Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Exp Med ; 221(3)2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38334978

ABSTRACT

An effective cancer therapy requires killing cancer cells and targeting the tumor microenvironment (TME). Searching for molecules critical for multiple cell types in the TME, we identified NR4A1 as one such molecule that can maintain the immune suppressive TME. Here, we establish NR4A1 as a valid target for cancer immunotherapy and describe a first-of-its-kind proteolysis-targeting chimera (PROTAC, named NR-V04) against NR4A1. NR-V04 degrades NR4A1 within hours in vitro and exhibits long-lasting NR4A1 degradation in tumors with an excellent safety profile. NR-V04 inhibits and frequently eradicates established tumors. At the mechanistic level, NR-V04 induces the tumor-infiltrating (TI) B cells and effector memory CD8+ T (Tem) cells and reduces monocytic myeloid-derived suppressor cells (m-MDSC), all of which are known to be clinically relevant immune cell populations in human melanomas. Overall, NR-V04-mediated NR4A1 degradation holds promise for enhancing anticancer immune responses and offers a new avenue for treating various types of cancers such as melanoma.


Subject(s)
Melanoma , Myeloid-Derived Suppressor Cells , Humans , Cell Line, Tumor , Immunotherapy , Melanoma/pathology , Myeloid-Derived Suppressor Cells/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Tumor Microenvironment , Proteolysis Targeting Chimera
2.
bioRxiv ; 2023 Aug 13.
Article in English | MEDLINE | ID: mdl-37609171

ABSTRACT

An effective cancer therapy requires both killing cancer cells and targeting tumor-promoting pathways or cell populations within the tumor microenvironment (TME). We purposely search for molecules that are critical for multiple tumor-promoting cell types and identified nuclear receptor subfamily 4 group A member 1 (NR4A1) as one such molecule. NR4A1 has been shown to promote the aggressiveness of cancer cells and maintain the immune suppressive TME. Using genetic and pharmacological approaches, we establish NR4A1 as a valid therapeutic target for cancer therapy. Importantly, we have developed the first-of-its kind proteolysis-targeting chimera (PROTAC, named NR-V04) against NR4A1. NR-V04 effectively degrades NR4A1 within hours of treatment in vitro and sustains for at least 4 days in vivo, exhibiting long-lasting NR4A1-degradation in tumors and an excellent safety profile. NR-V04 leads to robust tumor inhibition and sometimes eradication of established melanoma tumors. At the mechanistic level, we have identified an unexpected novel mechanism via significant induction of tumor-infiltrating (TI) B cells as well as an inhibition of monocytic myeloid derived suppressor cells (m-MDSC), two clinically relevant immune cell populations in human melanomas. Overall, NR-V04-mediated NR4A1 degradation holds promise for enhancing anti-cancer immune responses and offers a new avenue for treating various types of cancer.

3.
Blood Adv ; 5(5): 1388-1402, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33661300

ABSTRACT

Sickle cell disease (SCD) is associated with hemolysis, vascular inflammation, and organ damage. Affected patients experience chronic painful vaso-occlusive events requiring hospitalization. Hypoxia-induced polymerization of sickle hemoglobin S (HbS) contributes to sickling of red blood cells (RBCs) and disease pathophysiology. Dilution of HbS with nonsickling hemoglobin or hemoglobin with increased oxygen affinity, such as fetal hemoglobin or HbS bound to aromatic aldehydes, is clinically beneficial in decreasing polymerization. We investigated a novel alternate approach to modify HbS and decrease polymerization by inhibiting methionine aminopeptidase 2 (MetAP2), which cleaves the initiator methionine (iMet) from Val1 of α-globin and ßS-globin. Kinetic studies with MetAP2 show that ßS-globin is a fivefold better substrate than α-globin. Knockdown of MetAP2 in human umbilical cord blood-derived erythroid progenitor 2 cells shows more extensive modification of α-globin than ß-globin, consistent with kinetic data. Treatment of human erythroid cells in vitro or Townes SCD mice in vivo with selective MetAP2 inhibitors extensively modifies both globins with N-terminal iMet and acetylated iMet. HbS modification by MetAP2 inhibition increases oxygen affinity, as measured by decreased oxygen tension at which hemoglobin is 50% saturated. Acetyl-iMet modification on ßS-globin delays HbS polymerization under hypoxia. MetAP2 inhibitor-treated Townes mice reach 50% total HbS modification, significantly increasing the affinity of RBCs for oxygen, increasing whole blood single-cell RBC oxygen saturation, and decreasing fractional flow velocity losses in blood rheology under decreased oxygen pressures. Crystal structures of modified HbS variants show stabilization of the nonpolymerizing high O2-affinity R2 state, explaining modified HbS antisickling activity. Further study of MetAP2 inhibition as a potential therapeutic target for SCD is warranted.


Subject(s)
Anemia, Sickle Cell , Hemoglobin, Sickle , Aminopeptidases , Anemia, Sickle Cell/drug therapy , Animals , Antisickling Agents/pharmacology , Humans , Kinetics , Metalloendopeptidases , Methionyl Aminopeptidases , Mice , Polymerization
4.
Immunology ; 147(4): 399-413, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26694902

ABSTRACT

Retinoic acid receptor-related orphan nuclear receptor γ (RORγ) orchestrates a pro-inflammatory gene expression programme in multiple lymphocyte lineages including T helper type 17 (Th17) cells, γδ T cells, innate lymphoid cells and lymphoid tissue inducer cells. There is compelling evidence that RORγ-expressing cells are relevant targets for therapeutic intervention in the treatment of autoimmune and inflammatory diseases. Unlike Th17 cells, where RORγ expression is induced under specific pro-inflammatory conditions, γδ T cells and other innate-like immune cells express RORγ in the steady state. Small molecule mediated disruption of RORγ function in cells with pre-existing RORγ transcriptional complexes represents a significant and challenging pharmacological hurdle. We present data demonstrating that a novel, selective and potent small molecule RORγ inhibitor can block the RORγ-dependent gene expression programme in both Th17 cells and RORγ-expressing γδ T cells as well as a disease-relevant subset of human RORγ-expressing memory T cells. Importantly, systemic administration of this inhibitor in vivo limits pathology in an innate lymphocyte-driven mouse model of psoriasis.


Subject(s)
Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Benzamides/pharmacology , Gene Expression Regulation/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Pyridines/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dermatitis/drug therapy , Dermatitis/immunology , Dermatitis/metabolism , Dermatitis/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Immunologic Memory/drug effects , Interleukin-17/metabolism , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
5.
Bioorg Med Chem Lett ; 25(15): 2985-90, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26048789

ABSTRACT

The nuclear receptor RORγ plays a central role in controlling a pro-inflammatory gene expression program in several lymphocyte lineages including TH17 cells. RORγ-dependent inflammation has been implicated in the pathogenesis of several major autoimmune diseases and thus RORγ is an attractive target for therapeutic intervention in these diseases. Starting from a lead biaryl compound 4a, replacement of the head phenyl moiety with a substituted aminopyrazole group resulted in a series with improved physical properties. Further SAR exploration led to analogues (e.g., 4j and 5m) as potent RORγ inverse agonists.


Subject(s)
Benzamides/chemistry , Benzamides/pharmacology , Drug Inverse Agonism , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Pyrazoles/chemistry , Pyrazoles/pharmacology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Humans , Interleukin-17/immunology , Mice , Models, Molecular , Nuclear Receptor Subfamily 1, Group F, Member 3/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Spleen/cytology , Spleen/drug effects , Spleen/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
6.
Bioorg Med Chem Lett ; 25(15): 2991-7, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26048806

ABSTRACT

RORγt is a pivotal regulator of a pro-inflammatory gene expression program implicated in the pathology of several major human immune-mediated diseases. Evidence from mouse models demonstrates that genetic or pharmacological inhibition of RORγ activity can block the production of pathogenic cytokines, including IL-17, and convey therapeutic benefit. We have identified and developed a biaryl-carboxylamide series of RORγ inverse agonists via a structure based design approach. Co-crystal structures of compounds 16 and 48 supported the design approach and confirmed the key interactions with RORγ protein; the hydrogen bonding with His479 was key to the significant improvement in inverse agonist effect. The results have shown this is a class of potent and selective RORγ inverse agonists, with demonstrated oral bioavailability in rodents.


Subject(s)
Amides/chemistry , Amides/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Drug Inverse Agonism , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Amides/pharmacokinetics , Animals , Biphenyl Compounds/pharmacokinetics , Cell Line , Cytokines/immunology , Drug Discovery , Humans , Hydrogen Bonding , Interleukin-17/immunology , Mice , Molecular Docking Simulation , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Rats
8.
ACS Med Chem Lett ; 4(4): 414-8, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-24900686

ABSTRACT

To resolve the metabolite redox cycling associated with our earlier clinical compound 2, we carried out lead optimization of lead molecule 1. Compound 4 showed improved lipophilic ligand efficiency and demonstrated robust glucose lowering in diet-induced obese mice without a liability in predictive preclinical drug safety studies. Thus, it was selected as a clinical candidate and further studied in type 2 diabetic patients. Clinical data suggests no evidence of metabolite cycling, which is consistent with the preclinical profiling of metabolism.

9.
J Med Chem ; 55(16): 7021-36, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22809456

ABSTRACT

Glucokinase (GK) activation as a potential strategy to treat type 2 diabetes (T2D) is well recognized. Compound 1, a glucokinase activator (GKA) lead that we have previously disclosed, caused reversible hepatic lipidosis in repeat-dose toxicology studies. We hypothesized that the hepatic lipidosis was due to the structure-based toxicity and later established that it was due to the formation of a thiourea metabolite, 2. Subsequent SAR studies of 1 led to the identification of a pyrazine-based lead analogue 3, lacking the thiazole moiety. In vivo metabolite identification studies, followed by the independent synthesis and profiling of the cyclopentyl keto- and hydroxyl- metabolites of 3, led to the selection of piragliatin, 4, as the clinical lead. Piragliatin was found to lower pre- and postprandial glucose levels, improve the insulin secretory profile, increase ß-cell sensitivity to glucose, and decrease hepatic glucose output in patients with T2D.


Subject(s)
Benzeneacetamides/chemical synthesis , Diabetes Mellitus, Type 2/drug therapy , Enzyme Activators/chemical synthesis , Glucokinase/metabolism , Hypoglycemic Agents/chemical synthesis , Animals , Benzeneacetamides/pharmacokinetics , Benzeneacetamides/pharmacology , Dogs , Enzyme Activators/pharmacokinetics , Enzyme Activators/pharmacology , Female , Glucose/metabolism , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Lipidoses/metabolism , Liver/metabolism , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Postprandial Period , Rabbits , Rats , Rats, Wistar , Stereoisomerism , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 20(14): 4140-6, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20541935

ABSTRACT

7-N-Acetamide-4-methoxy-2-aminobenzothiazole 4-fluorobenzamide (compound 1) was chosen as a drug-like and non-xanthine based starting point for the discovery of A(2B) receptor antagonists because of its slight selectivity against A(1) and A(2A) receptors and modest A(2B) potency. SAR exploration of compound 1 described herein included modifications to the 7-N-acetamide group, substitution of the 4-methoxy group by halogens as well as replacement of the p-flouro-benzamide side chain. This work culminated in the identification of compound 37 with excellent A(2B) potency, modest selectivity versus A(2A) and A(1) receptors, and good rodent PK properties.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Benzothiazoles/pharmacology , Receptor, Adenosine A2B/metabolism , Xanthine/chemistry , Adenosine A2 Receptor Antagonists/chemistry , Benzothiazoles/chemistry , Structure-Activity Relationship
11.
J Med Chem ; 53(9): 3618-25, 2010 May 13.
Article in English | MEDLINE | ID: mdl-20405948

ABSTRACT

Glucokinase (GK) is a glucose sensor that couples glucose metabolism to insulin release. The important role of GK in maintaining glucose homeostasis is illustrated in patients with GK mutations. In this publication, identification of the hit molecule 1 and its SAR development, which led to the discovery of potent allosteric GK activators 9a and 21a, is described. Compound 21a (RO0281675) was used to validate the clinical relevance of targeting GK to treat type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucokinase/drug effects , Hypoglycemic Agents/chemistry , Sulfones/pharmacology , Thiazoles/pharmacology , Animals , Blood Glucose , Cell Line , Cytotoxins , Dose-Response Relationship, Drug , Drug Discovery , Humans , Insulin , Male , Mice , Pharmacokinetics , Structure-Activity Relationship , Sulfones/chemistry , Sulfones/toxicity , Thiazoles/chemistry , Thiazoles/toxicity
12.
Curr Med Chem ; 14(23): 2471-81, 2007.
Article in English | MEDLINE | ID: mdl-17979700

ABSTRACT

Factor Xa (fXa) is a critical serine protease situated at the confluence of the intrinsic and extrinsic pathways of the blood coagulation cascade. FXa catalyses the conversion of prothrombin to thrombin via the prothrombinase complex. Its singular role in thrombin generation, coupled with its potentiating effects on clot formation render it an attractive target for therapeutic intervention. Otamixaban is a synthetically derived parenteral fXa inhibitor currently in late stage clinical development at Sanofi-Aventis for the management of acute coronary syndrome. Otamixaban is a potent (Ki = 0.5 nM), selective, rapid acting, competitive and reversible fXa inhibitor that effectively inhibits both free and prothrombinase-bound fXa. In vivo experiments have demonstrated that Otamixaban is highly efficacious in rodent, canine and porcine models of thrombosis. In addition, recent clinical findings indicate that Otamixaban is efficacious, safe and well tolerated in humans and therefore has considerable potential for the treatment of acute coronary syndrome. This review article chronicles the discovery and pre-clinical data surrounding the fXa inhibitor Otamixaban as well as the recent clinical findings in humans.


Subject(s)
Cyclic N-Oxides/pharmacology , Factor Xa Inhibitors , Pyridines/pharmacology , Animals , Clinical Trials as Topic , Cyclic N-Oxides/chemistry , Dogs , Drug Design , Esters/chemistry , Haplorhini , Humans , Mice , Models, Biological , Models, Chemical , Molecular Conformation , Pyridines/chemistry , Rabbits , Serine Endopeptidases/chemistry , Signal Transduction , Treatment Outcome
13.
Curr Med Chem ; 13(15): 1839-43, 2006.
Article in English | MEDLINE | ID: mdl-16787225

ABSTRACT

Glucokinase (GK) is a molecular sensor that regulates glucose induced insulin secretion in pancreatic beta-cells and glucose homeostasis in the liver via catalysis of glucose to glucose-6-phosphate. The recent discovery and development of small molecule glucokinase activators represents a potentially important development for the management of type 2 diabetes. Since the discovery of the first orally active small molecule GK activator RO0281675, a number of research groups have reported the identification of potent activators. In this review, the biological significance of GK in whole body glucose homeostasis is briefly described coupled with the recent progress regarding the identification of novel small molecule GK activators.


Subject(s)
Blood Glucose/analysis , Diabetes Mellitus, Type 2/drug therapy , Enzyme Activators/pharmacology , Enzyme Activators/therapeutic use , Glucokinase/metabolism , Animals , Humans
14.
Bioorg Med Chem Lett ; 13(17): 2895-8, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-14611852
15.
Science ; 301(5631): 370-3, 2003 Jul 18.
Article in English | MEDLINE | ID: mdl-12869762

ABSTRACT

Glucokinase (GK) plays a key role in whole-body glucose homeostasis by catalyzing the phosphorylation of glucose in cells that express this enzyme, such as pancreatic beta cells and hepatocytes. We describe a class of antidiabetic agents that act as nonessential, mixed-type GK activators (GKAs) that increase the glucose affinity and maximum velocity (Vmax) of GK. GKAs augment both hepatic glucose metabolism and glucose-induced insulin secretion from isolated rodent pancreatic islets, consistent with the expression and function of GK in both cell types. In several rodent models of type 2 diabetes mellitus, GKAs lowered blood glucose levels, improved the results of glucose tolerance tests, and increased hepatic glucose uptake. These findings may lead to the development of new drug therapies for diabetes.


Subject(s)
Carrier Proteins , Diabetes Mellitus, Type 2/drug therapy , Glucokinase/metabolism , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/drug effects , Liver/drug effects , Thiazoles/pharmacology , Adaptor Proteins, Signal Transducing , Allosteric Regulation , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Activation , Enzyme Activators/chemistry , Enzyme Activators/pharmacology , Glucose Tolerance Test , Homeostasis , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Insulin/blood , Insulin Secretion , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/metabolism , Keto Acids/metabolism , Liver/metabolism , Liver Glycogen/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Proteins/metabolism , Proteins/pharmacology , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Stereoisomerism , Thiazoles/chemistry
16.
Bioorg Med Chem Lett ; 12(12): 1667-70, 2002 Jun 17.
Article in English | MEDLINE | ID: mdl-12039586

ABSTRACT

A systematic modification of the C(3) side-chain of the beta-aminoester class of factor Xa inhibitors and a survey of P(4) variations is described. These changes have resulted in the identification of sub-nanomolar inhibitors with improved selectivity versus related proteases. Coagulation parameters (i.e., APTT doubling concentrations) are also improved.


Subject(s)
Factor Xa Inhibitors , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Esters
17.
Bioorg Med Chem Lett ; 12(12): 1671-4, 2002 Jun 17.
Article in English | MEDLINE | ID: mdl-12039587

ABSTRACT

Further optimization of the beta-aminoester class of factor Xa (fXa) inhibitors is described culminating in the identification of 9c (FXV673), a potent and selective factor Xa inhibitor with excellent in vivo anticoagulant activity. An X-ray structure of FXV673 bound to human fXa is also presented. Based on its selectivity, potent in vivo activity and favorable pre-clinical safety profile, FXV673 was selected for further development and is currently undergoing clinical trials.


Subject(s)
Anticoagulants/chemistry , Anticoagulants/pharmacology , Cyclic N-Oxides/chemistry , Cyclic N-Oxides/pharmacology , Factor Xa Inhibitors , Pyridines/chemistry , Pyridines/pharmacology , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Crystallography, X-Ray , Esters , Humans , Models, Molecular , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...