Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 125
1.
Biochem Pharmacol ; 196: 114725, 2022 02.
Article En | MEDLINE | ID: mdl-34384758

Aberrant cholesterol metabolism and homeostasis in the form of elevated cholesterol biosynthesis and dysregulated efflux and metabolism is well recognized as a major feature of metabolic reprogramming in solid tumors. Recent studies have emphasized on major drivers and regulators such as Myc, mutant p53, SREBP2, LXRs and oncogenic signaling pathways that play crucial roles in tumor cholesterol metabolic reprogramming. Therapeutics such as statins targeting the mevalonate pathway were tried at the clinic without showing consistent benefits to cancer patients. Nuclear receptors are prominent regulators of mammalian metabolism. Their de-regulation often drives tumorigenesis. RORγ and its immune cell-specific isoform RORγt play important functions in control of mammalian metabolism, circadian rhythm and immune responses. Although RORγ, together with its closely related members RORα and RORß were identified initially as orphan receptors, recent studies strongly support the conclusion that specific intermediates and metabolites of cholesterol pathways serve as endogenous ligands of RORγ. More recent studies also reveal a critical role of RORγ in tumorigenesis through major oncogenic pathways including acting a new master-like regulator of tumor cholesterol biosynthesis program. Importantly, an increasing number of RORγ orthosteric and allosteric ligands are being identified that display potent activities in blocking tumor growth and autoimmune disorders in preclinical models. This review summarizes the recent preclinical and clinical progress on RORγ with emphasis on its role in reprogramming tumor cholesterol metabolism and its regulation. It will also discuss RORγ functional mechanisms, context-specificity and its value as a therapeutic target for effective cancer treatment.


Anticholesteremic Agents/administration & dosage , Autoimmune Diseases/metabolism , Cholesterol/biosynthesis , Neoplasms/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Oncogenes/physiology , Animals , Antineoplastic Agents/administration & dosage , Autoimmune Diseases/drug therapy , Drug Delivery Systems/trends , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Neoplasms/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Oncogenes/drug effects
2.
Front Immunol ; 12: 699987, 2021.
Article En | MEDLINE | ID: mdl-34552583

Objective: IL-17A plays a major role in the pathogenesis of spondyloarthritis (SpA). Here we assessed the impact of inhibition of RAR related orphan receptor-γ (RORC), the key transcription factor controlling IL-17 production, on experimental SpA in HLA-B27 transgenic (tg) rats. Methods: Experimental SpA was induced by immunization of HLA-B27 tg rats with heat-inactivated Mycobacterium tuberculosis. Splenocytes obtained at day 7, 14 and 21 after immunization were restimulated ex vivo to assess the induction of pro-inflammatory cytokines. Rats were then prophylactically treated with a RORC inhibitor versus vehicle control. The biologic effect of RORC inhibition was assessed by pro-inflammatory cytokine expression in draining lymph nodes. Arthritis and spondylitis were monitored clinically, and the degree of peripheral and axial inflammation, destruction and new bone formation was confirmed by histology. Results: Ex vivo mRNA and protein analyses revealed the rapid and selective induction of IL-17A and IL-22 production by a variety of lymphocyte subsets upon disease induction in HLA-B27 tg rats. Prophylactic RORC inhibition in vivo suppressed the expression of IL-17A, IL17F, and IL-22 without affecting the expression of other T helper cell subset related genes. This biological effect did not translate into clinical efficacy as RORC inhibition significantly accelerated the onset of arthritis and spondylitis, and aggravated the clinical severity of arthritis. This worsening of experimental SpA was confirmed by histopathological demonstration of increased inflammation, destruction, and new bone formation. Conclusion: Despite a significant suppression of the IL-17 axis, RORC inhibitor treatment accelerates and aggravates experimental SpA in the HLA-B27 tg rat model.


Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Spondylarthritis/immunology , Spondylarthritis/pathology , Animals , Disease Models, Animal , Female , HLA-B27 Antigen/genetics , Male , Rats , Rats, Inbred Lew , Rats, Transgenic
3.
Acta Biochim Biophys Sin (Shanghai) ; 53(10): 1290-1299, 2021 Oct 12.
Article En | MEDLINE | ID: mdl-34379099

Clostridioides difficile infection (CDI) is a common infection of the gastrointestinal tract. Typically, 20%-30% of CDI patients experience recurrent C.difficile infection (RCDI). Although the role of Th17 in infectious and inflammatory diseases including CDI has gained attention, reports on the correlation between Th17 and RCDI are scarce. In this study, CDI and RCDI mice models were challenged with C. difficile. Serum lactic acid dehydrogenase, inflammatory factor levels, reverse transcriptase-polymerase chain reaction, western blot analysis, hematoxylin and eosin staining, immunohistochemistry, flow cytometry analysis, and enzyme-linked immunosorbent assay were performed on the CDI, RCDI, and control group mice. The results showed more serious clinical manifestations in the RCDI group compared with those in the CDI group. More severe gut barrier disruption and higher degree of microbiota translocation were observed in the RCDI group compared with those in the CDI group. Moreover, extremely severe apoptosis was observed in HCT-116 cells incubated with the serum from RCDI mice model. In addition, higher levels of Th17 and IL-17 were detected in the blood or serum from the RCDI mouse model. Treatment with RORγt small molecule inhibitor SR1001 increased the expression of occludin, decreased the apoptotic rate of HCT-116 cells, and decreased the concentrations of Th17 and IL-17. Concisely, Th17 and IL-17 are potential indicators of RCDI and may serve as therapeutic targets for RCDI treatment. This study lays the foundation for future research on RCDI diagnosis and treatment.


Clostridium Infections/drug therapy , Clostridium Infections/immunology , Th17 Cells/metabolism , Animals , Apoptosis , Cell Line, Tumor , Clostridium Infections/metabolism , Clostridium Infections/pathology , Colon/drug effects , Colon/pathology , Disease Models, Animal , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Janus Kinase 2/metabolism , Male , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Recurrence , STAT3 Transcription Factor/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Th17 Cells/immunology , Thiazoles/pharmacology , Thiazoles/therapeutic use
4.
Basic Clin Pharmacol Toxicol ; 129(5): 357-368, 2021 Nov.
Article En | MEDLINE | ID: mdl-34384137

The management of acute pancreatitis (AP) remains a challenge to clinicians worldwide for limited effective interventions. Retinoid orphan receptor gamma t (RORγt) is a therapeutic target for several diseases; however, it is unclear whether inhibiting RORγt can ameliorate AP. The relative expression of RORγt, IL-17 and IL-23 in the peripheral blood mononuclear cells of AP patients was measured by RT-PCR. An AP mouse model was induced by ceruletide, and SR1001 was injected before ceruletide administration. RORγt+ cells, T helper 17 cells (Th17), regulatory T cells (Tregs) and γδ T cells were assessed in the pancreas and spleen by flow cytometry. Higher RORγt expression in patients indicated the potential role of RORγt in AP progression. Analyses of the IL-17/IL-23 axis confirmed its role. SR1001 significantly alleviated AP histologically in the mouse model. Serum levels of amylase, IL-6, TNFalpha, IL-17 and IL-23 decreased upon SR1001 treatment. SR1001 selectively decreased the number of RORγt+, Th17, Tregs and γδ T cells in the pancreas but not the spleen. Collectively, these results showed that SR1001 exerted therapeutic effects on AP by suppressing IL-17-secreting Th17 and γδ T cells in the pancreas. Thus, SR1001 may be a promising drug for the treatment of AP in the clinic.


Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Pancreatitis/drug therapy , Sulfonamides/pharmacology , Thiazoles/pharmacology , Acute Disease , Adult , Aged , Animals , Case-Control Studies , Ceruletide , Disease Models, Animal , Disease Progression , Female , Humans , Interleukin-17/blood , Intraepithelial Lymphocytes/metabolism , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Pancreatitis/physiopathology , Th17 Cells/metabolism
5.
Immunol Res ; 69(4): 378-390, 2021 08.
Article En | MEDLINE | ID: mdl-34219199

Retinoic­acid­receptor­related orphan nuclear hormone receptor gamma t (RORγt), a critical transcriptional factor of Th17 cells, is a potential therapeutic target for Th17-mediated autoimmune diseases. In addition, RORγt is essential for thymocyte survival and lymph node development, and RORγt inhibition or deficiency causes abnormal thymocyte development, thymus lymphoma, and lymph node defect. Recent study demonstrated that specific regulation of Th17 differentiation related to the hinge region of RORγt. In this research, we investigated the effect of RORγt inhibitor, 5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidine derivative (TTP), in the therapy of lupus nephritis and its safety on thymocyte development. We demonstrated that TTP repressed the development of Th17 cells and ameliorated the autoimmune disease manifestation in the pristane-induced lupus nephritis mice model. The treatment of TTP in the mice did not interfere with thymocyte development, including total thymocyte number and proportion of CD4+CD8+ double-positive populations in the thymus, and had no substantial effects on the pathogenesis of thymoma. The TTP had a stronger affinity with full-length RORγt protein compared with the truncated RORγt LBD region via surface plasmon resonance, which indicated TTP binding to RORγt beyond LBD region. Molecular docking computation showed that the best binding pocket of TTP to RORγt is located in the hinge region of RORγt. In summary, as a RORγt inhibitor, TTP had a potential to develop the clinical medicine for treating Th17-mediated autoimmune diseases with low safety risk for thymocyte development.


Lupus Nephritis/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Pyrimidines/therapeutic use , Animals , Antibodies/blood , Apoptosis/drug effects , Cell Differentiation/drug effects , Cytokines/genetics , DNA/immunology , Female , Immunosuppressive Agents , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Lupus Nephritis/chemically induced , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Pyrimidines/pharmacology , Spleen/drug effects , Spleen/immunology , Terpenes , Th17 Cells/drug effects , Th17 Cells/immunology , Thymocytes/drug effects
6.
J Med Chem ; 64(12): 8775-8797, 2021 06 24.
Article En | MEDLINE | ID: mdl-34121397

Receptor-related orphan receptor γ (RORγ) has emerged as an attractive therapeutic target for the treatment of cancer and inflammatory diseases. Herein, we report our effort on the discovery, optimization, and evaluation of benzothiazole and benzimidazole derivatives as novel inverse agonists of RORγ. The representative compound 27h (designated as XY123) potently inhibited the RORγ transcription activity with a half-maximal inhibitory concentration (IC50) value of 64 nM and showed excellent selectivity against other nuclear receptors. 27h also potently suppressed cell proliferation, colony formation, and the expression of androgen receptor (AR)-regulated genes in AR-positive prostate cancer cell lines. In addition, 27h demonstrated good metabolic stability and a pharmacokinetic property with reasonable oral bioavailability (32.41%) and moderate half-life (t1/2 = 4.98 h). Significantly, oral administration of compound 27h achieved complete and long-lasting tumor regression in the 22Rv1 xenograft tumor model in mice. Compound 27h may serve as a new valuable lead compound for further development of drugs for the treatment of prostate cancer.


Antineoplastic Agents/therapeutic use , Benzeneacetamides/therapeutic use , Benzimidazoles/therapeutic use , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Benzeneacetamides/chemical synthesis , Benzeneacetamides/pharmacokinetics , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacokinetics , Benzothiazoles/chemical synthesis , Benzothiazoles/pharmacokinetics , Benzothiazoles/therapeutic use , Cell Proliferation/drug effects , Drug Inverse Agonism , Drug Stability , Male , Mice, Inbred NOD , Mice, SCID , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Rats, Sprague-Dawley , Structure-Activity Relationship , Xenograft Model Antitumor Assays
7.
Sci Rep ; 11(1): 11066, 2021 05 26.
Article En | MEDLINE | ID: mdl-34040108

The nuclear receptor retinoid-related orphan receptor gamma t (RORγt) plays a critical role in driving Th17 cell differentiation and expansion, as well as IL-17 production in innate and adaptive immune cells. The IL-23/IL-17 axis is implicated in several autoimmune and inflammatory diseases, and biologics targeting IL-23 and IL-17 have shown significant clinical efficacy in treating psoriasis and psoriatic arthritis. JNJ-61803534 is a potent RORγt inverse agonist, selectively inhibiting RORγt-driven transcription versus closely-related family members, RORα and RORß. JNJ-61803534 inhibited IL-17A production in human CD4+ T cells under Th17 differentiation conditions, but did not inhibit IFNγ production under Th1 differentiation conditions, and had no impact on in vitro differentiation of regulatory T cells (Treg), nor on the suppressive activity of natural Tregs. In the mouse collagen-induced arthritis model, JNJ-61803534 dose-dependently attenuated inflammation, achieving ~ 90% maximum inhibition of clinical score. JNJ-61803534 significantly inhibited disease score in the imiquimod-induced mouse skin inflammation model, and dose-dependently inhibited the expression of RORγt-regulated genes, including IL-17A, IL-17F, IL-22 and IL-23R. Preclinical 1-month toxicity studies in rats and dogs identified doses that were well tolerated supporting progression into first-in-human studies. An oral formulation of JNJ-61803534 was studied in a phase 1 randomized double-blind study in healthy human volunteers to assess safety, pharmacokinetics, and pharmacodynamics. The compound was well tolerated in single ascending doses (SAD) up to 200 mg, and exhibited dose-dependent increases in exposure upon oral dosing, with a plasma half-life of 164 to 170 h. In addition, dose-dependent inhibition of ex vivo stimulated IL-17A production in whole blood was observed, demonstrating in vivo target engagement. In conclusion, JNJ-61803534 is a potent and selective RORγt inhibitor that exhibited acceptable preclinical safety and efficacy, as well as an acceptable safety profile in a healthy volunteer SAD study, with clear evidence of a pharmacodynamic effect in humans.


Anti-Inflammatory Agents/pharmacology , CD4-Positive T-Lymphocytes/drug effects , Gene Expression Regulation/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/drug effects , Animals , Anti-Inflammatory Agents/therapeutic use , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Inflammation/drug therapy , Inflammation/metabolism , Interleukins/genetics , Interleukins/metabolism , Mice , Skin Diseases/drug therapy , Skin Diseases/metabolism , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/metabolism
8.
Respir Res ; 22(1): 158, 2021 May 22.
Article En | MEDLINE | ID: mdl-34022896

BACKGROUND: RORγt is a transcription factor that enables elaboration of Th17-associated cytokines (including IL-17 and IL-22) and is proposed as a pharmacological target for severe asthma. METHODS: IL-17 immunohistochemistry was performed in severe asthma bronchial biopsies (specificity confirmed with in situ hybridization). Primary human small airway epithelial cells in air liquid interface and primary bronchial smooth muscle cells were stimulated with recombinant human IL-17 and/or IL-22 and pro-inflammatory cytokines measured. Balb/c mice were challenged intratracheally with IL-17 and/or IL-22 and airway hyperreactivity, pro-inflammatory cytokines and airway neutrophilia measured. Balb/c mice were sensitized intraperitoneally and challenged intratracheally with house dust mite extract and the effect of either a RORγt inhibitor (BIX119) or an anti-IL-11 antibody assessed on airway hyperreactivity, pro-inflammatory cytokines and airway neutrophilia measured. RESULTS: We confirmed in severe asthma bronchial biopsies both the presence of IL-17-positive lymphocytes and that an IL-17 transcriptome profile in a severe asthma patient sub-population. Both IL-17 and IL-22 stimulated the release of pro-inflammatory cytokine and chemokine release from primary human lung cells and in mice. Furthermore, IL-22 in combination with IL-17, but neither alone, elicits airway hyperresponsiveness (AHR) in naïve mice. A RORγt inhibitor specifically blocked both IL-17 and IL-22, AHR and neutrophilia in a mouse house dust mite model unlike other registered or advanced pipeline modes of action. Full efficacy versus these parameters was associated with 90% inhibition of IL-17 and 50% inhibition of IL-22. In contrast, anti-IL-17 also blocked IL-17, but not IL-22, AHR or neutrophilia. Moreover, the deregulated genes in the lungs from these mice correlated well with deregulated genes from severe asthma biopsies suggesting that this model recapitulates significant severe asthma-relevant biology. Furthermore, these genes were reversed upon RORγt inhibition in the HDM model. Cell deconvolution suggested that the responsible cells were corticosteroid insensitive γδ-T-cells. CONCLUSION: These data strongly suggest that both IL-17 and IL-22 are required for Th2-low endotype associated biology and that a RORγt inhibitor may provide improved clinical benefit in a severe asthma sub-population of patients by blocking both IL-17 and IL-22 biology compared with blocking IL-17 alone.


Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Interleukin-17/metabolism , Interleukins/antagonists & inhibitors , Lung/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Th17 Cells/drug effects , Adolescent , Adult , Aged , Animals , Asthma/immunology , Asthma/metabolism , Asthma/physiopathology , Cells, Cultured , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Humans , Interleukins/metabolism , Lung/immunology , Lung/metabolism , Lung/physiopathology , Male , Mice, Inbred BALB C , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Pyroglyphidae/immunology , Signal Transduction , Th17 Cells/immunology , Th17 Cells/metabolism , Young Adult , Interleukin-22
9.
Sci Rep ; 11(1): 9132, 2021 04 28.
Article En | MEDLINE | ID: mdl-33911101

Clinical success of IL-17/IL-23 pathway biologics for the treatment of moderate to severe psoriasis suggests that targeting RORγt, a master regulator for the proliferation and function of Th17 cells, could be an effective alternative. However, oral RORγ antagonists (VTP43742, TAK828) with high systemic exposure showed toxicity in phase I/II clinical trials and terminated development. To alleviate the potential safety concerns, identifying compounds with skin-restricted exposure amenable for topical use is of great interest. Systematic structure activity relationship study and multi-parameter optimization led to the discovery of a novel RORγ antagonist (SHR168442) with desired properties for a topical drug. It suppressed the transcription of IL-17 gene, leading to reduction of IL-17 cytokine secretion. It showed high exposure in skin, but low in plasma. Topical application of SHR168442 in Vaseline exhibited excellent efficacy in the imiquimod-induced and IL-23-induced psoriasis-like skin inflammation mouse models and correlated with the reduction of Th17 pathway cytokines, IL-6, TNFα and IL-17A. This work demonstrated restricted skin exposure of RORγ antagonist may provide a new topical treatment option as targeted therapeutics for mild to moderate psoriasis patients and may be suitable for the treatment of any other inflammatory disorders that are accessible locally.


Benzimidazoles/therapeutic use , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Psoriasis/drug therapy , Administration, Topical , Animals , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Benzimidazoles/pharmacology , Binding Sites , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Stability , Humans , Imiquimod/toxicity , Interleukin-17/metabolism , Interleukin-23/metabolism , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Psoriasis/chemically induced , Psoriasis/pathology , Rats , Rats, Sprague-Dawley , Severity of Illness Index , Skin/drug effects , Skin/pathology
10.
J Med Chem ; 64(5): 2714-2724, 2021 03 11.
Article En | MEDLINE | ID: mdl-33591748

SAR efforts directed at identifying RORγt inverse agonists structurally different from our clinical compound 1 (BMS-986251) led to tricyclic-carbocyclic analogues represented by 3-7 and culminated in the identification of 3d (BMS-986313), with structural differences distinct from 1. The X-ray co-crystal structure of 3d with the ligand binding domain of RORγt revealed several key interactions, which are different from 1. The in vitro and in vivo PK profiles of 3d are described. In addition, we demonstrate robust efficacy of 3d in two preclinical models of psoriasis-the IMQ-induced skin lesion model and the IL-23-induced acanthosis model. The efficacy seen with 3d in these models is comparable to the results observed with 1.


Amides/therapeutic use , Hydrocarbons, Cyclic/therapeutic use , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Psoriasis/drug therapy , Amides/chemistry , Amides/pharmacokinetics , Animals , Drug Inverse Agonism , Female , Humans , Hydrocarbons, Cyclic/chemistry , Hydrocarbons, Cyclic/pharmacokinetics , Interleukin-23 , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Molecular Structure , Psoriasis/chemically induced , Rats , Structure-Activity Relationship
11.
Clin Exp Dermatol ; 46(1): 122-129, 2021 Jan.
Article En | MEDLINE | ID: mdl-32767679

BACKGROUND: Transcription factor retinoic acid-related orphan receptor 2 (RORC2/RORγT) mediates interleukin (IL)-17A and IL-17F expression. IL-17A plays a central role in the pathogenesis of several inflammatory disorders, including psoriasis. The RORC2 inhibitor PF-06763809 has been hypothesized to inhibit IL-17A production in T-helper 17 (Th17) cells, thereby reducing psoriasis symptoms. AIM: To assess the safety, tolerability and effect on skin infiltrate thickness of PF-06763809 in participants with mild/moderate chronic plaque psoriasis. METHODS: This was a randomized, double-blind, first-in-human study (trial registration: ClinicalTrials.gov NCT03469336). Participants received each of the following six treatments once daily for 18 days: three topical doses (2.3%, 0.8%, 0.23%) of PF-06763809, a vehicle and two active comparators (betamethasone and calcipotriol). Primary endpoints included change from baseline in psoriatic skin infiltrate thickness [echo-poor band (EPB) on ultrasonography] at Day 19, and safety. Change in psoriasis-associated gene expression (Day 19), evaluated by real-time reverse transcription PCR of skin biopsies, was an exploratory endpoint. RESULTS: In total, 17 participants completed the study. Change from baseline in the EPB on Day 19 for all three doses of PF-06763809 was not significantly different from that of vehicle (P > 0.05). A significant reduction in EPB from baseline was observed with betamethasone on Day 19 relative to all other treatments (P < 0.0001). Treatment-related adverse events were mild/moderate. There were no significant differences in gene expression on Day 19 between vehicle and PF-06763809-treated skin lesions. CONCLUSION: Using a psoriasis plaque test design, PF-06763809 was found to be well tolerated with an acceptable safety profile in participants with psoriasis, but without reduction in skin infiltrate thickness or disease biomarkers.


Boron Compounds/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Interleukin-17/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Psoriasis/drug therapy , Administration, Topical , Boron Compounds/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Double-Blind Method , Gene Expression , Humans , Interleukin-17/genetics , Interleukin-17/metabolism , Male , Middle Aged , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Organic Chemicals/adverse effects , Organic Chemicals/therapeutic use , Psoriasis/pathology , Real-Time Polymerase Chain Reaction , Skin/pathology , Treatment Failure
12.
Mol Diagn Ther ; 25(1): 77-85, 2021 01.
Article En | MEDLINE | ID: mdl-33314011

BACKGROUND: The field of antisense oligonucleotide therapeutics is rapidly growing and in addition to small molecules and therapeutic antibodies, oligonucleotide-based gene expression modifiers have been developed as fully accepted therapeutics. Antisense oligonucleotides are designed to modify gene expression of their specific target genes. However, as their effect relies on Watson-Crick base pairing, they could also bind to other unintended complementary RNAs showing sufficient sequence homology, which in turn could lead to off-target effects. It is assumed that these off-target effects depend on the degree of complementarity between the antisense oligonucleotides and off-target sequences. OBJECTIVE: Aim of this study was the investigation of the effects of antisense oligonucleotides on the expression of potential off-targets having a defined number of mismatches to the oligonucleotide sequence. METHODS: We extend recent studies by investigating the off-target profile of two 17-mer antisense oligonucleotides in two distinct human cell lines by a whole-transcriptome study using RNA sequencing. RESULTS: The relatively high percentage of significantly downregulated off-target genes for which one mismatch is present corroborates the requirement for intense bioinformatic screens and stringent specificity criteria to design antisense oligonucleotides with only minimal sequence complementarity to any non-target sequence. CONCLUSIONS: Avoiding suppression of off-target genes by a thorough bioinformatics screen should strongly reduce the risk for toxicities caused by antisense oligonucleotide-mediated off-target RNA suppression and finally result in safer antisense oligonucleotide-based therapeutics.


Gene Expression Profiling/methods , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Oligonucleotides, Antisense/genetics , Base Pairing , Base Sequence , Cell Line , Down-Regulation , Humans , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Oligonucleotides, Antisense/pharmacology , Sequence Analysis, RNA , Sequence Homology, Nucleic Acid
13.
Bioorg Med Chem Lett ; 30(23): 127521, 2020 12 01.
Article En | MEDLINE | ID: mdl-32882417

In order to rapidly develop C6 and C8 SAR of our reported tricyclic sulfone series of RORγt inverse agonists, a late-stage bromination was employed. Although not regioselective, the bromination protocol allowed us to explore new substitution patterns/vectors that otherwise would have to be incorporated at the very beginning of the synthesis. Based on the SAR obtained from this exercise, compound 15 bearing a C8 fluorine was developed as a very potent and selective RORγt inverse agonist. This analog's in vitro profile, pharmacokinetic (PK) data and efficacy in an IL-23 induced mouse acanthosis model will be discussed.


Heterocyclic Compounds, 3-Ring/therapeutic use , Melanosis/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Sulfones/therapeutic use , Animals , Crystallography, X-Ray , Drug Inverse Agonism , Female , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Interleukin-18 , Male , Melanosis/chemically induced , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Structure , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Protein Binding , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/pharmacokinetics
14.
Bioorg Med Chem Lett ; 30(19): 127441, 2020 10 01.
Article En | MEDLINE | ID: mdl-32736080

In an effort to discover oral inverse agonists of RORγt to treat inflammatory diseases, a new 2,6-difluorobenzyl ether series of cyclopentyl sulfones were found to be surprisingly more potent than the corresponding alcohol derivatives. When combined with a more optimized phenyl ((R)-3-phenylpyrrolidin-3-yl)sulfone template, the 2,6-difluorobenzyl ethers yielded a set of very potent RORγt inverse agonists (e.g., compound 26, RORγt Gal4 EC50 11 nM) that are highly selective against PXR, LXRα and LXRß. After optimizing for stability in human and mouse liver microsomes, compounds 29 and 38 were evaluated in vivo and found to have good oral bioavailability (56% and 101%, respectively) in mice. X-ray co-crystal structure of compound 27 in RORγt revealed that the bulky benzyl ether group causes helix 11 of the protein to partially uncoil to create a new, enlarged binding site, which nicely accommodates the benzyl ether moiety, leading to net potency gain.


Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Pyrrolidines/pharmacology , Sulfones/pharmacology , Animals , Crystallography, X-Ray , Drug Discovery , Drug Inverse Agonism , Drug Stability , Hep G2 Cells , Humans , Mice , Microsomes, Liver/metabolism , Molecular Structure , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Pyrrolidines/chemical synthesis , Pyrrolidines/metabolism , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/metabolism
15.
Bioorg Med Chem Lett ; 30(19): 127466, 2020 10 01.
Article En | MEDLINE | ID: mdl-32763309

RORγt is the master regulator of the IL-23/IL-17 axis, a pathway that is clinically validated for the treatment of various immunological disorders. Over the last few years, our group has reported different chemotypes that potently act as inverse agonists of RORγt. One of them, the tricyclic pyrrolidine chemotype, has demonstrated biologic-like preclinical efficacy and has led to our clinical candidate BMS-986251. In this letter, we discuss the invention of an annulation reaction which enabled the synthesis of a tricyclic exocyclic amide chemotype and the identification of compounds with RORγt inverse agonist activity. Preliminary structure activity relationships are disclosed.


Amides/chemistry , Hydrocarbons, Cyclic/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Sulfones/chemistry , Amides/chemical synthesis , Amides/metabolism , Animals , Cyclization , Drug Inverse Agonism , Humans , Hydrocarbons, Cyclic/chemical synthesis , Hydrocarbons, Cyclic/metabolism , Mice , Microsomes, Liver/metabolism , Molecular Docking Simulation , Molecular Structure , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/metabolism
16.
Food Funct ; 11(8): 6875-6888, 2020 Aug 01.
Article En | MEDLINE | ID: mdl-32686813

T helper 17 (Th17) cells that express interleukin-17 (IL-17) play a key role in various inflammatory diseases, such as multiple sclerosis (MS), and its animal model experimental autoimmune encephalomyelitis (EAE). The retinoic acid receptor-related orphan receptors γt (RORγt) have an indispensable effect on the differentiation of this cell type, and are thus considered a valuable target in the treatment of Th17-related disorders. In this study, we found that eriodictyol (EDT), a natural flavonoid abundant in citrus fruits and peanuts, was located directly in the binding pocket of RORγt, and induced a conformational change that resulted in the effective suppression of the receptor's activity, thus offering insight into the transcriptional inhibition of RORγt-dependent genes. Consistent with this, EDT dose-dependently (5-10 µM) blocked murine Th17 differentiation, and markedly reduced IL-17A secretion in vitro. Furthermore, this compound has been found to have novel properties for directly inhibiting Th1 cell development and promoting Treg cell differentiation at high doses (≥10 µM). EDT administration significantly decreased the clinical severity in the EAE model, with inhibited demyelination and reduced inflammatory responses in the periphery and in the central nervous system (CNS). In the adoptive transfer model, EDT also remarkably suppressed the Th17 cell infiltration and pathogenicity. Collectively, our data demonstrated that EDT, as an agent for the pharmacological inhibition of RORγt, has great potential for immunomodulation, and for use in the treatment of Th17-mediated autoimmune disease.


Cell Differentiation/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Flavanones/pharmacology , Interleukin-17/metabolism , Th17 Cells/immunology , Adoptive Transfer , Animals , Cell Differentiation/immunology , Dose-Response Relationship, Immunologic , Encephalomyelitis, Autoimmune, Experimental/immunology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Th17 Cells/cytology
17.
Eur J Immunol ; 50(9): 1400-1411, 2020 09.
Article En | MEDLINE | ID: mdl-32388860

Inflammatory bowel diseases (IBD) are chronic conditions that result from uncontrolled intestinal inflammation. Pathogenic Th17 cells, characterized by production of IL-17A in the absence of IL-10, are thought to contribute to this inflammation, but in humans, antibody-mediated blockade of IL-17A is an ineffective IBD therapy whereas IL-23 blockade is effective. Here, we investigated the effects of pharmacological inhibition of RORC2, the Th17 cell lineage-defining transcription factor, on in vivo-differentiated human Th17 cells and Th17-like Tregs (Th17-Tregs). BMS-336, a small molecule RORC2 inverse agonist, inhibited expression of RORC2-regulated genes in peripheral Th17 cells (CD4+ CD25- CD127+ CXCR3- CCR4+ CCR6+ ) in a dose-dependent manner, with similar inhibitory effects on laminar propria mononuclear cells from IBD and non-IBD subjects. Exposure of peripheral Th17-Tregs (CD4+ CD25hi CD127lo CXCR3- CCR4+ CCR6+ ) to BMS-336 also inhibited IL-17A production and prevented inflammatory cytokine-induced destabilization, as evidenced by preserved FOXP3 expression and epigenetic status of the Treg-specific demethylation region. In parallel, RORC2 inhibition increased the production of IL-10 in Th17-Tregs, resulting in enhanced suppression of inflammatory cytokines from myeloid cells. Thus, via its ability to simultaneously inhibit Th17 cells and enhance the stability and function of Th17-Tregs, pharmacological inhibition of RORC2 is a promising approach to suppress inflammation and promote immune regulation in IBD.


Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Adult , Cell Differentiation/drug effects , Female , Humans , Inflammatory Bowel Diseases , Male , Middle Aged
18.
Int J Mol Sci ; 21(10)2020 May 17.
Article En | MEDLINE | ID: mdl-32429598

Diabetic retinopathy is a diabetes-mediated retinal microvascular disease that is the leading cause of blindness in the working-age population worldwide. Interleukin (IL)-17A is an inflammatory cytokine that has been previously shown to play a pivotal role in the promotion and progression of diabetic retinopathy. Retinoic acid-related orphan receptor gammaT (RORγt) is a ligand-dependent transcription factor that mediates IL-17A production. However, the role of RORγt in diabetes-mediated retinal inflammation and capillary degeneration, as well as its potential therapeutic attributes for diabetic retinopathy has not yet been determined. In the current study, we examined retinal inflammation and vascular pathology in streptozotocin-induced diabetic mice. We found RORγt expressing cells in the retinal vasculature of diabetic mice. Further, diabetes-mediated retinal inflammation, oxidative stress, and retinal endothelial cell death were all significantly lower in RORγt-/- mice. Finally, when a RORγt small molecule inhibitor (SR1001) was subcutaneously injected into diabetic mice, retinal inflammation and capillary degeneration were ameliorated. These findings establish a pathologic role for RORγt in the onset of diabetic retinopathy and identify a potentially novel therapeutic for this blinding disease.


Capillaries/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Interleukin-17/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Retinal Vessels/metabolism , Sulfonamides/pharmacology , Thiazoles/pharmacology , Animals , Capillaries/pathology , Cell Death/genetics , Cell Survival/drug effects , Cell Survival/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetic Retinopathy/chemically induced , Diabetic Retinopathy/drug therapy , Drug Inverse Agonism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hyperglycemia/blood , Hyperglycemia/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Oxidative Stress/genetics , Retinal Vessels/drug effects , Retinal Vessels/pathology , Sulfonamides/therapeutic use , Thiazoles/therapeutic use
19.
Nature ; 580(7805): 621-627, 2020 04.
Article En | MEDLINE | ID: mdl-32179876

Frequently referred to as the 'magic methyl effect', the installation of methyl groups-especially adjacent (α) to heteroatoms-has been shown to dramatically increase the potency of biologically active molecules1-3. However, existing methylation methods show limited scope and have not been demonstrated in complex settings1. Here we report a regioselective and chemoselective oxidative C(sp3)-H methylation method that is compatible with late-stage functionalization of drug scaffolds and natural products. This combines a highly site-selective and chemoselective C-H hydroxylation with a mild, functional-group-tolerant methylation. Using a small-molecule manganese catalyst, Mn(CF3PDP), at low loading (at a substrate/catalyst ratio of 200) affords targeted C-H hydroxylation on heterocyclic cores, while preserving electron-neutral and electron-rich aryls. Fluorine- or Lewis-acid-assisted formation of reactive iminium or oxonium intermediates enables the use of a mildly nucleophilic organoaluminium methylating reagent that preserves other electrophilic functionalities on the substrate. We show this late-stage C(sp3)-H methylation on 41 substrates housing 16 different medicinally important cores that include electron-rich aryls, heterocycles, carbonyls and amines. Eighteen pharmacologically relevant molecules with competing sites-including drugs (for example, tedizolid) and natural products-are methylated site-selectively at the most electron rich, least sterically hindered position. We demonstrate the syntheses of two magic methyl substrates-an inverse agonist for the nuclear receptor RORc and an antagonist of the sphingosine-1-phosphate receptor-1-via late-stage methylation from the drug or its advanced precursor. We also show a remote methylation of the B-ring carbocycle of an abiraterone analogue. The ability to methylate such complex molecules at late stages will reduce synthetic effort and thereby expedite broader exploration of the magic methyl effect in pursuit of new small-molecule therapeutics and chemical probes.


Biological Products/chemistry , Biological Products/chemical synthesis , Carbon/chemistry , Chemistry Techniques, Synthetic , Hydrogen/chemistry , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/chemical synthesis , Androstenes/chemical synthesis , Androstenes/chemistry , Catalysis , Drug Inverse Agonism , Electrons , Fluorine/chemistry , Hydroxylation , Lewis Acids/chemistry , Manganese/chemistry , Methylation , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Oxazolidinones/chemical synthesis , Oxazolidinones/chemistry , Oxidation-Reduction , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Tetrazoles/chemical synthesis , Tetrazoles/chemistry
20.
Inflammation ; 43(3): 962-974, 2020 Jun.
Article En | MEDLINE | ID: mdl-32170603

Esculetin, a coumarin derivative from various natural plants, has an anti-inflammatory property. In the present study, we examined if esculetin has any salutary effects against lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. Acute lung injury (ALI) was induced via the intratracheal administration of LPS, and esculetin (20 and 40 mg/kg) was given intraperitoneally 30 min before LPS challenge. After 6 h of LPS administration, lung tissues were collected for analysis. Pretreatment with esculetin significantly attenuated histopathological changes, inflammatory cell infiltration, and production of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1ß, and IL-6, in the lung tissue. Furthermore, esculetin inhibited the protein kinase B (AKT), extracellular signal-regulated kinase (ERK), and nuclear factor-kappa B (NF-κB) pathways and downregulated the expression of RORγt and IL-17 in LPS-induced ALI. Our results indicated that esculetin possesses anti-inflammatory and protective effects against LPS-induced ALI via inhibition of the AKT/ERK/NF-κB and RORγt/IL-17 pathways.


Acute Lung Injury/drug therapy , Interleukin-17/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , NF-kappa B/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Umbelliferones/therapeutic use , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Dose-Response Relationship, Drug , Interleukin-17/metabolism , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Umbelliferones/pharmacology
...