. The mass-spectrometry-based approach described here opens the path to further co-complex crystal structures of this potential cancer drug target in complex with covalent inhibitors.


Subject(s)
Enzyme Inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Protein Binding , Protein Conformation
5.
J Med Chem ; 63(21): 12574-12594, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33108181

ABSTRACT

Despite extensive research on small molecule thrombin inhibitors for oral application in the past decades, only a single double prodrug with very modest oral bioavailability has reached human therapy as a marketed drug. We have undertaken major efforts to identify neutral, non-prodrug inhibitors. Using a holistic analysis of all available internal data, we were able to build computational models and apply these for the selection of a lead series with the highest possibility of achieving oral bioavailability. In our design, we relied on protein structure knowledge to address potency and identified a small window of favorable physicochemical properties to balance absorption and metabolic stability. Protein structure information on the pregnane X receptor helped in overcoming a persistent cytochrome P450 3A4 induction problem. The selected compound series was optimized to a highly potent, neutral, non-prodrug thrombin inhibitor by designing, synthesizing, and testing derivatives. The resulting optimized compound, BAY1217224, has reached first clinical trials, which have confirmed the desired pharmacokinetic properties.


Subject(s)
Anticoagulants/chemical synthesis , Drug Design , Thrombin/antagonists & inhibitors , Administration, Oral , Animals , Anticoagulants/chemistry , Anticoagulants/pharmacokinetics , Anticoagulants/pharmacology , Benzoxazoles/chemistry , Benzoxazoles/metabolism , Benzoxazoles/pharmacology , Binding Sites , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Half-Life , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Imidazoles/pharmacology , Inhibitory Concentration 50 , Male , Molecular Docking Simulation , Oxazolidinones/chemistry , Oxazolidinones/metabolism , Oxazolidinones/pharmacology , Pregnane X Receptor/genetics , Pregnane X Receptor/metabolism , Rats , Rats, Wistar , Structure-Activity Relationship , Thrombin/metabolism , Transcriptional Activation/drug effects
6.
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
7.
ChemMedChem ; 15(10): 827-832, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32237114

ABSTRACT

Due to its frequent mutations in multiple lethal cancers, KRAS is one of the most-studied anticancer targets nowadays. Since the discovery of the druggable allosteric binding site containing a G12C mutation, KRASG12C has been the focus of attention in oncology research. We report here a computationally driven approach aimed at identifying novel and selective KRASG12C covalent inhibitors. The workflow involved initial enumeration of virtual molecules tailored for the KRAS allosteric binding site. Tools such as pharmacophore modeling, docking, and free-energy perturbations were deployed to prioritize the compounds with the best profiles. The synthesized naphthyridinone scaffold showed the ability to react with G12C and inhibit KRASG12C . Analogues were prepared to establish structure-activity relationships, while molecular dynamics simulations and crystallization of the inhibitor-KRASG12C complex highlighted an unprecedented binding mode.


Subject(s)
Enzyme Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Allosteric Regulation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Structure-Activity Relationship
8.
J Med Chem ; 62(24): 11194-11217, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31746599

ABSTRACT

The P2X4 receptor is a ligand-gated ion channel that is expressed on a variety of cell types, especially those involved in inflammatory and immune processes. High-throughput screening led to a new class of P2X4 inhibitors with substantial CYP 3A4 induction in human hepatocytes. A structure-guided optimization with respect to decreased pregnane X receptor (PXR) binding was started. It was found that the introduction of larger and more polar substituents on the ether linker led to less PXR binding while maintaining the P2X4 inhibitory potency. This translated into significantly reduced CYP 3A4 induction for compounds 71 and 73. Unfortunately, the in vivo pharmacokinetic (PK) profiles of these compounds were insufficient for the desired profile in humans. However, BAY-1797 (10) was identified and characterized as a potent and selective P2X4 antagonist. This compound is suitable for in vivo studies in rodents, and the anti-inflammatory and anti-nociceptive effects of BAY-1797 were demonstrated in a mouse complete Freund's adjuvant (CFA) inflammatory pain model.


Subject(s)
Acetamides/pharmacology , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A/metabolism , Drug Discovery , Inflammation/drug therapy , Pain/drug therapy , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Purinergic P2X4/chemistry , Acetamides/chemistry , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Cytochrome P-450 CYP3A Inducers/chemistry , Enzyme Induction , Female , Gene Expression Regulation , Humans , Inflammation/metabolism , Inflammation/pathology , Ligands , Male , Mice , Mice, Inbred C57BL , Pain/metabolism , Pain/pathology , Purinergic P2X Receptor Antagonists/chemistry , Rats , Rats, Wistar
9.
Proc Natl Acad Sci U S A ; 116(7): 2551-2560, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30683722

ABSTRACT

Since the late 1980s, mutations in the RAS genes have been recognized as major oncogenes with a high occurrence rate in human cancers. Such mutations reduce the ability of the small GTPase RAS to hydrolyze GTP, keeping this molecular switch in a constitutively active GTP-bound form that drives, unchecked, oncogenic downstream signaling. One strategy to reduce the levels of active RAS is to target guanine nucleotide exchange factors, which allow RAS to cycle from the inactive GDP-bound state to the active GTP-bound form. Here, we describe the identification of potent and cell-active small-molecule inhibitors which efficiently disrupt the interaction between KRAS and its exchange factor SOS1, a mode of action confirmed by a series of biophysical techniques. The binding sites, mode of action, and selectivity were elucidated using crystal structures of KRASG12C-SOS1, SOS1, and SOS2. By preventing formation of the KRAS-SOS1 complex, these inhibitors block reloading of KRAS with GTP, leading to antiproliferative activity. The final compound 23 (BAY-293) selectively inhibits the KRAS-SOS1 interaction with an IC50 of 21 nM and is a valuable chemical probe for future investigations.


Subject(s)
Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , SOS1 Protein/antagonists & inhibitors , Cell Line , Crystallography, X-Ray , Drug Discovery , Fluorescence Resonance Energy Transfer , High-Throughput Screening Assays , Humans , Protein Binding , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/chemistry , SOS1 Protein/metabolism , Signal Transduction
10.
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
11.
Proc Natl Acad Sci U S A ; 113(17): 4711-6, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27078104

ABSTRACT

Cancerous cells have an acutely increased demand for energy, leading to increased levels of human glucose transporter 1 (hGLUT1). This up-regulation suggests hGLUT1 as a target for therapeutic inhibitors addressing a multitude of cancer types. Here, we present three inhibitor-bound, inward-open structures of WT-hGLUT1 crystallized with three different inhibitors: cytochalasin B, a nine-membered bicyclic ring fused to a 14-membered macrocycle, which has been described extensively in the literature of hGLUTs, and two previously undescribed Phe amide-derived inhibitors. Despite very different chemical backbones, all three compounds bind in the central cavity of the inward-open state of hGLUT1, and all binding sites overlap the glucose-binding site. The inhibitory action of the compounds was determined for hGLUT family members, hGLUT1-4, using cell-based assays, and compared with homology models for these hGLUT members. This comparison uncovered a probable basis for the observed differences in inhibition between family members. We pinpoint regions of the hGLUT proteins that can be targeted to achieve isoform selectivity, and show that these same regions are used for inhibitors with very distinct structural backbones. The inhibitor cocomplex structures of hGLUT1 provide an important structural insight for the design of more selective inhibitors for hGLUTs and hGLUT1 in particular.


Subject(s)
Cytochalasins/chemistry , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/ultrastructure , Glucose/chemistry , Phenylalanine/analogs & derivatives , Amino Acid Sequence , Binding Sites , Computer Simulation , Conserved Sequence , Humans , Models, Chemical , Models, Molecular , Phenylalanine/chemistry , Protein Binding , Protein Conformation
12.
J Med Chem ; 59(10): 4578-600, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27075367

ABSTRACT

Protein lysine methyltransferases have recently emerged as a new target class for the development of inhibitors that modulate gene transcription or signaling pathways. SET and MYND domain containing protein 2 (SMYD2) is a catalytic SET domain containing methyltransferase reported to monomethylate lysine residues on histone and nonhistone proteins. Although several studies have uncovered an important role of SMYD2 in promoting cancer by protein methylation, the biology of SMYD2 is far from being fully understood. Utilization of highly potent and selective chemical probes for target validation has emerged as a concept which circumvents possible limitations of knockdown experiments and, in particular, could result in an improved exploration of drug targets with a complex underlying biology. Here, we report the development of a potent, selective, and cell-active, substrate-competitive inhibitor of SMYD2, which is the first reported inhibitor suitable for in vivo target validation studies in rodents.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Pyridazines/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HEK293 Cells , Histone-Lysine N-Methyltransferase/metabolism , Humans , Models, Molecular , Molecular Structure , Pyridazines/chemical synthesis , Pyridazines/chemistry , Structure-Activity Relationship , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
13.
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
14.
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
15.
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
16.
Acta Crystallogr D Biol Crystallogr ; 67(Pt 3): 156-66, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21358046

ABSTRACT

A crystallographic fragment screen was carried out to identify starting points for the development of inhibitors of protein kinase Pim-1, a potential target for tumour therapy. All fragment hits identified via soaking in this study turned out to bind to the unusually hydrophobic pocket at the hinge region. The most potent fragments, two cinnamic acid derivatives (with a best IC(50) of 130 µM), additionally form a well defined hydrogen bond. The balance between hydrophobic and polar interactions makes these molecules good starting points for further optimization. Pim-2 inhibitors from a recently reported high-throughput screening campaign also feature a cinnamic acid moiety. Two of these Pim-2 inhibitors were synthesized, their potencies against Pim-1 were determined and their cocrystal structures were elucidated in order to determine to what degree the binding modes identified by fragment screening are conserved in optimized inhibitors. The structures show that the cinnamic acid moieties indeed adopt the same binding mode. Fragment screening thus correctly identified binding modes which are maintained when fragments are grown into larger and higher affinity inhibitors. The high-throughput screening-derived compound (E)-3-{3-[6-(4-aminocyclohexylamino)-pyrazin-2-yl]phenyl}acrylic acid (compound 1) is the most potent inhibitor of the cinnamic acid series for which the three-dimensional binding mode is known (IC(50) = 17 nM, K(d) = 28 nM). The structure reveals the molecular basis for the large gain in potency between the initial fragment hit and this optimized inhibitor.


Subject(s)
Cinnamates/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-pim-1/chemistry , Cinnamates/metabolism , Cinnamates/pharmacology , Crystallography, X-Ray , Inhibitory Concentration 50 , Ligands , Models, Molecular , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/metabolism , Structure-Activity Relationship , Thermodynamics
17.
J Mol Biol ; 377(1): 206-19, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18241888

ABSTRACT

Molecular interactions between near-IR fluorescent probes and specific antibodies may be exploited to generate novel smart probes for diagnostic imaging. Using a new phage display technology, we developed such antibody Fab fragments with subnanomolar binding affinity for tetrasulfocyanine, a near-IR in vivo imaging agent. Unexpectedly, some Fabs induced redshifts of the dye absorption peak of up to 44 nm. This is the largest shift reported for a biological system so far. Crystal structure determination and absorption spectroscopy in the crystal in combination with microcalorimetry and small-angle X-ray scattering in solution revealed that the redshift is triggered by formation of a Fab dimer, with tetrasulfocyanine being buried in a fully closed protein cavity within the dimer interface. The derived principle of shifting the absorption peak of a symmetric dye via packaging within a Fab dimer interface may be transferred to other diagnostic fluorophores, opening the way towards smart imaging probes that change their wavelength upon interaction with an antibody.


Subject(s)
Antibodies, Monoclonal/immunology , Coloring Agents/chemistry , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Indoles/chemistry , Indoles/immunology , Solvents/chemistry , Absorption , Amino Acid Sequence , Antibodies, Monoclonal/chemistry , Antibody Affinity , Calorimetry , Chromatography, Gel , Complementarity Determining Regions/chemistry , Crystallography, X-Ray , Dimerization , Fluorescence , Humans , Models, Molecular , Molecular Sequence Data , Peptide Library , Scattering, Small Angle , Spectrophotometry, Ultraviolet
18.
Expert Opin Drug Discov ; 3(4): 391-401, 2008 Apr.
Article in English | MEDLINE | ID: mdl-23489095

ABSTRACT

BACKGROUND: With long and costly drug development times there is a need in the pharmaceutical industry to prioritize targets early in the drug discovery process. One of the possible criteria is 'protein drugability', a term with multiple understandings in the literature. Among others, it is the likelihood of finding a selective, low-molecular weight molecule that binds with high affinity to the target. OBJECTIVE: Which methods are available for drugability prediction? What can be achieved by such predictions and how can they influence the target prioritization process? METHODS: The main focus is on sequence- and structure-related computational methods for drugability prediction, giving an overview on their background as well as their bias and limitations with an emphasis on the structural biology point of view. RESULTS/CONCLUSION: Structural drugability assessment presents one criterion for prioritization of a target portfolio by enabling classification of targets into low, average, or high drugability.

19.
J Mol Biol ; 369(3): 735-45, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17449059

ABSTRACT

The Ser/Thr protein kinase MAPKAP kinase 2 (MK2) plays a crucial role in inflammation. We determined the structure of the kinase domain of MK2 in complex with a low molecular mass inhibitor in two different crystal forms, obtained from soaking and co-crystallization. To our knowledge, these are the first structures of MK2 showing the binding mode of an inhibitor with high binding affinity (IC50 8.5 nM). The two crystal forms revealed conformational flexibility in the binding site and extend the experimental basis for rational drug design. Crystal form-1 contained one MK2 molecule per asymmetric unit. Form-2 contained 12 molecules, which arrange into two different types of MK2 trimers. One of them may serve as a model for an intermediate state during substrate phosphorylation, as each MK2 monomer places its activation segment into the substrate peptide binding groove of the trimer neighbor.


Subject(s)
Enzyme Inhibitors/chemistry , Protein Kinases/chemistry , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Binding Sites , Crystallization , Dimerization , Drug Design , Electrons , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Serine-Threonine Kinases
20.
Article in English | MEDLINE | ID: mdl-17329818

ABSTRACT

Tetrasulfocyanine (TSC) has been described as a fluorescent probe for tumour imaging. The complex of TSC and the Fab antibody fragment MOR03268 has been crystallized in three different crystal forms. MOR03268 was identified from the HuCAL GOLD library and further optimized to bind TSC with high affinity (Kd = 0.6 nM). For two of the three crystal forms (forms 1 and 2), data sets could be collected to 2.8 and 2.85 A resolution, respectively. Form 1 belongs to space group I222, with unit-cell parameters a = 72, b = 99, c = 154 A. Form 2 belongs to space group P4(3)2(1)2, with unit-cell parameters a = b = 77, c = 379 A. Form 3 only diffracted to 8 A and was not analyzed further. Molecular-replacement solutions for forms 1 and 2 were found and rebuilding and refinement is in progress. Form 1 contains one Fab molecule per asymmetric unit, while form 2 harbours two. Judging from the green colour of the crystals, both forms contain the Fab molecule bound to the green TSC dye and in both the hydrolysis-sensitive dye molecule is protected from degradation for several weeks to months. The structures should reveal the molecular basis of the high-affinity recognition of TSC by the Fab molecule MOR03268.


Subject(s)
Fluorescent Dyes/chemistry , Immunoglobulin Fab Fragments/chemistry , Indoles , Amino Acid Sequence , Binding Sites, Antibody , Crystallization , Fluorescent Dyes/metabolism , Humans , Immunoglobulin Fab Fragments/biosynthesis , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Indoles/chemical synthesis , Indoles/metabolism , Molecular Sequence Data , Solutions/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...