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










Publication year range
1.
ACS Med Chem Lett ; 15(5): 714-721, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38746903

ABSTRACT

We herein report the discovery, synthesis, and evolution of a series of indazoles and azaindazoles as CNS-penetrant IRAK4 inhibitors. Described is the use of structure-based and property-based drug design strategically leveraged to guide the property profile of a key series into a favorable property space while maintaining potency and selectivity. Our rationale that led toward functionalities with potency improvements, CNS-penetration, solubility, and favorable drug-like properties is portrayed. In vivo evaluation of an advanced analogue showed significant, dose-dependent modulation of inflammatory cytokines in a mouse model. In pursuit of incorporating a highly engineered bridged ether that was crucial to metabolic stability in this series, significant synthetic challenges were overcome to enable the preparation of the analogues.

2.
J Med Chem ; 67(10): 8383-8395, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38695469

ABSTRACT

Interleukin receptor associated kinase 4 (IRAK4) plays an important role in innate immune signaling through Toll-like and interleukin-1 receptors and represents an attractive target for the treatment of inflammatory diseases and cancer. We previously reported the development of a potent, selective, and brain-penetrant imidazopyrimidine series of IRAK4 inhibitors. However, lead molecule BIO-7488 (1) suffered from low solubility which led to variable PK, compound accumulation, and poor in vivo tolerability. Herein, we describe the discovery of a series of pyridone analogs with improved solubility which are highly potent, selective and demonstrate desirable PK profiles including good oral bioavailability and excellent brain penetration. BIO-8169 (2) reduced the in vivo production of pro-inflammatory cytokines, was well tolerated in safety studies in rodents and dog at margins well above the predicted efficacious exposure and showed promising results in a mouse model for multiple sclerosis.


Subject(s)
Brain , Interleukin-1 Receptor-Associated Kinases , Protein Kinase Inhibitors , Animals , Dogs , Male , Mice , Rats , Brain/metabolism , Brain/drug effects , Drug Discovery , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/chemical synthesis , Pyrimidines/therapeutic use , Structure-Activity Relationship
3.
J Med Chem ; 67(6): 4676-4690, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38467640

ABSTRACT

Interleukin receptor-associated kinase 4 (IRAK4) is a key node of signaling within the innate immune system that regulates the production of inflammatory cytokines and chemokines. The presence of damage-associated molecular patterns (DAMPs) after tissue damage such as stroke or traumatic brain injury (TBI) initiates signaling through the IRAK4 pathway that can lead to a feed-forward inflammatory loop that can ultimately hinder patient recovery. Herein, we describe the first potent, selective, and CNS-penetrant IRAK4 inhibitors for the treatment of neuroinflammation. Lead compounds from the series were evaluated in CNS PK/PD models of inflammation, as well as a mouse model of ischemic stroke. The SAR optimization detailed within culminates in the discovery of BIO-7488, a highly selective and potent IRAK4 inhibitor that is CNS penetrant and has excellent ADME properties.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Ischemic Stroke , Mice , Animals , Humans , Signal Transduction , Cytokines , Pyrimidines/pharmacology , Pyrimidines/therapeutic use
4.
Toxicol Pathol ; 52(1): 35-54, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38385340

ABSTRACT

Recombinant adeno-associated virus (AAV)-mediated degeneration of sensory neurons in the dorsal root ganglia (DRG) and trigeminal ganglia (TG) has been observed in non-human primates (NHPs) following intravenous (IV) and intrathecal (IT) delivery. Administration of recombinant AAV encoding a human protein transgene via a single intra-cisterna magna (ICM) injection in New Zealand white rabbits resulted in histopathology changes very similar to NHPs: mononuclear cell infiltration, degeneration/necrosis of sensory neurons, and nerve fiber degeneration of sensory tracts in the spinal cord and of multiple nerves. AAV-associated clinical signs and incidence/severity of histologic findings indicated that rabbits were equally or more sensitive than NHPs to sensory neuron damage. Another study using human and rabbit transgene constructs of the same protein demonstrated comparable changes suggesting that the effects are not an immune response to the non-self protein transgene. Rabbit has not been characterized as a species for general toxicity testing of AAV gene therapies, but these studies suggest that it may be an alternative model to investigate mechanisms of AAV-mediated neurotoxicity and test novel AAV designs mitigating these adverse effects.


Subject(s)
Dependovirus , Ganglia, Spinal , Animals , Rabbits , Dependovirus/genetics , Genetic Vectors , Male , Humans , Transgenes , Female , Sensory Receptor Cells
5.
J Clin Invest ; 134(4)2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38357922

ABSTRACT

Chronic and elevated levels of the antiviral cytokine IFN-α in the brain are neurotoxic. This is best observed in patients with genetic cerebral interferonopathies such as Aicardi-Goutières syndrome. Cerebral interferonopathies typically manifest in early childhood and lead to debilitating disease and premature death. There is no cure for these diseases with existing treatments largely aimed at managing symptoms. Thus, an effective therapeutic strategy is urgently needed. Here, we investigated the effect of antisense oligonucleotides targeting the murine IFN-α receptor (Ifnar1 ASOs) in a transgenic mouse model of cerebral interferonopathy. Intracerebroventricular injection of Ifnar1 ASOs into transgenic mice with brain-targeted chronic IFN-α production resulted in a blunted cerebral interferon signature, reduced neuroinflammation, restoration of blood-brain barrier integrity, absence of tissue destruction, and lessened neuronal damage. Remarkably, Ifnar1 ASO treatment was also effective when given after the onset of neuropathological changes, as it reversed such disease-related features. We conclude that ASOs targeting the IFN-α receptor halt and reverse progression of IFN-α-mediated neuroinflammation and neurotoxicity, opening what we believe to be a new and promising approach for the treatment of patients with cerebral interferonopathies.


Subject(s)
Interferon Type I , Nervous System Diseases , Child, Preschool , Humans , Mice , Animals , Neuroinflammatory Diseases , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Interferon-alpha/genetics , Mice, Transgenic
6.
J Proteome Res ; 20(7): 3689-3700, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34085531

ABSTRACT

Novel therapies and biomarkers are needed for the treatment of acute ischemic stroke (AIS). This study aimed to provide comprehensive insights into the dynamic proteome changes and underlying molecular mechanisms post-ischemic stroke. TMT-coupled proteomic analysis was conducted on mouse brain cortex tissue from five time points up to 4 weeks poststroke in the distal hypoxic-middle cerebral artery occlusion (DH-MCAO) model. We found that nearly half of the detected proteome was altered following stroke, but only ∼8.6% of the changes were at relatively large scales. Clustering on the changed proteome defined four distinct expression patterns characterized by temporal and quantitative changes in innate and adaptive immune response pathways and cytoskeletal and neuronal remodeling. Further analysis on a subset of 309 "top hits", which temporally responded to stroke with relatively large and sustained changes, revealed that they were mostly secreted proteins, highly correlated to different cortical cytokines, and thereby potential pharmacodynamic biomarker candidates for inflammation-targeting therapies. Closer examination of the top enriched neurophysiologic pathways identified 57 proteins potentially associated with poststroke recovery. Altogether, our study generated a rich dataset with candidate proteins worthy of further validation as biomarkers and/or therapeutic targets for stroke. The proteomics data are available in the PRIDE Archive with identifier PXD025077.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Animals , Mice , Proteome/genetics , Proteomics
7.
Proc Natl Acad Sci U S A ; 116(16): 7941-7950, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30944222

ABSTRACT

Studies over the past decade have revealed a central role for innate immune sensors in autoimmune and autoinflammatory diseases. cGAS, a cytosolic DNA sensor, detects both foreign and host DNA and generates a second-messenger cGAMP, which in turn binds and activates stimulator of IFN genes (STING), leading to induction of type I interferons and inflammatory cytokines. Recently, gain-of-function mutations in STING have been identified in patients with STING-associated vasculopathy with onset in infancy (SAVI). SAVI patients present with early-onset systemic inflammation and interstitial lung disease, resulting in pulmonary fibrosis and respiratory failure. Here, we describe two independent SAVI mouse models, harboring the two most common mutations found in patients. A direct comparison of these strains reveals a hierarchy of immune abnormalities, lung inflammation and fibrosis, which do not depend on either IFN-α/ß receptor signaling or mixed lineage kinase domain-like pseudokinase (MLKL)-dependent necroptotic cell death pathways. Furthermore, radiation chimera experiments reveal how bone marrow from the V154M mutant mice transfer disease to the WT host, whereas the N153S does not, indicating mutation-specific disease outcomes. Moreover, using radiation chimeras we find that T cell lymphopenia depends on T cell-intrinsic expression of the SAVI mutation. Collectively, these mutant mice recapitulate many of the disease features seen in SAVI patients and highlight mutation-specific functions of STING that shed light on the heterogeneity observed in SAVI patients.


Subject(s)
Disease Models, Animal , Interferon Type I/metabolism , Vascular Diseases , Animals , Cell Death/immunology , Cytokines/metabolism , Gain of Function Mutation , Inflammation/immunology , Inflammation/physiopathology , Mice , Vascular Diseases/genetics , Vascular Diseases/immunology , Vascular Diseases/physiopathology
8.
Blood Adv ; 2(21): 2904-2916, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30396910

ABSTRACT

The main complication of replacement therapy with factor in hemophilia A (HemA) is the formation of inhibitors (neutralizing anti-factor VIII [FVIII] antibodies) in ∼30% of severe HemA patients. Because these inhibitors render replacement FVIII treatment essentially ineffective, preventing or eliminating them is of top priority in disease management. The extended half-life recombinant FVIII Fc fusion protein (rFVIIIFc) is an approved therapy for HemA patients. In addition, it has been reported that rFVIIIFc may induce tolerance to FVIII more readily than FVIII alone in HemA patients that have developed inhibitors. Given that the immunoglobulin G1 Fc region has the potential to interact with immune cells expressing Fc receptors (FcRs) and thereby affect the immune response to rFVIII, we investigated how human macrophages, expressing both FcRs and receptors reported to bind FVIII, respond to rFVIIIFc. We show herein that rFVIIIFc, but not rFVIII, uniquely skews macrophages toward an alternatively activated regulatory phenotype. rFVIIIFc initiates signaling events that result in morphological changes, as well as a specific gene expression and metabolic profile that is characteristic of the regulatory type Mox/M2-like macrophages. Further, these changes are dependent on rFVIIIFc-FcR interactions. Our findings elucidate mechanisms of potential immunomodulatory properties of rFVIIIFc.


Subject(s)
Factor VIII/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Macrophage Activation/drug effects , Macrophages/metabolism , Recombinant Fusion Proteins/pharmacology , Cells, Cultured , Factor VIII/therapeutic use , Gene Expression Regulation/drug effects , Hemophilia A/drug therapy , Hemophilia A/pathology , Humans , Immunoglobulin Fc Fragments/therapeutic use , Leukocytes, Mononuclear/cytology , Macrophages/cytology , Macrophages/drug effects , Receptors, Fc/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/therapeutic use , Signal Transduction/drug effects
9.
Sci Signal ; 11(541)2018 07 31.
Article in English | MEDLINE | ID: mdl-30065029

ABSTRACT

Members of the family of nuclear factor κB (NF-κB) transcription factors are critical for multiple cellular processes, including regulating innate and adaptive immune responses, cell proliferation, and cell survival. Canonical NF-κB complexes are retained in the cytoplasm by the inhibitory protein IκBα, whereas noncanonical NF-κB complexes are retained by p100. Although activation of canonical NF-κB signaling through the IκBα kinase complex is well studied, few regulators of the NF-κB-inducing kinase (NIK)-dependent processing of noncanonical p100 to p52 and the subsequent nuclear translocation of p52 have been identified. We discovered a role for cyclin-dependent kinase 12 (CDK12) in transcriptionally regulating the noncanonical NF-κB pathway. High-content phenotypic screening identified the compound 919278 as a specific inhibitor of the lymphotoxin ß receptor (LTßR), and tumor necrosis factor (TNF) receptor superfamily member 12A (FN14)-dependent nuclear translocation of p52, but not of the TNF-α receptor-mediated nuclear translocation of p65. Chemoproteomics identified CDK12 as the target of 919278. CDK12 inhibition by 919278, the CDK inhibitor THZ1, or siRNA-mediated knockdown resulted in similar global transcriptional changes and prevented the LTßR- and FN14-dependent expression of MAP3K14 (which encodes NIK) as well as NIK accumulation by reducing phosphorylation of the carboxyl-terminal domain of RNA polymerase II. By coupling a phenotypic screen with chemoproteomics, we identified a pathway for the activation of the noncanonical NF-κB pathway that could serve as a therapeutic target in autoimmunity and cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , NF-kappa B/metabolism , Osteosarcoma/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Cyclins/metabolism , Gene Expression Profiling , High-Throughput Screening Assays , Humans , Indoles/pharmacology , Lymphotoxin beta Receptor/antagonists & inhibitors , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B p52 Subunit/genetics , NF-kappa B p52 Subunit/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Propionates/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteome , Signal Transduction , TWEAK Receptor/antagonists & inhibitors , TWEAK Receptor/genetics , TWEAK Receptor/metabolism , Tumor Cells, Cultured , NF-kappaB-Inducing Kinase
10.
Transfusion ; 54(1): 179-89, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23621760

ABSTRACT

BACKGROUND: Red blood cell (RBC) alloantibodies to nonself antigens may develop after transfusion or pregnancy, leading to morbidity and mortality in the form of hemolytic transfusion reactions or hemolytic disease of the newborn. A better understanding of the mechanisms of RBC alloantibody induction, or strategies to mitigate the consequences of such antibodies, may ultimately improve transfusion safety. However, such studies are inherently difficult in humans. STUDY DESIGN AND METHODS: We recently generated transgenic mice with RBC-specific expression of the human KEL glycoprotein, specifically the KEL2 or KEL1 antigens. Herein, we investigate recipient alloimmune responses to transfused RBCs in this system. RESULTS: Transfusion of RBCs from KEL2 donors into wild-type recipients (lacking the human KEL protein but expressing the murine KEL ortholog) resulted in dose-dependent anti-KEL glycoprotein immunoglobulin (Ig)M and IgG antibody responses, enhanced by recipient inflammation with poly(I:C). Boostable responses were evident upon repeat transfusion, with morbid-appearing alloimmunized recipients experiencing rapid clearance of transfused KEL2 but not control RBCs. Although KEL1 RBCs were also immunogenic after transfusion into wild-type recipients, transfusion of KEL1 RBCs into KEL2 recipients or vice versa failed to lead to detectable anti-KEL1 or anti-KEL2 responses. CONCLUSIONS: This murine model, with reproducible and clinically significant KEL glycoprotein alloantibody responses, provides a platform for future mechanistic studies of RBC alloantibody induction and consequences. Long-term translational goals of these studies include improving transfusion safety for at-risk patients.


Subject(s)
Erythrocyte Transfusion/methods , Erythrocytes/immunology , Isoantibodies/biosynthesis , Kell Blood-Group System/immunology , Anemia, Hemolytic/genetics , Anemia, Hemolytic/immunology , Animals , Antibody Formation/drug effects , Antibody Formation/genetics , Blood Group Incompatibility/genetics , Blood Group Incompatibility/immunology , Erythrocytes/metabolism , Humans , Inflammation/immunology , Kell Blood-Group System/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C
11.
Blood ; 122(8): 1494-504, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23801629

ABSTRACT

Exposure to nonself red blood cell (RBC) antigens, either from transfusion or pregnancy, may result in alloimmunization and incompatible RBC clearance. First described as a pregnancy complication 80 years ago, hemolytic disease of the fetus and newborn (HDFN) is caused by alloimmunization to paternally derived RBC antigens. Despite the morbidity/mortality of HDFN, women at risk for RBC alloimmunization have few therapeutic options. Given that alloantibodies to antigens in the KEL family are among the most clinically significant, we developed a murine model with RBC-specific expression of the human KEL antigen to evaluate the impact of maternal/fetal KEL incompatibility. After exposure to fetal KEL RBCs during successive pregnancies with KEL-positive males, 21 of 21 wild-type female mice developed anti-KEL alloantibodies; intrauterine fetal anemia and/or demise occurred in a subset of KEL-positive pups born to wild type, but not agammaglobulinemic mothers. Similar to previous observations in humans, pregnancy-associated alloantibodies were detrimental in a transfusion setting, and transfusion-associated alloantibodies were detrimental in a pregnancy setting. This is the first pregnancy-associated HDFN model described to date, which will serve as a platform to develop targeted therapies to prevent and/or mitigate the dangers of RBC alloantibodies to fetuses and newborns.


Subject(s)
Anemia, Hemolytic/immunology , Erythrocytes/cytology , Isoantibodies/immunology , Kell Blood-Group System/immunology , Models, Animal , Anemia, Hemolytic/genetics , Animals , Blood Transfusion , Cytokines/metabolism , Female , Green Fluorescent Proteins/metabolism , Immunoglobulin G/immunology , Kell Blood-Group System/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Pregnancy, Animal
12.
Transfusion ; 52(12): 2620-30, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22494154

ABSTRACT

BACKGROUND: KEL1, also known as "K", is one of the most immunogenic red blood cell (RBC) antigens. KEL2, also known as "k," differs from KEL1 by a single amino acid. Anti-Kell system antibodies can lead to significant adverse clinical outcomes in humans, including hemolytic complications in alloimmunized transfusion recipients or in infants of alloimmunized mothers. To provide a platform for in-depth immunologic studies of alloimmunization and subsequent sequelae, we generated transgenic mice expressing the human KEL1 or KEL2 antigens. STUDY DESIGN AND METHODS: Vectors were created in which cDNAs encoding either KEL1 or KEL2 were regulated by an erythroid specific ß-globin promoter and enhancer. Pronuclear microinjections were carried out into a C57BL6 background, and founder pups were identified by polymerase chain reaction and screened for expression by flow cytometry. RBC life span and antigen stability were assessed by dye labeling RBCs, transfusing into agammaglobulinemic (µMT) recipients, and tracking by flow cytometry. RESULTS: The expression of either KEL1 or KEL2 is RBC specific and first occurs on early RBC precursors. Both KEL1 and KEL2 RBCs have a normal circulatory life span and stable antigen expression. Expression of KEL1 or KEL2 does not result in altered levels of murine Kell, and resulting RBCs have normal hematologic variables. CONCLUSION: The KEL1 and KEL2 mice represent the first murine system of RBC immunity with antithetical antigens, allowing a more precise modeling of human RBC immunology in general and also a platform for development of novel therapeutics to prevent or minimize the dangers of RBC alloimmunization to the KEL1 and KEL2 antigens in particular.


Subject(s)
Erythrocytes/immunology , Kell Blood-Group System/genetics , Kell Blood-Group System/immunology , Mice, Transgenic , Models, Animal , Animals , Blood Transfusion , Epitopes/genetics , Epitopes/immunology , Erythrocytes/cytology , Gene Library , Humans , Mice , Mice, Inbred C57BL , Transgenes/genetics
13.
Ethics Behav ; 5(1): 25-48, 1995.
Article in English | MEDLINE | ID: mdl-11654169

ABSTRACT

A series of studies investigated the capacity of children between the ages of 7 and 12 to give free and informed consent to participation in psychological research. Children were reasonably accurate in describing the purpose of studies, but many did not understand the possible benefits or especially the possible risks of participating. In several studies children's consent was not affected by the knowledge that their parents had given their permission or by the parents saying that they would not be upset if the children refused. In contrast, other studies found that children were much more likely to stop their participation if the experimenter said explicitly that she would not be upset if they stopped. We suggest that experimenters should pay more attention to describing the possible risks and benefits of participation in research, and that they should also make it clearer to children that they are free to stop once they have begun.


Subject(s)
Behavioral Research , Child , Cognition , Comprehension , Informed Consent , Mental Competency , Psychology , Research , Canada , Coercion , Empirical Research , Evaluation Studies as Topic , Human Experimentation , Humans , Parent-Child Relations , Research Subjects , Researcher-Subject Relations , Risk , Risk Assessment
SELECTION OF CITATIONS
SEARCH DETAIL
...