Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Chem Biol ; 29(2): 249-258.e5, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34547225

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates plasma low-density lipoprotein cholesterol (LDL-C) levels by promoting hepatic LDL receptor (LDLR) degradation. Therapeutic antibodies that disrupt PCSK9-LDLR binding reduce LDL-C concentrations and cardiovascular disease risk. The epidermal growth factor precursor homology domain A (EGF-A) of the LDLR serves as a primary contact with PCSK9 via a flat interface, presenting a challenge for identifying small molecule PCSK9-LDLR disruptors. We employ an affinity-based screen of 1013in vitro-translated macrocyclic peptides to identify high-affinity PCSK9 ligands that utilize a unique, induced-fit pocket and partially disrupt the PCSK9-LDLR interaction. Structure-based design led to molecules with enhanced function and pharmacokinetic properties (e.g., 13PCSK9i). In mice, 13PCSK9i reduces plasma cholesterol levels and increases hepatic LDLR density in a dose-dependent manner. 13PCSK9i functions by a unique, allosteric mechanism and is the smallest molecule identified to date with in vivo PCSK9-LDLR disruptor function.


Subject(s)
Peptides/pharmacology , Proprotein Convertase 9/metabolism , Receptors, LDL/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Peptides/chemical synthesis , Peptides/chemistry , Protein Conformation , Receptors, LDL/metabolism
2.
ACS Comb Sci ; 22(3): 120-128, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32040908

ABSTRACT

The use of DNA-encoded libraries has emerged as a powerful hit generation technology. Combining the power of combinatorial chemistry to enumerate large compound collections with the efficiency of affinity selection in pools, the methodology makes it possible to interrogate vast chemical space against biological targets of pharmaceutical relevance. Thus, the chemical transformations employed for the synthesis of encoded libraries play a crucial role in the identification of diverse and drug-like starting points. Currently established transformations have mostly been limited to water-compatible reactions to accommodate the growing oligonucleotide tag. Herein, we describe the development of a practical catch-and-release methodology utilizing a cationic, amphiphilic PEG-based polymer to perform chemical transformations on immobilized DNA conjugates under anhydrous conditions. We demonstrate the usefulness of our APTAC (amphiphilic polymer-facilitated transformations under anhydrous conditions) approach by performing several challenging transformations on DNA-conjugated small molecules in pure organic solvents: the addition of a carbanion equivalent to a DNA-conjugated ketone in tetrahydrofuran, the synthesis of saturated heterocycles using the tin (Sn) amine protocol (SnAP) in dichloromethane, and the dual-catalytic (Ir/Ni) metallaphotoredox decarboxylative cross-coupling of carboxylic acids to DNA-conjugated aryl halides in DMSO. In addition, we demonstrate the feasibility of the latter in multititer-plate format.


Subject(s)
Combinatorial Chemistry Techniques , DNA/chemistry , Polymers/chemistry , Small Molecule Libraries/chemical synthesis , Surface-Active Agents/chemistry , Molecular Structure , Small Molecule Libraries/chemistry
3.
J Med Chem ; 61(18): 8120-8135, 2018 09 27.
Article in English | MEDLINE | ID: mdl-30137981

ABSTRACT

Chronic myelogenous leukemia (CML) arises from the constitutive activity of the BCR-ABL1 oncoprotein. Tyrosine kinase inhibitors (TKIs) that target the ATP-binding site have transformed CML into a chronic manageable disease. However, some patients develop drug resistance due to ATP-site mutations impeding drug binding. We describe the discovery of asciminib (ABL001), the first allosteric BCR-ABL1 inhibitor to reach the clinic. Asciminib binds to the myristate pocket of BCR-ABL1 and maintains activity against TKI-resistant ATP-site mutations. Although resistance can emerge due to myristate-site mutations, these are sensitive to ATP-competitive inhibitors so that combinations of asciminib with ATP-competitive TKIs suppress the emergence of resistance. Fragment-based screening using NMR and X-ray yielded ligands for the myristate pocket. An NMR-based conformational assay guided the transformation of these inactive ligands into ABL1 inhibitors. Further structure-based optimization for potency, physicochemical, pharmacokinetic, and drug-like properties, culminated in asciminib, which is currently undergoing clinical studies in CML patients.


Subject(s)
Drug Discovery , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Niacinamide/analogs & derivatives , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Allosteric Regulation , Animals , Dogs , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice , Models, Molecular , Molecular Structure , Mutation , Niacinamide/chemistry , Niacinamide/pharmacology , Phosphorylation , Protein Conformation , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Nature ; 543(7647): 733-737, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28329763

ABSTRACT

Chronic myeloid leukaemia (CML) is driven by the activity of the BCR-ABL1 fusion oncoprotein. ABL1 kinase inhibitors have improved the clinical outcomes for patients with CML, with over 80% of patients treated with imatinib surviving for more than 10 years. Second-generation ABL1 kinase inhibitors induce more potent molecular responses in both previously untreated and imatinib-resistant patients with CML. Studies in patients with chronic-phase CML have shown that around 50% of patients who achieve and maintain undetectable BCR-ABL1 transcript levels for at least 2 years remain disease-free after the withdrawal of treatment. Here we characterize ABL001 (asciminib), a potent and selective allosteric ABL1 inhibitor that is undergoing clinical development testing in patients with CML and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukaemia. In contrast to catalytic-site ABL1 kinase inhibitors, ABL001 binds to the myristoyl pocket of ABL1 and induces the formation of an inactive kinase conformation. ABL001 and second-generation catalytic inhibitors have similar cellular potencies but distinct patterns of resistance mutations, with genetic barcoding studies revealing pre-existing clonal populations with no shared resistance between ABL001 and the catalytic inhibitor nilotinib. Consistent with this profile, acquired resistance was observed with single-agent therapy in mice; however, the combination of ABL001 and nilotinib led to complete disease control and eradicated CML xenograft tumours without recurrence after the cessation of treatment.


Subject(s)
Allosteric Site/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Niacinamide/analogs & derivatives , Pyrazoles/pharmacology , Allosteric Regulation/drug effects , Animals , Catalytic Domain/drug effects , Cell Proliferation/drug effects , Dasatinib/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Therapy, Combination , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mutation , Niacinamide/pharmacology , Niacinamide/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Xenograft Model Antitumor Assays
5.
Angew Chem Int Ed Engl ; 54(48): 14575-9, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26457482

ABSTRACT

Targeting drugs to their desired site of action can increase their safety and efficacy. Bisphosphonates are prototypical examples of drugs targeted to bone. However, bisphosphonate bone affinity is often considered too strong and cannot be significantly modulated without losing activity on the enzymatic target, farnesyl pyrophosphate synthase (FPPS). Furthermore, bisphosphonate bone affinity comes at the expense of very low and variable oral bioavailability. FPPS inhibitors were developed with a monophosphonate as a bone-affinity tag that confers moderate affinity to bone, which can furthermore be tuned to the desired level, and the relationship between structure and bone affinity was evaluated by using an NMR-based bone-binding assay. The concept of targeting drugs to bone with moderate affinity, while retaining oral bioavailability, has broad application to a variety of other bone-targeted drugs.


Subject(s)
Bone and Bones/drug effects , Drug Delivery Systems , Administration, Oral , Biological Availability , Bone and Bones/enzymology , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Geranyltranstransferase/antagonists & inhibitors , Humans
6.
ChemMedChem ; 10(11): 1884-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26381451

ABSTRACT

Farnesyl pyrophosphate synthase (FPPS) is an established target for the treatment of bone diseases, but also shows promise as an anticancer and anti-infective drug target. Currently available anti-FPPS drugs are active-site-directed bisphosphonate inhibitors, the peculiar pharmacological profile of which is inadequate for therapeutic indications beyond bone diseases. The recent discovery of an allosteric binding site has paved the way toward the development of novel non-bisphosphonate FPPS inhibitors with broader therapeutic potential, notably as immunomodulators in oncology. Herein we report the discovery, by an integrated lead finding approach, of two new chemical classes of allosteric FPPS inhibitors that belong to the salicylic acid and quinoline chemotypes. We present their synthesis, biochemical and cellular activities, structure-activity relationships, and provide X-ray structures of several representative FPPS complexes. These novel allosteric FPPS inhibitors are devoid of any affinity for bone mineral and could serve as leads to evaluate their potential in none-bone diseases.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Geranyltranstransferase/antagonists & inhibitors , Quinolines/pharmacology , Salicylic Acid/pharmacology , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Geranyltranstransferase/metabolism , Humans , Molecular Structure , Quinolines/chemical synthesis , Quinolines/chemistry , Salicylic Acid/chemical synthesis , Salicylic Acid/chemistry , Structure-Activity Relationship
7.
Nat Chem Biol ; 6(9): 660-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20711197

ABSTRACT

Bisphosphonates are potent inhibitors of farnesyl pyrophosphate synthase (FPPS) and are highly efficacious in the treatment of bone diseases such as osteoporosis, Paget's disease and tumor-induced osteolysis. In addition, the potential for direct antitumor effects has been postulated on the basis of in vitro and in vivo studies and has recently been demonstrated clinically in early breast cancer patients treated with the potent bisphosphonate zoledronic acid. However, the high affinity of bisphosphonates for bone mineral seems suboptimal for the direct treatment of soft-tissue tumors. Here we report the discovery of the first potent non-bisphosphonate FPPS inhibitors. These new inhibitors bind to a previously unknown allosteric site on FPPS, which was identified by fragment-based approaches using NMR and X-ray crystallography. This allosteric and druggable pocket allows the development of a new generation of FPPS inhibitors that are optimized for direct antitumor effects in soft tissue.


Subject(s)
Diphosphonates , Drug Evaluation, Preclinical , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Geranyltranstransferase/antagonists & inhibitors , Allosteric Regulation , Allosteric Site , Bone and Bones/chemistry , Bone and Bones/metabolism , Crystallography, X-Ray , Diphosphonates/analysis , Diphosphonates/chemistry , Diphosphonates/metabolism , Diphosphonates/pharmacology , Enzyme Inhibitors/chemistry , Geranyltranstransferase/metabolism , Humans , Imidazoles/analysis , Imidazoles/chemistry , Imidazoles/pharmacology , Magnetic Resonance Spectroscopy , Soft Tissue Neoplasms/drug therapy , Zoledronic Acid
8.
J Am Chem Soc ; 132(20): 7043-8, 2010 May 26.
Article in English | MEDLINE | ID: mdl-20450175

ABSTRACT

Allosteric inhibitors of Bcr-Abl have emerged as a novel therapeutic option for the treatment of CML. Using fragment-based screening, a search for novel Abl inhibitors that bind to the myristate pocket was carried out. Here we show that not all myristate ligands are functional inhibitors, but that the conformational state of C-terminal helix_I is a structural determinant for functional activity. We present an NMR-based conformational assay to monitor the conformation of this crucial helix_I and show that myristate ligands that bend helix_I are functional antagonists, whereas ligands that bind to the myristate pocket but do not induce this conformational change are kinase agonists. Activation of c-Abl by allosteric agonists has been confirmed in a biochemical assay.


Subject(s)
Drug Evaluation, Preclinical/methods , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/agonists , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Allosteric Regulation , Animals , Enzyme Activation/drug effects , Humans , Ligands , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Myristic Acid/metabolism , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Structure, Secondary , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/metabolism
9.
Biochim Biophys Acta ; 1804(3): 454-62, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20152788

ABSTRACT

The ATP-competitive inhibitors dasatinib and nilotinib, which bind to catalytically different conformations of the Abl kinase domain, have recently been approved for the treatment of imatinib-resistant CML. These two new drugs, albeit very efficient against most of the imatinib-resistant mutants of Bcr-Abl, fail to effectively suppress the Bcr-Abl activity of the T315I (or gatekeeper) mutation. Generating new ATP site-binding drugs that target the T315I in Abl has been hampered, amongst others, by target selectivity, which is frequently an issue when developing ATP-competitive inhibitors. Recently, using an unbiased cellular screening approach, GNF-2, a non-ATP-competitive inhibitor, has been identified that demonstrates cellular activity against Bcr-Abl transformed cells. The exquisite selectivity of GNF-2 is due to the finding that it targets the myristate binding site located near the C-terminus of the Abl kinase domain, as demonstrated by genetic approaches, solution NMR and X-ray crystallography. GNF-2, like myristate, is able to induce and/or stabilize the clamped inactive conformation of Abl analogous to the SH2-Y527 interaction of Src. The molecular mechanism for allosteric inhibition by the GNF-2 inhibitor class, and the combined effects with ATP-competitive inhibitors such as nilotinib and imatinib on wild-type Abl and imatinib-resistant mutants, in particular the T315I gatekeeper mutant, are reviewed.


Subject(s)
Adenosine Triphosphate/chemistry , Myristic Acid/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/chemistry , Adenosine Triphosphate/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/genetics , Benzamides , Crystallography, X-Ray , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Imatinib Mesylate , Mutation, Missense , Myristic Acid/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Nuclear Magnetic Resonance, Biomolecular , Piperazines/chemistry , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/chemistry , Pyrimidines/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...