Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Haematologica ; 105(9): 2286-2297, 2020 09 01.
Article in English | MEDLINE | ID: mdl-33054053

ABSTRACT

Differentiation therapies achieve remarkable success in acute promyelocytic leukemia, a subtype of acute myeloid leukemia. However, excluding acute promyelocytic leukemia, clinical benefits of differentiation therapies are negligible in acute myeloid leukemia except for mutant isocitrate dehydrogenase 1/2. Dihydroorotate dehydrogenase catalyses the fourth step of the de novo pyrimidine synthesis pathway. ASLAN003 is a highly potent dihydroorotate dehydrogenase inhibitor that induces differentiation, as well as reduces cell proliferation and viability, of acute myeloid leukemia cell lines and primary acute myeloid leukemia blasts including in chemo-resistant cells. Apoptotic pathways are triggered by ASLAN003, and it also significantly inhibits protein synthesis and activates AP-1 transcription, contributing to its differentiation promoting capacity. Finally, ASLAN003 substantially reduces leukemic burden and prolongs survival in acute myeloid leukemia xenograft mice and acute myeloid leukemia patient-derived xenograft models. Notably, the drug has no evident effect on normal hematopoietic cells and exhibits excellent safety profiles in mice, even after a prolonged period of administration. Our results, therefore, suggest that ASLAN003 is an agent targeting dihydroorotate dehydrogenase with potential in the treatment of acute myeloid leukemia. ASLAN003 is currently being evaluated in phase 2a clinical trial in acute myeloid leukemia patients.


Subject(s)
Leukemia, Myeloid, Acute , Oxidoreductases Acting on CH-CH Group Donors , Animals , Cell Differentiation , Dihydroorotate Dehydrogenase , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mice , Oxidoreductases Acting on CH-CH Group Donors/genetics
2.
Invest New Drugs ; 36(5): 886-894, 2018 10.
Article in English | MEDLINE | ID: mdl-29766337

ABSTRACT

Background The MET tyrosine kinase and its ligand, hepatocyte growth factor (HGF) also known as scatter factor, are associated with tumourigenesis and metastasis by promotion of scattering, proliferation, angiogenesis, motility and invasion. ASLAN-002 is a potent inhibitor of MET as well as related kinases. A phase I dose escalation study was conducted to determine the safety and pharmacokinetics of ASLAN-002 in patients with advanced cancer. Methods Patients with advanced or metastatic solid tumours, who had progressed on standard therapy or for whom standard therapy was not known, were administered ASLAN-002 orally. The starting dose was 100 mg once daily (QD) with subsequent cohorts to receive doses of 200 mg QD, 300 mg QD, 450 mg QD, 600 mg QD, 300 mg twice daily (BID), 450 mg BID, and 600 mg BID. Results Forty patients were included across 7 dose cohorts. Cohort 8 (600 mg BID) was not opened due to the lack of appreciable pharmacokinetic (PK) differences between 300 mg BID and 450 mg BID and higher incidences of grade 3 or 4 adverse events (AE) in Cohort 7 (450 mg BID). Fifteen patients (37.5%) experienced a grade 3 or 4 AE. The most commonly reported AEs were nausea (55%), fatigue (47.5%) and constipation (30%). One dose limiting toxicity (DLT) of atrial fibrillation was observed with 450 mg BID. Conclusions ASLAN-002 is well tolerated at 300 mg BID and is the recommended dose for future phase II studies (RP2D). Clinical Trials Registry Number: NCT01721148 .


Subject(s)
Neoplasms/drug therapy , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Administration, Oral , Adult , Aged , Aged, 80 and over , Atrial Fibrillation/drug therapy , Atrial Fibrillation/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Fatigue/chemically induced , Fatigue/metabolism , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Nausea/chemically induced , Nausea/metabolism , Neoplasms/metabolism
3.
Clin Pharmacol Ther ; 104(2): 335-345, 2018 08.
Article in English | MEDLINE | ID: mdl-29134633

ABSTRACT

Current asthma treatments address symptoms rather than the underlying disease pathophysiology, a better understanding of which has led to the identification of the Th2 high endotype. The activation of Toll-like receptors to induce Type I interferons directly in the lungs represents a novel therapeutic approach to reset this underlying Th2 pathophysiology with the potential to provide long-term disease modification. We present the nonclinical data and phase I clinical profile of an inhaled TLR9 agonist, AZD1419, a C-type CpG designed to induce interferon in the lung. In healthy volunteers, AZD1419 was found to be safe and well-tolerated. Target engagement in the lung was demonstrated at all dose levels tested. No evidence of tolerization or amplification of responses was evident on repeated dosing and 15.4 mg was defined as the maximum tolerated dose. AZD1419 clinical data supports its continued development as a potentially disease-modifying therapeutic in asthma.


Subject(s)
Anti-Asthmatic Agents/administration & dosage , Interferon Type I/metabolism , Lung/drug effects , Oligonucleotides/administration & dosage , Th2 Cells/drug effects , Toll-Like Receptor 9/agonists , Administration, Inhalation , Adolescent , Adult , Animals , Anti-Asthmatic Agents/adverse effects , Anti-Asthmatic Agents/pharmacokinetics , Cells, Cultured , Dose-Response Relationship, Drug , Double-Blind Method , Female , Healthy Volunteers , Humans , Lung/immunology , Lung/metabolism , Macaca fascicularis , Male , Maximum Tolerated Dose , Mice , Middle Aged , Oligonucleotides/adverse effects , Oligonucleotides/pharmacokinetics , Risk Assessment , Th2 Cells/immunology , Th2 Cells/metabolism , Toll-Like Receptor 9/metabolism , Up-Regulation , Young Adult
4.
ACS Med Chem Lett ; 8(9): 981-986, 2017 Sep 14.
Article in English | MEDLINE | ID: mdl-28947948

ABSTRACT

N-(5-Bromo-3-methoxypyrazin-2-yl)-5-chlorothiophene-2-sulfonamide 1 was identified as a hit in a CCR4 receptor antagonist high-throughput screen (HTS) of a subset of the AstraZeneca compound bank. As a hit with a lead-like profile, it was an excellent starting point for a CCR4 receptor antagonist program and enabled the rapid progression through the Lead Identification and Lead Optimization phases resulting in the discovery of two bioavailable CCR4 receptor antagonist candidate drugs.

5.
Sci Transl Med ; 9(374)2017 01 25.
Article in English | MEDLINE | ID: mdl-28123075

ABSTRACT

Bone destruction occurs in aging and numerous diseases, including osteoporosis and cancer. Many cancer patients have bone osteolysis that is refractory to state-of-the-art treatments, which block osteoclast activity with bisphosphonates or by inhibiting the receptor activator of nuclear factor κB ligand (RANKL) pathway. We previously showed that macrophage-stimulating protein (MSP) signaling, which is elevated in about 40% of breast cancers, promotes osteolytic bone metastasis by activation of the MSP signaling pathway in tumor cells or in the bone microenvironment. We show that MSP signals through its receptor, RON tyrosine kinase, expressed on host cells, to activate osteoclasts directly by a previously undescribed pathway that is complementary to RANKL signaling and converges on proto-oncogene, non-receptor tyrosine kinase SRC (SRC). Genetic or pharmacologic inhibition of RON kinase blocked cancer-mediated bone destruction and osteoporosis in several mouse models. Furthermore, the RON kinase inhibitor BMS-777607/ASLAN002 altered markers of bone turnover in a first-in-human clinical cancer study, indicating the inhibitor's potential for normalizing bone loss in patients. These findings uncover a new therapeutic target for pathogenic bone loss and provide a rationale for treatment of bone destruction in various diseases with RON inhibitors.


Subject(s)
Bone and Bones/pathology , Neoplasms/pathology , Osteoporosis/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Aminopyridines/pharmacology , Animals , Estrogens/metabolism , Female , Humans , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Transplantation , Osteoblasts/metabolism , Osteoclasts/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Pyridones/pharmacology , Signal Transduction , Treatment Outcome
6.
Thorax ; 69(3): 240-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24127021

ABSTRACT

BACKGROUND: Defective rhinovirus (RV)-induced interferon (IFN)-ß and IFN-λ production and increased RV replication have been reported in primary human bronchial epithelial cells (HBECs) from subjects with asthma. How universal this defect is in asthma is unknown. Additionally, the IFN subtypes induced by RV infection in primary HBECs have not been comprehensively investigated. OBJECTIVE: To study RV induction of IFN-α, IFN-ß and IFN-λ and RV replication in HBECs from subjects with atopic asthma and healthy controls. METHODS: HBECs were obtained from subjects with asthma and healthy controls and infected with RV16 and RV1B, and cells and supernatants harvested at 8, 24 and 48h. IFN proteins were analysed by ELISA and IFN mRNA and viral RNA expression by quantitative PCR. Virus release was assessed in cell supernatants. RESULTS: IFN-ß and IFN-λ were the only IFNs induced by RV in HBECs and IFN-λ protein induction was substantially greater than IFN-ß. Induction of IFN-λ1 mRNA by RV16 at 48h was significantly greater in HBECs from subjects with asthma; otherwise there were no significant differences between subjects with asthma and controls in RV replication, or in induction of type I or III IFN protein or mRNA. CONCLUSIONS: IFN-λ and, to a lesser degree, IFN-ß are the major IFN subtypes induced by RV infection of HBECs. Neither defective IFN induction by RV nor increased RV replication was observed in the HBECs from subjects with well controlled asthma reported in this study.


Subject(s)
Antiviral Agents/therapeutic use , Asthma/immunology , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Rhinovirus/immunology , Adult , Asthma/metabolism , Asthma/virology , Bronchoalveolar Lavage , Case-Control Studies , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Humans , Male , Signal Transduction , Time Factors , Virus Replication/drug effects
7.
PLoS One ; 8(6): e65921, 2013.
Article in English | MEDLINE | ID: mdl-23824215

ABSTRACT

Defective Rhinovirus induced interferon-ß and interferon-λ production has been reported in bronchial epithelial cells from asthmatics but the mechanisms of defective interferon induction in asthma are unknown. Virus infection can induce interferon through Toll like Receptors (TLR)3, TLR7 and TLR8. The role of these TLRs in interferon induction in asthma is unclear. This objective of this study was to measure the type I and III interferon response to TLR in bronchial epithelial cells and peripheral blood cells from atopic asthmatics and non-atopic non-asthmatics. Bronchial epithelial cells and peripheral blood mononuclear cells from atopic asthmatic and non-atopic non-asthmatic subjects were stimulated with agonists to TLR3, TLR4 & TLRs7-9 and type I and III interferon and pro-inflammatory cytokine, interleukin(IL)-6 and IL-8, responses assessed. mRNA expression was analysed by qPCR. Interferon proteins were analysed by ELISA. Pro-inflammatory cytokines were induced by each TLR ligand in both cell types. Ligands to TLR3 and TLR7/8, but not other TLRs, induced interferon-ß and interferon-λ in bronchial epithelial cells. The ligand to TLR7/8, but not those to other TLRs, induced only type I interferons in peripheral blood mononuclear cells. No difference was observed in TLR induced interferon or pro-inflammatory cytokine production between asthmatic and non-asthmatic subjects from either cell type. TLR3 and TLR7/8,, stimulation induced interferon in bronchial epithelial cells and peripheral blood mononuclear cells. Interferon induction to TLR agonists was not observed to be different in asthmatics and non-asthmatics.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/metabolism , Interferons/biosynthesis , Toll-Like Receptors/metabolism , Trachea/metabolism , Asthma/blood , Asthma/drug therapy , Case-Control Studies , Humans
8.
J Allergy Clin Immunol ; 129(6): 1506-1514.e6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22657407

ABSTRACT

BACKGROUND: Asthmatic patients have defective rhinovirus-induced IFN-ß and IFN-λ production from bronchial epithelial cells and IFN-λ from bronchoalveolar lavage (BAL) cells. Whether bronchoalveolar lavage cells have defective type I interferon responses to rhinovirus is unknown, as are mechanisms explaining defective rhinovirus interferon induction in asthmatic patients. OBJECTIVE: We sought to investigate rhinovirus induction of type I interferons in BAL and blood mononuclear cells from asthmatic patients and healthy subjects and to investigate mechanisms of any deficiency observed. METHODS: BAL and blood mononuclear cells from atopic asthmatic patients and healthy subjects were infected with rhinovirus ex vivo. Interferon proteins were analyzed by using ELISA. mRNA expression of key components of interferon induction pathways were analyzed by using quantitative PCR. RESULTS: Rhinovirus induction of type I interferon protein was delayed and deficient in BAL cells from asthmatic patients, and lower interferon levels were associated with greater airway hyperresponsiveness and skin prick test response positivity. Expression of Toll-like receptor (TLR) 3, TLR7, TLR8, retinoic acid-inducible gene I (RIG-I), melanoma differentiation-associated gene 5 (MDA-5), TIR domain-containing adapter-inducing IFN-ß (TRIF), myeloid differentiation primary response gene 88 (MyD88), caspase recruitment domain adaptor inducing IFN-ß (CARDIF), IL-1 receptor-associated kinase 4 (IRAK4), IκB kinase ß (IKKB), IκB kinase ι (IKKI), interferon regulatory factors 3 and 7, and rhinovirus induction of expression of the virus-inducible molecules TLR3, TLR7, RIG-I, and MDA-5 were not impaired in these interferon-deficient BAL cells in asthmatic patients. Defective rhinovirus interferon induction was not observed in blood mononuclear cells. CONCLUSIONS: Rhinovirus induction of type I interferons in BAL cells is delayed and deficient and might be a marker of more severe asthma. Defective rhinovirus interferon induction in asthmatic patients was not accompanied by differences in the expression or induction of key molecules implicated in viral induction of interferons.


Subject(s)
Asthma/immunology , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , Rhinovirus/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Asthma/metabolism , Asthma/virology , Bronchoalveolar Lavage Fluid , Case-Control Studies , Epithelial Cells/metabolism , Epithelial Cells/virology , Female , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Male , RNA Helicases/metabolism , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/metabolism , Signal Transduction , Skin Tests , Time Factors , Toll-Like Receptor 3/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
J Clin Invest ; 119(9): 2564-76, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19726873

ABSTRACT

CpG-containing immunostimulatory DNA sequences (ISS), which signal through TLR9, are being developed as a therapy for allergic indications and have proven to be safe and well tolerated in humans when administrated via the pulmonary route. In contrast, ISS inhalation has unexplained toxicity in rodents, which express TLR9 in monocyte/macrophage lineage cells as well as in plasmacytoid DCs (pDCs) and B cells, the principal TLR9-expressing cells in humans. We therefore investigated the mechanisms underlying this rodent-specific toxicity and its implications for humans. Mice responded to intranasally administered 1018 ISS, a representative B class ISS, with strictly TLR9-dependent toxicity, including lung inflammation and weight loss, that was fully reversible and pDC and B cell independent. Knockout mouse experiments demonstrated that ISS-induced toxicity was critically dependent on TNF-alpha, with IFN-alpha required for TNF-alpha induction. In contrast, human PBMCs, human alveolar macrophages, and airway-derived cells from Ascaris suum-allergic cynomolgus monkeys did not produce appreciable TNF-alpha in vitro in response to ISS stimulation. Moreover, sputum of allergic humans exposed to inhaled ISS demonstrated induction of IFN-inducible genes but minimal TNF-alpha induction. These data demonstrate that ISS induce rodent-specific TNF-alpha-dependent toxicity that is absent in humans and reflective of differential TLR9 expression patterns in rodents versus humans.


Subject(s)
Oligodeoxyribonucleotides/toxicity , Tumor Necrosis Factor-alpha/metabolism , Adjuvants, Immunologic/toxicity , Administration, Inhalation , Animals , Asthma/genetics , Asthma/immunology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , In Vitro Techniques , Lung/drug effects , Lung/immunology , Lung/pathology , Macaca fascicularis , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/immunology , Species Specificity , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
10.
Altern Lab Anim ; 34(3): 295-303, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16831061

ABSTRACT

A method for the routine, rapid and simultaneous cloning of drug targets from multiple mammalian species is described. This expedites the generation of recombinant proteins and cell lines that can provide alternatives to animal experiments. This was achieved by the collection of RNA from a comprehensive range of tissues from a variety of species, and the optimisation of cDNA synthesis. This "zooplate" has been successfully used for the simultaneous amplification and cloning of drug targets from multiple species. These products have subsequently been used to develop in vitro assays that support efficacy and safety studies in new drug discovery programmes. Within the framework of the Three Rs, these reagents can reduce the number of animals required to provide material for ex vivo assays and can refine the in vivo studies that are still necessary.


Subject(s)
Animal Testing Alternatives/methods , Cloning, Molecular/methods , Drug Evaluation, Preclinical/methods , Recombinant Proteins/biosynthesis , Animals , Humans , RNA/chemistry , RNA/genetics , Receptors, CXCR3 , Receptors, Chemokine/chemistry , Receptors, Chemokine/genetics , Recombinant Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
J Invest Dermatol ; 123(5): 892-901, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15482477

ABSTRACT

In this study, the phenotype of psoriatic keratinocytes and fibroblasts in reconstructed skin models was compared to those constructed from normal cells. Characterization of this model by immunohistochemistry showed that classical markers of keratinocyte differentiation exhibited similar patterns of distribution in the psoriatic models to those derived from normal cells and generally reflected in vivo observations. Some crucial differences, however, were observed between normal and psoriatic models when pro-inflammatory gene expression and keratinocyte proliferation were investigated. Notably, the chemokine receptor CXCR2 was overexpressed in the psoriatic models, and, moreover, was localized to the granular layer of keratinocytes as seen in psoriasis in vivo. Pro-inflammatory genes (tumor necrosis factor alpha [TNF-alpha], interferon gamma [IFN-gamma], and interleukin 8 [IL-8]) were expressed at high levels in the psoriatic models, but were only minimally expressed in the normal models. Models derived from uninvolved psoriatic skin showed the same gene expression profile as those derived from involved skin along with an increased proliferation rate when compared to normal models. These results suggest that psoriatic individuals possess an inherent predisposition to develop the disease phenotype even in the absence of T cells. This study represents a comprehensive characterization of psoriatic human skin reconstructed in vitro, and demonstrates the potential of this model as a valuable tool in drug discovery.


Subject(s)
Keratinocytes/cytology , Organ Culture Techniques/methods , Psoriasis/pathology , Psoriasis/physiopathology , Adult , Aged , Antibody Specificity , Cells, Cultured , Chemokine CXCL1 , Chemokines, CXC/genetics , Chemokines, CXC/immunology , Female , Gene Expression/immunology , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-8/genetics , Interleukin-8/immunology , Keratinocytes/physiology , Male , Middle Aged , Phenotype , Psoriasis/immunology , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/immunology , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...