Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Mov Disord ; 38(3): 386-398, 2023 03.
Article in English | MEDLINE | ID: mdl-36807624

ABSTRACT

BACKGROUND: Leucine-rich repeat kinase 2 (LRRK2) inhibition is a promising therapeutic approach for the treatment of Parkinson's disease (PD). OBJECTIVE: The aim of this study was to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of the potent, selective, CNS-penetrant LRRK2 inhibitor BIIB122 (DNL151) in healthy participants and patients with PD. METHODS: Two randomized, double-blind, placebo-controlled studies were completed. The phase 1 study (DNLI-C-0001) evaluated single and multiple doses of BIIB122 for up to 28 days in healthy participants. The phase 1b study (DNLI-C-0003) evaluated BIIB122 for 28 days in patients with mild to moderate PD. The primary objectives were to investigate the safety, tolerability, and plasma pharmacokinetics of BIIB122. Pharmacodynamic outcomes included peripheral and central target inhibition and lysosomal pathway engagement biomarkers. RESULTS: A total of 186/184 healthy participants (146/145 BIIB122, 40/39 placebo) and 36/36 patients (26/26 BIIB122, 10/10 placebo) were randomized/treated in the phase 1 and phase 1b studies, respectively. In both studies, BIIB122 was generally well tolerated; no serious adverse events were reported, and the majority of treatment-emergent adverse events were mild. BIIB122 cerebrospinal fluid/unbound plasma concentration ratio was ~1 (range, 0.7-1.8). Dose-dependent median reductions from baseline were observed in whole-blood phosphorylated serine 935 LRRK2 (≤98%), peripheral blood mononuclear cell phosphorylated threonine 73 pRab10 (≤93%), cerebrospinal fluid total LRRK2 (≤50%), and urine bis (monoacylglycerol) phosphate (≤74%). CONCLUSIONS: At generally safe and well-tolerated doses, BIIB122 achieved substantial peripheral LRRK2 kinase inhibition and modulation of lysosomal pathways downstream of LRRK2, with evidence of CNS distribution and target inhibition. These studies support continued investigation of LRRK2 inhibition with BIIB122 for the treatment of PD. © 2023 Denali Therapeutics Inc and The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Leukocytes, Mononuclear/metabolism , Healthy Volunteers , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Biomarkers/metabolism , Mutation
2.
Clin Transl Sci ; 15(8): 2010-2023, 2022 08.
Article in English | MEDLINE | ID: mdl-35649245

ABSTRACT

RIPK1 is a master regulator of inflammatory signaling and cell death and increased RIPK1 activity is observed in human diseases, including Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS). RIPK1 inhibition has been shown to protect against cell death in a range of preclinical cellular and animal models of diseases. SAR443060 (previously DNL747) is a selective, orally bioavailable, central nervous system (CNS)-penetrant, small-molecule, reversible inhibitor of RIPK1. In three early-stage clinical trials in healthy subjects and patients with AD or ALS (NCT03757325 and NCT03757351), SAR443060 distributed into the cerebrospinal fluid (CSF) after oral administration and demonstrated robust peripheral target engagement as measured by a reduction in phosphorylation of RIPK1 at serine 166 (pRIPK1) in human peripheral blood mononuclear cells compared to baseline. RIPK1 inhibition was generally safe and well-tolerated in healthy volunteers and patients with AD or ALS. Taken together, the distribution into the CSF after oral administration, the peripheral proof-of-mechanism, and the safety profile of RIPK1 inhibition to date, suggest that therapeutic modulation of RIPK1 in the CNS is possible, conferring potential therapeutic promise for AD and ALS, as well as other neurodegenerative conditions. However, SAR443060 development was discontinued due to long-term nonclinical toxicology findings, although these nonclinical toxicology signals were not observed in the short duration dosing in any of the three early-stage clinical trials. The dose-limiting toxicities observed for SAR443060 preclinically have not been reported for other RIPK1-inhibitors, suggesting that these toxicities are compound-specific (related to SAR443060) rather than RIPK1 pathway-specific.


Subject(s)
Alzheimer Disease , Amyotrophic Lateral Sclerosis , Receptor-Interacting Protein Serine-Threonine Kinases , Alzheimer Disease/drug therapy , Amyotrophic Lateral Sclerosis/drug therapy , Double-Blind Method , Healthy Volunteers , Humans , Leukocytes, Mononuclear , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors
3.
Sci Transl Med ; 14(648): eabj2658, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35675433

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are the most common genetic risk factors for Parkinson's disease (PD). Increased LRRK2 kinase activity is thought to impair lysosomal function and may contribute to the pathogenesis of PD. Thus, inhibition of LRRK2 is a potential disease-modifying therapeutic strategy for PD. DNL201 is an investigational, first-in-class, CNS-penetrant, selective, ATP-competitive, small-molecule LRRK2 kinase inhibitor. In preclinical models, DNL201 inhibited LRRK2 kinase activity as evidenced by reduced phosphorylation of both LRRK2 at serine-935 (pS935) and Rab10 at threonine-73 (pT73), a direct substrate of LRRK2. Inhibition of LRRK2 by DNL201 demonstrated improved lysosomal function in cellular models of disease, including primary mouse astrocytes and fibroblasts from patients with Gaucher disease. Chronic administration of DNL201 to cynomolgus macaques at pharmacologically relevant doses was not associated with adverse findings. In phase 1 and phase 1b clinical trials in 122 healthy volunteers and in 28 patients with PD, respectively, DNL201 at single and multiple doses inhibited LRRK2 and was well tolerated at doses demonstrating LRRK2 pathway engagement and alteration of downstream lysosomal biomarkers. Robust cerebrospinal fluid penetration of DNL201 was observed in both healthy volunteers and patients with PD. These data support the hypothesis that LRRK2 inhibition has the potential to correct lysosomal dysfunction in patients with PD at doses that are generally safe and well tolerated, warranting further clinical development of LRRK2 inhibitors as a therapeutic modality for PD.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Parkinson Disease , Animals , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Lysosomes/metabolism , Mice , Mutation , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Phosphorylation
4.
Ann Clin Transl Neurol ; 7(7): 1103-1116, 2020 07.
Article in English | MEDLINE | ID: mdl-32515902

ABSTRACT

OBJECTIVE: To investigate neurodegenerative and inflammatory biomarkers in people with amyotrophic lateral sclerosis (PALS), evaluate their predictive value for ALS progression rates, and assess their utility as pharmacodynamic biomarkers for monitoring treatment effects. METHODS: De-identified, longitudinal plasma, and cerebrospinal fluid (CSF) samples from PALS (n = 108; 85 with samples from ≥2 visits) and controls without neurological disease (n = 41) were obtained from the Northeast ALS Consortium (NEALS) Biofluid Repository. Seventeen of 108 PALS had familial ALS, of whom 10 had C9orf72 mutations. Additional healthy control CSF samples (n = 35) were obtained from multiple sources. We stratified PALS into fast- and slow-progression subgroups using the ALS Functional Rating Scale-Revised change rate. We compared cytokines/chemokines and neurofilament (NF) levels between PALS and controls, among progression subgroups, and in those with C9orf72 mutations. RESULTS: We found significant elevations of cytokines, including MCP-1, IL-18, and neurofilaments (NFs), indicators of neurodegeneration, in PALS versus controls. Among PALS, these cytokines and NFs were significantly higher in fast-progression and C9orf72 mutation subgroups versus slow progressors. Analyte levels were generally stable over time, a key feature for monitoring treatment effects. We demonstrated that CSF/plasma neurofilament light chain (NFL) levels may predict disease progression, and stratification by NFL levels can enrich for more homogeneous patient groups. INTERPRETATION: Longitudinal stability of cytokines and NFs in PALS support their use for monitoring responses to immunomodulatory and neuroprotective treatments. NFs also have prognostic value for fast-progression patients and may be used to select similar patient subsets in clinical trials.


Subject(s)
Amyotrophic Lateral Sclerosis/diagnosis , Amyotrophic Lateral Sclerosis/metabolism , Cytokines/metabolism , Disease Progression , Neurofilament Proteins/metabolism , Adult , Aged , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Biological Specimen Banks , Biomarkers/blood , Biomarkers/cerebrospinal fluid , C9orf72 Protein/genetics , Cytokines/blood , Cytokines/cerebrospinal fluid , Female , Humans , Longitudinal Studies , Male , Middle Aged , Neurofilament Proteins/blood , Neurofilament Proteins/cerebrospinal fluid , Prognosis
5.
Clin Pharmacol Ther ; 107(2): 406-414, 2020 02.
Article in English | MEDLINE | ID: mdl-31437302

ABSTRACT

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) regulates inflammation, cytokine release, and necroptotic cell death and is implicated in pathogenic cellular pathways in amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), and multiple sclerosis. Inhibition of RIPK1 activity protects against inflammation and cell death in multiple animal models. DNL104 is a selective, brain-penetrant inhibitor of RIPK1 phosphorylation in clinical development for AD and ALS. DNL104 was tested in 68 healthy volunteers to investigate safety and tolerability, pharmacokinetic profile in plasma and cerebrospinal fluid, and pharmacodynamic effects of RIPK1 inhibition in peripheral blood mononuclear cells in a first-in-human, placebo-controlled, double-blind, randomized single-ascending dose (SAD) and multiple-ascending dose (MAD) study. DNL104 was well-tolerated in the SAD group and during the dosing period of the MAD group. However, postdose liver toxicity in 37.5% of subjects was observed in the MAD, and assessed to be drug related. We demonstrate that DNL104 leads to RIP1 kinase inhibition, and this is not associated with central nervous system (CNS) toxicities, supporting future development of CNS penetrant RIPK1 inhibitors.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Adolescent , Adult , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Leukocytes, Mononuclear/drug effects , Male , Metabolic Clearance Rate , Middle Aged , Phosphorylation/drug effects , Young Adult
6.
Alzheimers Dement ; 16(1): 131-143, 2020 01.
Article in English | MEDLINE | ID: mdl-31668596

ABSTRACT

INTRODUCTION: Frontotemporal lobar degeneration (FTLD) is the most common form of dementia for those under 60 years of age. Increasing numbers of therapeutics targeting FTLD syndromes are being developed. METHODS: In March 2018, the Association for Frontotemporal Degeneration convened the Frontotemporal Degeneration Study Group meeting in Washington, DC, to discuss advances in the clinical science of FTLD. RESULTS: Challenges exist for conducting clinical trials in FTLD. Two of the greatest challenges are (1) the heterogeneity of FTLD syndromes leading to difficulties in efficiently measuring treatment effects and (2) the rarity of FTLD disorders leading to recruitment challenges. DISCUSSION: New personalized endpoints that are clinically meaningful to individuals and their families should be developed. Personalized approaches to analyzing MRI data, development of new fluid biomarkers and wearable technologies will help to improve the power to detect treatment effects in FTLD clinical trials and enable new, clinical trial designs, possibly leveraged from the experience of oncology trials. A computational visualization and analysis platform that can support novel analyses of combined clinical, genetic, imaging, biomarker data with other novel modalities will be critical to the success of these endeavors.


Subject(s)
Biomarkers , Clinical Trials as Topic , Frontotemporal Lobar Degeneration/genetics , Magnetic Resonance Imaging , Atrophy , Congresses as Topic , Humans
7.
Alzheimers Res Ther ; 10(1): 96, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30231896

ABSTRACT

BACKGROUND: We investigated the effect of crenezumab, a humanized anti-amyloid-beta (Aß) immunoglobulin (Ig)G4 monoclonal antibody, on biomarkers of amyloid pathology, neurodegeneration, and disease progression in patients with mild-to-moderate Alzheimer's disease (AD). METHODS: This double-blind, placebo-controlled, randomized phase II study enrolled patients with mild-to-moderate AD and a Mini-Mental State Examination (MMSE) score of 18-26. In part 1 of the study, patients were 2:1 randomized to receive low-dose subcutaneous (SC) 300 mg crenezumab every 2 weeks (q2w) or placebo for 68 weeks; in part 2, patients were 2:1 randomized to receive high-dose intravenous (IV) 15 mg/kg crenezumab every 4 weeks (q4w) or placebo for 68 weeks. The primary endpoint was change in amyloid burden from baseline to week 69 assessed by florbetapir positron emission tomography (PET) in the modified intent-to-treat population. Secondary endpoints were change from baseline to week 69 in cerebrospinal fluid (CSF) biomarkers and fluorodeoxyglucose PET, and change from baseline to week 73 in 12-point Alzheimer's Disease Assessment Scale cognitive subscale (ADAS-Cog12) and Clinical Dementia Rating Sum of Boxes (CDR-SB). Safety was assessed in patients who received at least one dose of study treatment. RESULTS: From August 2011 to September 2012, 91 patients were enrolled and randomized (low-dose SC cohort: crenezumab (n = 26) or placebo (n = 13); high-dose IV cohort: crenezumab (n = 36) or placebo (n = 16)). The primary endpoint was not met using a prespecified cerebellar reference region to calculate standard uptake value ratios (SUVRs) from florbetapir PET. Exploratory analyses using subcortical white matter reference regions showed nonsignificant trends toward slower accumulation of plaque amyloid in the high-dose IV cohort. In both cohorts, a significant mean increase from baseline in CSF Aß(1-42) levels versus placebo was observed. Nonsignificant trends toward ADAS-Cog12 and CDR-SB benefits were identified in a mild (MMSE 20-26) subset of the high-dose IV cohort. No amyloid-related imaging abnormalities due to edema/effusion were observed. CONCLUSION: The primary endpoint was not met. Exploratory findings suggest potential Aß target engagement with crenezumab and possible slower accumulation of plaque amyloid. Studies investigating the effects of higher doses of crenezumab on amyloid load and disease progression are ongoing. TRIAL REGISTRATION: ClinicalTrials.gov, NCT01397578 . Registered on 18 July 2011.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/cerebrospinal fluid , Antibodies, Monoclonal/therapeutic use , Brain/diagnostic imaging , Aged , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/diagnostic imaging , Amyloid beta-Peptides/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Double-Blind Method , Female , Humans , Magnetic Resonance Imaging , Male , Positron-Emission Tomography , Treatment Outcome
8.
Neurology ; 90(21): e1889-e1897, 2018 05 22.
Article in English | MEDLINE | ID: mdl-29695589

ABSTRACT

OBJECTIVE: To evaluate the safety and efficacy of crenezumab in patients with mild to moderate Alzheimer disease (AD). METHODS: In this phase 2 trial, 431 patients with mild to moderate AD 50 to 80 years of age were randomized 2:1 (crenezumab:placebo). Patients received low-dose subcutaneous crenezumab 300 mg or placebo every 2 weeks (n = 184) or high-dose intravenous crenezumab 15 mg/kg or placebo every 4 weeks (n = 247) for 68 weeks. Primary outcome measures were change in Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-Cog12) and Clinical Dementia Rating-Sum of Boxes scores from baseline to week 73. RESULTS: The primary and secondary endpoints were not met. In an exploratory post hoc analysis, a reduction in decline on the ADAS-Cog12 was observed in the high-dose group. Separation from the placebo group on the ADAS-Cog12 was greatest in the milder subsets of AD patients and reached statistical significance in the group with Mini-Mental State Examination scores of 22 to 26. In both groups, there was a significant increase in CSF ß-amyloid1-42 levels that correlated with crenezumab CSF levels. The overall rate of adverse events was balanced between groups. One case of amyloid-related imaging abnormalities indicative of vasogenic edema or effusions was reported. CONCLUSIONS: Although prespecified criteria for testing treatment effects were not met, these data suggest a potential treatment effect in patients with mild AD treated with high-dose crenezumab. Together with the safety profile for crenezumab, these data support the exploration of crenezumab treatment at even higher doses in patients with early AD. CLINICALTRIALSGOV IDENTIFIER: NCT 01343966. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that, for people with AD, crenezumab does not significantly improve cognition or function at 18 months. The study is rated Class II because <80% of enrolled patients completed the study.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/psychology , Antibodies, Monoclonal/therapeutic use , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized , Double-Blind Method , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Treatment Outcome
9.
Sci Transl Med ; 9(395)2017 06 21.
Article in English | MEDLINE | ID: mdl-28637922

ABSTRACT

Geographic atrophy is an advanced form of age-related macular degeneration (AMD) and a leading cause of vision loss for which there are no approved treatments. Genetic studies in AMD patients have implicated dysregulation of the alternative complement pathway in the pathogenesis of geographic atrophy. Lampalizumab is a potential therapeutic that targets complement factor D, a pivotal activator of the alternative complement pathway. The MAHALO phase 2 clinical trial was a multicenter, randomized, controlled study that evaluated lampalizumab administered by intravitreal injection monthly (n = 42) and every other month (n = 41) versus sham control (n = 40) in patients with geographic atrophy secondary to AMD. The primary endpoint was the mean change in lesion area from baseline to month 18 as measured by fundus autofluorescence. Specific AMD-associated genetic polymorphisms were also analyzed. The MAHALO study met its primary efficacy endpoint with an acceptable safety profile; monthly lampalizumab treatment demonstrated a 20% reduction in lesion area progression versus sham control [80% confidence interval (CI), 4 to 37%]. A more substantial monthly treatment benefit of 44% reduction in geographic atrophy area progression versus sham control (95% CI, 15 to 73%) was observed in a subgroup of complement factor I (CFI) risk-allele carriers (57% of the patients analyzed were CFI risk-allele carriers). The MAHALO study shows a potential treatment effect in patients with geographic atrophy and supports therapeutic targeting of the alternative complement pathway for treating AMD pathogenesis.


Subject(s)
Geographic Atrophy/drug therapy , Geographic Atrophy/metabolism , Immunoglobulin Fab Fragments/therapeutic use , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Aged , Complement Factor D/antagonists & inhibitors , Complement Factor D/metabolism , Complement Pathway, Alternative , Disease Progression , Female , Geographic Atrophy/pathology , Humans , Macular Degeneration/pathology , Male , Middle Aged
11.
Sci Transl Med ; 6(241): 241cm5, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24944190

ABSTRACT

As the prevalence of Alzheimer's disease (AD) grows, so do the costs it imposes on society. Scientific, clinical, and financial interests have focused current drug discovery efforts largely on the single biological pathway that leads to amyloid deposition. This effort has resulted in slow progress and disappointing outcomes. Here, we describe a "portfolio approach" in which multiple distinct drug development projects are undertaken simultaneously. Although a greater upfront investment is required, the probability of at least one success should be higher with "multiple shots on goal," increasing the efficiency of this undertaking. However, our portfolio simulations show that the risk-adjusted return on investment of parallel discovery is insufficient to attract private-sector funding. Nevertheless, the future cost savings of an effective AD therapy to Medicare and Medicaid far exceed this investment, suggesting that government funding is both essential and financially beneficial.


Subject(s)
Alzheimer Disease/drug therapy , Cooperative Behavior , Cost-Benefit Analysis , Drug Discovery , Humans , Private Sector , Public Sector
12.
J Neurosci ; 32(28): 9677-89, 2012 Jul 11.
Article in English | MEDLINE | ID: mdl-22787053

ABSTRACT

Passive immunization against ß-amyloid (Aß) has become an increasingly desirable strategy as a therapeutic treatment for Alzheimer's disease (AD). However, traditional passive immunization approaches carry the risk of Fcγ receptor-mediated overactivation of microglial cells, which may contribute to an inappropriate proinflammatory response leading to vasogenic edema and cerebral microhemorrhage. Here, we describe the generation of a humanized anti-Aß monoclonal antibody of an IgG4 isotype, known as MABT5102A (MABT). An IgG4 subclass was selected to reduce the risk of Fcγ receptor-mediated overactivation of microglia. MABT bound with high affinity to multiple forms of Aß, protected against Aß1-42 oligomer-induced cytotoxicity, and increased uptake of neurotoxic Aß oligomers by microglia. Furthermore, MABT-mediated amyloid plaque removal was demonstrated using in vivo live imaging in hAPP((V717I))/PS1 transgenic mice. When compared with a human IgG1 wild-type subclass, containing the same antigen-binding variable domains and with equal binding to Aß, MABT showed reduced activation of stress-activated p38MAPK (p38 mitogen-activated protein kinase) in microglia and induced less release of the proinflammatory cytokine TNFα. We propose that a humanized IgG4 anti-Aß antibody that takes advantage of a unique Aß binding profile, while also possessing reduced effector function, may provide a safer therapeutic alternative for passive immunotherapy for AD. Data from a phase I clinical trial testing MABT is consistent with this hypothesis, showing no signs of vasogenic edema, even in ApoE4 carriers.


Subject(s)
Alzheimer Disease/therapy , Amyloid beta-Peptides/immunology , Immunoglobulin G/pharmacology , Microglia/drug effects , Microglia/metabolism , Neuroprotective Agents/pharmacology , Peptide Fragments/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/blood , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Newborn , CX3C Chemokine Receptor 1 , Cells, Cultured , Cerebral Cortex/cytology , Disease Models, Animal , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Double-Blind Method , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Humans , Immunoglobulin G/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Confocal , Middle Aged , Mutation/genetics , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/metabolism , Plaque, Amyloid/immunology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Presenilin-1/genetics , Protein Binding/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Chemokine/genetics , Statistics, Nonparametric , Time Factors , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Sci Transl Med ; 3(101): 101ra92, 2011 Sep 21.
Article in English | MEDLINE | ID: mdl-21937757

ABSTRACT

An important goal for personalized health care is the identification of biomarkers that predict the likelihood of treatment responses. Here, we tested the hypothesis that quantitative mRNA assays for B lineage cells in blood could serve as baseline predictors of therapeutic response to B cell depletion therapy in subjects with rheumatoid arthritis (RA). In samples from the REFLEX trial of rituximab in inadequate responders to antibodies to tumor necrosis factor-α, a 25% subgroup of treated subjects with elevated baseline mRNA levels of IgJ, a marker for antibody-secreting plasmablasts, showed reduced clinical response rates. There were no significant efficacy differences in the placebo arm subjects stratified by this marker. Prospective testing of the IgJ biomarker in the DANCER and SERENE rituximab clinical trial cohorts and the SCRIPT ocrelizumab cohort confirmed the utility of this marker to predict nonresponse to anti-CD20 therapy. A combination mRNA biomarker, IgJhiFCRL5lo, showed improved test performance over IgJhi alone. This study demonstrates that baseline blood levels of molecular markers for late-stage B lineage plasmablasts identify a ~20% subgroup of active RA subjects who are unlikely to gain substantial clinical benefit from anti-CD20 B cell depletion therapy.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antigens, CD20/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Plasma Cells/immunology , Biomarkers/blood , Cell Lineage/genetics , Cohort Studies , Demography , Female , Humans , Immunoglobulin J-Chains/genetics , Immunoglobulin J-Chains/metabolism , Male , Middle Aged , RNA, Messenger/blood , RNA, Messenger/genetics , Receptors, Cell Surface/immunology , Receptors, Fc , Reverse Transcriptase Polymerase Chain Reaction , Rituximab , Treatment Outcome
14.
J Neurosci ; 29(27): 8649-54, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19587271

ABSTRACT

The myelin-derived neurite growth inhibitor Nogo has been proposed to play a major role in blocking axon regeneration in the CNS after injuries. However, past studies have produced mixed results regarding the regenerative phenotype of various Nogo-deficient mouse lines after experimental spinal cord injury. Two lines did not display enhanced corticospinal tract (CST) regeneration, and one displayed modest regeneration. A fourth line, a Nogo-A,B gene-trap mutant, was instead reported to exhibit extensive CST regeneration, but the results were later found to be inadvertently confounded with an axon labeling artifact. Of the four Nogo mutant lines studied so far, three continue to express some isoform(s) of Nogo, leaving open the question whether any remaining Nogo protein contributes to the modest regenerative phenotype reported in some. The remaining Nogo mutant line studied was confounded by the unexplained rescue of embryonic lethality associated with this mutation. To gain a better understanding of the contribution of Nogo as an inhibitor of regeneration of CNS axons, and particularly CST axons, we reanalyzed the Nogo-A,B gene-trap mutant line and analyzed a novel, fully viable Nogo deletion mutant line that is null for all known isoforms of Nogo. Our analyses failed to reveal any enhanced CST regeneration after experimental spinal cord injury in either line. These results indicate that Nogo alone does not account for lack of CST regeneration and have implications for current therapeutic development for spinal cord injury in humans by targeting Nogo.


Subject(s)
Myelin Proteins/deficiency , Myelin Proteins/genetics , Nerve Regeneration/genetics , Pyramidal Tracts/physiology , Alleles , Animals , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nerve Regeneration/physiology , Nogo Proteins , Pyramidal Tracts/pathology , Recombination, Genetic , Spinal Cord Injuries/genetics , Spinal Cord Injuries/physiopathology
15.
J Neurosci ; 29(22): 7116-23, 2009 Jun 03.
Article in English | MEDLINE | ID: mdl-19494134

ABSTRACT

Mature neurons have diminished intrinsic regenerative capacity. Axotomy of the peripheral branch of adult dorsal root ganglia (a "conditioning" lesion) triggers a transcription-dependent axon growth program. Here, we show that this growth program requires the function of the transcription factor Smad1. After peripheral axotomy, neuronal Smad1 is upregulated, and phosphorylated Smad1 accumulates in the nucleus. Both events precede the onset of axonal extension. Reducing Smad1 by RNA interference in vitro impairs axonal growth, and the continued presence of Smad1 is required to maintain the growth program. Furthermore, intraganglionic injection of BMP2 or 4, which activates Smad1, markedly enhances axonal growth capacity, mimicking the effect of a conditioning lesion. Thus, activation of Smad1 by axotomy is a key component of the transcriptional switch that promotes an enhanced growth state of adult sensory neurons.


Subject(s)
Axons/physiology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/physiology , Smad1 Protein/metabolism , Analysis of Variance , Animals , Axons/drug effects , Axotomy/methods , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cells, Cultured , Enzyme Activation/drug effects , Functional Laterality , Ganglia, Spinal/cytology , Gene Expression Profiling/methods , Gene Expression Regulation/drug effects , Mice , Microarray Analysis , RNA, Small Interfering/pharmacology , Sensory Receptor Cells/drug effects , Smad1 Protein/genetics , Tubulin/metabolism
16.
Neuron ; 53(5): 649-62, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17329206

ABSTRACT

We report a role for Nogo receptors (NgRs) in macrophage efflux from sites of inflammation in peripheral nerve. Increasing numbers of macrophages in crushed rat sciatic nerves express NgR1 and NgR2 on the cell surface in the first week after injury. These macrophages show reduced binding to myelin and MAG in vitro, which is reversed by NgR siRNA knockdown and by inhibiting Rho-associated kinase. Fourteen days after sciatic nerve crush, regenerating nerves with newly synthesized myelin have fewer macrophages than cut/ligated nerves that lack axons and myelin. Almost all macrophages in the cut/ligated nerves lie within the Schwann cell basal lamina, while in the crushed regenerating nerves the majority migrate out. Furthermore, crush-injured nerves of NgR1- and MAG-deficient mice and Y-27632-treated rats show impaired macrophage efflux from Schwann cell basal lamina containing myelinated axons. These data have implications for the resolution of inflammation in peripheral nerve and CNS pathologies.


Subject(s)
Macrophages/pathology , Myelin Proteins/immunology , Receptors, Peptide/immunology , Sciatic Nerve/injuries , Animals , Cell Movement/immunology , Cells, Cultured , Female , GPI-Linked Proteins , Ligands , Macrophages/metabolism , Mice , Mice, Knockout , Microscopy, Electron , Monocytes/cytology , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin Sheath/immunology , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Nerve Crush , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Nerve Regeneration/immunology , Neurons/cytology , Neurons/immunology , Neurons/pathology , Nogo Proteins , Nogo Receptor 1 , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Schwann Cells/immunology , Schwann Cells/pathology , Schwann Cells/ultrastructure , Sciatic Nerve/immunology , Sciatic Nerve/pathology
17.
Nat Med ; 12(11): 1232; author reply 1232-3, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17088884
18.
Proc Natl Acad Sci U S A ; 102(4): 1205-10, 2005 Jan 25.
Article in English | MEDLINE | ID: mdl-15647357

ABSTRACT

Axon regeneration failure in the adult mammalian CNS is attributed in part to the inhibitory nature of CNS myelin. Three myelin-associated, structurally distinct proteins, Nogo, myelin-associated glycoprotein, and oligodendrocyte myelin glycoprotein, have been implicated in this inhibition. Neuronal Nogo receptor (NgR) binds to each of the three inhibitors and has been proposed to mediate their inhibitory signals by complexing with a signal-transducing coreceptor, the neurotrophin receptor p75(NTR). To assess the contribution of NgR to mediating myelin inhibitory signals and regeneration failure in vivo, we generated and characterized NgR-deficient mice. Nogo transcripts are up-regulated in NgR mutants, indicating that NgR regulates Nogo in vivo. However, neurite outgrowth from NgR-deficient postnatal dorsal root ganglion or cerebellar granule neurons is inhibited by myelin and by a Nogo-66 substrate to the same extent as is from wild-type neurons, whereas p75(NTR)-deficient neurons are less inhibited. The NgR ligand-binding domain promotes neurite outgrowth on Nogo-66, regardless of the genotype of the neurons, indicating that the NgR ligand-binding domain can act independent of NgR. Thus, NgR is not essential for mediating inhibitory signals from CNS myelin, at least in the neurons tested, whereas p75(NTR) plays a central role in this response. Neither NgR-nor p75(NTR)-deficient mice showed enhanced regeneration of corticospinal tract axons in comparison with wild-type controls after spinal dorsal hemisection. Our results thus fail to support a central role for NgR in axonal growth inhibition in vitro or in corticospinal tract regeneration block in vivo.


Subject(s)
Cerebral Cortex/physiology , Myelin Proteins/physiology , Nerve Regeneration , Neurites/physiology , Receptors, Peptide/physiology , Spinal Cord/physiology , Animals , Female , Mice , Mice, Inbred C57BL , Nogo Proteins , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/physiology
19.
J Comp Neurol ; 472(4): 463-77, 2004 May 10.
Article in English | MEDLINE | ID: mdl-15065120

ABSTRACT

In rodents, the main contingent of corticospinal tract (CST) axons descends in the ventral part of the dorsal column. There is, however, a contingent of CST axons that descends in the dorsolateral column (the "dorsolateral corticospinal tract," or DLCST). Here, we define some of the features of the DLCST by tracing CST projections following injections of biotinylated dextran amine into the sensorimotor cortex, assessing the distribution of DLCST axons and terminal arborizations in intact mice and in mice in which the main contingent of CST axons in the dorsal column had been transected. Axons of the DLCST diverge from the main tract at the pyramidal decussation, gather in fascicles in the dorsolateral gray matter below the spinomedullary junction, and project in a gradual trajectory laterally toward the dorsolateral column over the first few cervical segments. DLCST axons then project along the dorsolateral column to sacral levels, giving rise to collaterals that project into the gray matter. Labeled DLCST axons were most abundant in cervical segments, where they were often collected in fascicles, and progressively decreased in number in more caudal segments. Tracing of DLCST axons in mice with selective lesions of the dorsal column revealed that DLCST axons arborize extensively throughout the dorsal and ventral horns and that the overall territory that the DLCST axons invade is similar to the territory innervated by the CST axons in the main tract. Some DLCST axon arbors with varicosities are seen near large neurons in the ventral horn (presumed motoneurons). Substantial numbers of DLCST axons project across the midline to the gray matter on the contralateral side. Thus, the DLCST provides an alternate route for CST input to caudal segments, which is of particular relevance for studies of CST distribution and function following partial spinal cord injuries.


Subject(s)
Biotin/analogs & derivatives , Pyramidal Tracts/anatomy & histology , Pyramidal Tracts/physiology , Animals , Axons/pathology , Brain/pathology , Dextrans , Female , Hindlimb/innervation , Histocytochemistry , Male , Mice , Mice, Inbred C57BL , Pyramidal Tracts/pathology , Rhodamines , Spinal Cord/anatomy & histology , Spinal Cord/cytology
20.
Neuron ; 38(2): 213-24, 2003 Apr 24.
Article in English | MEDLINE | ID: mdl-12718856

ABSTRACT

The failure of regeneration of severed axons in the adult mammalian central nervous system is thought to be due partly to the presence of endogenous inhibitors of axon regeneration. The nogo gene encodes three proteins (Nogo-A, -B, and -C) that have been proposed to contribute to this inhibition. To determine whether deletion of nogo enhances regenerative ability, we generated two lines of mutant mice, one lacking Nogo-A and -B but not -C (Nogo-A/B mutant), and one deficient in all three isoforms (Nogo-A/B/C mutant). Although Nogo-A/B-deficient myelin has reduced inhibitory activity in a neurite outgrowth assay in vitro, tracing of corticospinal tract fibers after dorsal hemisection of the spinal cord did not reveal an obvious increase in regeneration or sprouting of these fibers in either mouse line, suggesting that elimination of Nogo alone is not sufficient to induce extensive axon regeneration.


Subject(s)
Myelin Proteins/deficiency , Nerve Regeneration/physiology , Spinal Cord/physiology , Animals , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Myelin Proteins/genetics , Myelin Proteins/metabolism , Myelin Sheath/physiology , Nerve Regeneration/genetics , Neurites/physiology , Nogo Proteins , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/metabolism , Spinal Cord/cytology , Spinal Cord/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...