Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
J Med Chem ; 64(24): 18193-18208, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34894681

ABSTRACT

As a result of emerging biological data suggesting that within the c-Jun N-terminal kinase (JNK) family, JNK1 and not JNK2 or JNK3 may be primarily responsible for fibrosis pathology, we sought to identify JNK inhibitors with an increased JNK1 bias relative to our previous clinical compound tanzisertib (CC-930). This manuscript reports the synthesis and structure-activity relationship (SAR) studies for a novel series of JNK inhibitors demonstrating an increased JNK1 bias. SAR optimization on a series of 2,4-dialkylamino-pyrimidine-5-carboxamides resulted in the identification of compounds possessing low nanomolar JNK inhibitory potency, overall kinome selectivity, and the ability to inhibit cellular phosphorylation of the direct JNK substrate c-Jun. Optimization of physicochemical properties in this series resulted in compounds that demonstrated excellent systemic exposure following oral dosing, enabling in vivo efficacy studies and the selection of a candidate for clinical development, CC-90001, which is currently in clinical trials (Phase II) in patients with idiopathic pulmonary fibrosis (NCT03142191).


Subject(s)
Cyclohexylamines/pharmacology , Drug Discovery , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Cyclohexylamines/therapeutic use , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Phosphorylation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Structure-Activity Relationship , Substrate Specificity
2.
Bioorg Med Chem Lett ; 30(23): 127625, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33096160
3.
J Mol Med (Berl) ; 98(8): 1161-1173, 2020 08.
Article in English | MEDLINE | ID: mdl-32632752

ABSTRACT

Small molecules targeting the cereblon-containing E3 ubiquitin ligase including thalidomide, lenalidomide, and pomalidomide modulate turnover of downstream client proteins and demonstrate pre-clinical and clinical anti-myeloma activity. Different drugs that engage with cereblon hold the potential of unique phenotypic effects, and we therefore studied the novel protein homeostatic modulator (PHM™) BTX306 with a unique thiophene-fused scaffold bearing a substituted phenylurea and glutarimide. This agent much more potently reduced human-derived myeloma cell line viability, with median inhibitory concentrations in the single nanomolar range versus micromolar values for lenalidomide or pomalidomide, and more potently activated caspases 3/8/9. While lenalidomide and pomalidomide induced greater degradation of Ikaros and Aiolos in myeloma cells, BTX306 more potently reduced levels of GSPT1, eRF1, CK1α, MCL-1, and c-MYC. Suppression of cereblon or overexpression of Aiolos or Ikaros induced relative resistance to BTX306, and this agent did not impact viability of murine hematopoietic cells in an in vivo model, demonstrating its specificity for human cereblon. Interestingly, BTX306 did show some reduced activity in lenalidomide-resistant cell line models but nonetheless retained its nanomolar potency in vitro, overcame bortezomib resistance, and was equipotent against otherwise isogenic cell line models with either wild-type or knockout TP53. Finally, BTX306 demonstrated strong activity against primary CD138-positive plasma cells, showed enhanced anti-proliferative activity in combination with bortezomib and dexamethasone, and was effective in an in vivo systemic model of multiple myeloma. Taken together, the data support further translational studies of BTX306 and its derivatives to the clinic for patients with relapsed and/or refractory myeloma. KEY MESSAGES: BTX306 has a unique thiophene-fused scaffold bearing phenylurea and glutarimide. BTX306 is more potent against myeloma cells than lenalidomide or pomalidomide. BTX306 overcomes myeloma cell resistance to lenalidomide or bortezomib in vitro. BTX306 is active against primary myeloma cells, and shows efficacy in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Drug Resistance, Neoplasm/drug effects , Lenalidomide/pharmacology , Proteostasis/drug effects , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Humans , Mice , Multiple Myeloma , Ubiquitin-Protein Ligases/antagonists & inhibitors
4.
ACS Med Chem Lett ; 10(5): 811-815, 2019 May 09.
Article in English | MEDLINE | ID: mdl-31098004

ABSTRACT

Fragment-based drug discovery (FBDD) is a well-established technology for lead compound generation in drug discovery. As this technology has evolved, the design of fragments for screening has also evolved to engender not just an understanding of the role of modulating the physicochemical properties of fragments (Rule of Three, Ro3) but also the importance and implications of incorporating shape and, in particular, 3D characteristics into fragments. Herein, we describe the design and synthesis of pyrrolidine-based fragments with good fragment-like (Ro3) physicochemical properties that effectively sample three-dimensional molecular space.

5.
Bioorg Med Chem Lett ; 29(3): 491-495, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30553737
6.
Cell ; 175(1): 171-185.e25, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30146162

ABSTRACT

CKIα ablation induces p53 activation, and CKIα degradation underlies the therapeutic effect of lenalidomide in a pre-leukemia syndrome. Here we describe the development of CKIα inhibitors, which co-target the transcriptional kinases CDK7 and CDK9, thereby augmenting CKIα-induced p53 activation and its anti-leukemic activity. Oncogene-driving super-enhancers (SEs) are highly sensitive to CDK7/9 inhibition. We identified multiple newly gained SEs in primary mouse acute myeloid leukemia (AML) cells and demonstrate that the inhibitors abolish many SEs and preferentially suppress the transcription elongation of SE-driven oncogenes. We show that blocking CKIα together with CDK7 and/or CDK9 synergistically stabilize p53, deprive leukemia cells of survival and proliferation-maintaining SE-driven oncogenes, and induce apoptosis. Leukemia progenitors are selectively eliminated by the inhibitors, explaining their therapeutic efficacy with preserved hematopoiesis and leukemia cure potential; they eradicate leukemia in MLL-AF9 and Tet2-/-;Flt3ITD AML mouse models and in several patient-derived AML xenograft models, supporting their potential efficacy in curing human leukemia.


Subject(s)
Casein Kinase Ialpha/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Animals , Apoptosis/drug effects , Casein Kinase Ialpha/physiology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/physiology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/physiology , DNA-Binding Proteins , Disease Models, Animal , Enhancer Elements, Genetic/genetics , Hematopoiesis , Humans , Mice , Mice, Inbred C57BL , Oncogene Proteins, Fusion/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Tumor Suppressor Protein p53/physiology , Xenograft Model Antitumor Assays
7.
J Med Chem ; 60(21): 8989-9002, 2017 11 09.
Article in English | MEDLINE | ID: mdl-28991472

ABSTRACT

Triple negative breast cancer (TNBC) remains a serious unmet medical need with discouragingly high relapse rates. We report here the synthesis and structure-activity relationship (SAR) of a novel series of 2,4,5-trisubstituted-7H-pyrrolo[2,3-d]pyrimidines with potent activity against TNBC tumor cell lines. These compounds were discovered from a TNBC phenotypic screen and possess a unique dual inhibition profile targeting TTK (mitotic exit) and CLK2 (mRNA splicing). Design and optimization, driven with a TNBC tumor cell assay, identified potent and selective compounds with favorable in vitro and in vivo activity profiles and good iv PK properties. This cell-based driven SAR produced compounds with strong single agent in vivo efficacy in multiple TNBC xenograft models without significant body weight loss. These data supported the nomination of CC-671 into IND-enabling studies as a single agent TNBC therapy.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/chemical synthesis , Triple Negative Breast Neoplasms/drug therapy , Animals , Cell Line, Tumor , Female , Heterografts , Humans , Mice , Mitosis/drug effects , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA Splicing/drug effects , Structure-Activity Relationship , Triple Negative Breast Neoplasms/enzymology
10.
ACS Chem Biol ; 4(6): 473-83, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19413326

ABSTRACT

As part of our effort to inhibit bacterial fatty acid biosynthesis through the recently validated target biotin carboxylase, we employed a unique combination of two emergent lead discovery strategies. We used both de novo fragment-based drug discovery and virtual screening, which employs 3D shape and electrostatic property similarity searching. We screened a collection of unbiased low-molecular-weight molecules and identified a structurally diverse collection of weak-binding but ligand-efficient fragments as potential building blocks for biotin carboxylase ATP-competitive inhibitors. Through iterative cycles of structure-based drug design relying on successive fragment costructures, we improved the potency of the initial hits by up to 3000-fold while maintaining their ligand-efficiency and desirable physicochemical properties. In one example, hit-expansion efforts resulted in a series of amino-oxazoles with antibacterial activity. These results successfully demonstrate that virtual screening approaches can substantially augment fragment-based screening approaches to identify novel antibacterial agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbon-Nitrogen Ligases/antagonists & inhibitors , Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Anti-Bacterial Agents/chemistry , Binding Sites , Carbon-Nitrogen Ligases/metabolism , Combinatorial Chemistry Techniques , Enzyme Inhibitors/chemistry , Ligands , Microbial Sensitivity Tests , Molecular Weight , Structure-Activity Relationship
11.
Am J Respir Crit Care Med ; 176(12): 1251-60, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17901412

ABSTRACT

RATIONALE: Cytokines secreted by T cells play a pivotal role in the pathogenesis of lung injury and fibrosis, and the transcription factors nuclear factor (NF)-kappaB and activator protein (AP)-1 are involved in the expression of cytokines from T cells during lung injury. OBJECTIVES: We assessed the potential therapeutic effect of SP100030, a specific inhibitor of T-cell NF-kappaB and AP-1 in lung fibrosis. METHODS: The effect of SP100030 was evaluated using a mouse model of chronic lung fibrosis. MEASUREMENTS AND MAIN RESULTS: Mice treated with SP100030, as compared with untreated mice, had significantly less cachexia and less lung injury and had decreased levels of inflammatory cytokines and growth factors, decreased activation of coagulation activation, and decreased collagen deposition in the lung. The inhibitory activity of SP100030 was dose dependent and was effective in acute and chronic phases of lung fibrosis. SP100030 inhibited the activation of the protein kinase C-isoform in T-cell lines and suppressed NF-kappaB-driven cytokine expression in CD4(+) and CD8(+) T cells. CONCLUSIONS: These results suggest that the specific inhibition of NF-kappaB could be useful for the treatment of lung fibrosis.


Subject(s)
Immunosuppressive Agents/pharmacology , NF-kappa B/drug effects , Pulmonary Fibrosis/drug therapy , T-Lymphocytes/drug effects , Animals , Bronchoalveolar Lavage Fluid/cytology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Humans , Jurkat Cells , Mice , Organic Chemicals/pharmacology , Pulmonary Fibrosis/chemically induced
12.
Bioorg Med Chem Lett ; 13(22): 4077-80, 2003 Nov 17.
Article in English | MEDLINE | ID: mdl-14592511

ABSTRACT

We have developed novel orally active quinazoline analogues as inhibitors of AP-1 and NF-kappaB mediated transcriptional activation. Among the derivatives prepared, 1-[2-(2-thienyl)quinazolin-4-ylamino]-3-methyl-3-pyrroline-2,5-dione (10) showed significant activity in an adjuvant-induced arthritis rat model by reducing the swelling by 65% in the non-injected foot. The synthesis, structure-activity relationship, and in vivo activity are described.


Subject(s)
NF-kappa B/antagonists & inhibitors , Quinazolines/chemical synthesis , Quinazolines/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Transcriptional Activation/drug effects , Administration, Oral , Animals , Arthritis, Experimental/drug therapy , Disease Models, Animal , Drug Design , Humans , Jurkat Cells , Quinazolines/administration & dosage , Rats , Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 12(18): 2573-7, 2002 Sep 16.
Article in English | MEDLINE | ID: mdl-12182863

ABSTRACT

Several analogues of ethyl 2-[(3-methyl-2,5-dioxo(3-pyrrolinyl))amino]-4-(trifluoromethyl)pyrimidine-5-carboxylate (1) were synthesized and tested as inhibitors of AP-1 and NF-kappaB mediated transcriptional activation in Jurkat T cells. From our SAR work, ethyl 2-[(3-methyl-2,5-dioxo(3-pyrrolinyl))-N-methylamino]-4-(trifluoromethyl)-pyrimidine-5-carboxylate was identified as a novel and potent inhibitor.


Subject(s)
Gene Expression Regulation/physiology , NF-kappa B/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrroles/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Humans , Jurkat Cells , NF-kappa B/physiology , Pyrimidines/chemistry , Pyrroles/chemistry , Structure-Activity Relationship , Transcription Factor AP-1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL