Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lee Honigberg is active.

Publication


Featured researches published by Lee Honigberg.


PLOS ONE | 2011

Functional Studies on the IBD Susceptibility Gene IL23R Implicate Reduced Receptor Function in the Protective Genetic Variant R381Q

Svetlana Pidasheva; Sara Trifari; Anne Phillips; Jason A. Hackney; Yan Ma; Ashley Smith; Sue J. Sohn; Hergen Spits; Randall David Little; Timothy W. Behrens; Lee Honigberg; Nico Ghilardi; Hilary Clark

Genome-wide association studies (GWAS) in several populations have demonstrated significant association of the IL23R gene with IBD (Crohns disease (CD) and ulcerative colitis (UC)) and psoriasis, suggesting that perturbation of the IL-23 signaling pathway is relevant to the pathophysiology of these diseases. One particular variant, R381Q (rs11209026), confers strong protection against development of CD. We investigated the effects of this variant in primary T cells from healthy donors carrying IL23RR381 and IL23RQ381 haplotypes. Using a proprietary anti-IL23R antibody, ELISA, flow cytometry, phosphoflow and real-time RT-PCR methods, we examined IL23R expression and STAT3 phosphorylation and activation in response to IL-23. IL23RQ381 was associated with reduced STAT3 phosphorylation upon stimulation with IL-23 and decreased number of IL-23 responsive T-cells. We also observed slightly reduced levels of proinflammatory cytokine secretion in IL23RQ381 positive donors. Our study shows conclusively that IL23RQ381 is a loss-of-function allele, further strengthening the implication from GWAS results that the IL-23 pathway is pathogenic in human disease. This data provides an explanation for the protective role of R381Q in CD and may lead to the development of improved therapeutics for autoimmune disorders like CD.


Science Translational Medicine | 2015

Effect of selective LRRK2 kinase inhibition on nonhuman primate lung

Reina N. Fuji; Michael Flagella; Miriam Baca; Marco A. S. Baptista; Jens Brodbeck; Bryan K. Chan; Brian K. Fiske; Lee Honigberg; Adrian M. Jubb; Paula Katavolos; Donna W. Lee; Sock-Cheng Lewin-Koh; Tori Lin; Xingrong Liu; Shannon Liu; Joseph P. Lyssikatos; Jennifer O'Mahony; Mike Reichelt; Merone Roose-Girma; Zejuan Sheng; Todd Sherer; Ashley Smith; Margaret Solon; Zachary Kevin Sweeney; Jacqueline M. Tarrant; Alison Urkowitz; Søren Warming; Murat Yaylaoglu; Shuo Zhang; Haitao Zhu

LRRK2 kinase inhibitors, under development for Parkinson’s disease, have an effect on type II pneumocytes in nonhuman primate lung, suggesting that pulmonary toxicity may be a critical safety liability. A lung phenotype for LRRK2 inhibitors Human genetic evidence implicates leucine-rich repeat kinase 2 (LRRK2) as a high-priority drug target for Parkinson’s disease. However, the benefit and risk of inhibiting the kinase activity of LRRK2 is unknown and is currently untested in humans. Using two selective LRRK2 kinase inhibitors, Fuji et al. report a safety liability in nonhuman primates characterized by morphological changes in lung. These results are consistent with observations in mice lacking LRRK2. These safety observations offer a cautionary note for pharmacological modulation of LRRK2 in humans. Inhibition of the kinase activity of leucine-rich repeat kinase 2 (LRRK2) is under investigation as a possible treatment for Parkinson’s disease. However, there is no clinical validation as yet, and the safety implications of targeting LRRK2 kinase activity are not well understood. We evaluated the potential safety risks by comparing human and mouse LRRK2 mRNA tissue expression, by analyzing a Lrrk2 knockout mouse model, and by testing selective brain-penetrating LRRK2 kinase inhibitors in multiple species. LRRK2 mRNA tissue expression was comparable between species. Phenotypic analysis of Lrrk2 knockout mice revealed morphologic changes in lungs and kidneys, similar to those reported previously. However, in preclinical toxicity assessments in rodents, no pulmonary or renal changes were induced by two distinct LRRK2 kinase inhibitors. Both of these kinase inhibitors induced abnormal cytoplasmic accumulation of secretory lysosome-related organelles known as lamellar bodies in type II pneumocytes of the lung in nonhuman primates, but no lysosomal abnormality was observed in the kidney. The pulmonary change resembled the phenotype of Lrrk2 knockout mice, suggesting that this was LRRK2-mediated rather than a nonspecific or off-target effect. A biomarker of lysosomal dysregulation, di-docosahexaenoyl (22:6) bis(monoacylglycerol) phosphate (di-22:6-BMP), was also decreased in the urine of Lrrk2 knockout mice and nonhuman primates treated with LRRK2 kinase inhibitors. Our results suggest a role for LRRK2 in regulating lysosome-related lamellar bodies and that pulmonary toxicity may be a critical safety liability for LRRK2 kinase inhibitors in patients.


Science Translational Medicine | 2017

Targeting factor D of the alternative complement pathway reduces geographic atrophy progression secondary to age-related macular degeneration

Brian L. Yaspan; David F. Williams; Frank G. Holz; Carl D. Regillo; Zhengrong Li; Amy Dressen; Menno van Lookeren Campagne; Kha Le; Robert R. Graham; Tatiana Beres; Tushar Bhangale; Lee Honigberg; Ashley Smith; Erin Henry; Carole Ho; Erich C. Strauss

The phase 2 MAHALO clinical trial shows that lampalizumab reduces geographic atrophy secondary to age-related macular degeneration and implicates complement dysregulation in disease pathogenesis. Illuminating a new treatment for macular degeneration Geographic atrophy secondary to age-related macular degeneration is a major cause of vision loss for which there is no treatment. Yaspan et al. now report the results of the MAHALO phase 2 randomized, controlled trial that evaluated lampalizumab in patients with geographic atrophy secondary to age-related macular degeneration. Lampalizumab is a specific inhibitor of complement factor D, a pivotal regulator of the alternative complement pathway. The MAHALO study met its primary efficacy endpoint with a 20% reduction in lesion area progression compared to sham control with monthly lampalizumab treatment. Moreover, lampalizumab showed an acceptable safety profile. A more substantial monthly treatment benefit of 44% reduction in geographic atrophy progression versus sham control was observed in a subgroup of patients who were complement factor I risk-allele carriers. 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.


Proteomics Clinical Applications | 2015

Development and evaluation of a multiplexed mass spectrometry based assay for measuring candidate peptide biomarkers in Alzheimer's Disease Neuroimaging Initiative (ADNI) CSF

Daniel S. Spellman; Kristin Wildsmith; Lee Honigberg; Marianne Tuefferd; David Baker; Nandini Raghavan; Angus C. Nairn; Pascal Croteau; Michael Schirm; Rene Allard; Julie Lamontagne; Daniel Chelsky; S.C. Hoffmann; William Z. Potter; Alzheimer's Disease Neuroimaging Initiative

We describe the outcome of the Biomarkers Consortium CSF Proteomics Project (where CSF is cerebral spinal fluid), a public–private partnership of government, academia, nonprofit, and industry. The goal of this study was to evaluate a multiplexed MS‐based approach for the qualification of candidate Alzheimers disease (AD) biomarkers using CSF samples from the AD Neuroimaging Initiative.


Alzheimers & Dementia | 2016

CLINICAL TRIAL DESIGN OF CREAD: A RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED, PARALLEL-GROUP PHASE 3 STUDY TO EVALUATE CRENEZUMAB TREATMENT IN PATIENTS WITH PRODROMAL-TO-MILD ALZHEIMER’S DISEASE

Thomas Blaettler; Janice Smith; Jillian Smith; Robert Paul; Veronica Asnaghi; Reina N. Fuji; Angelica Quartino; Lee Honigberg; Michael Rabbia; Susan Yule; Susanne Ostrowitzki; Paulo Fontoura

with sustained suppression at all doses. After multiple doses, change from baseline in mean CSF Ab42 was 63.2 and 79.3% in the 15 and 50 mg AZD3293 groups, respectively, with similar reductions in Ab40 (Table 2). Baseline CSFAb42 levels were similar in the healthy Japanese and non-Japanese subjects. Conclusions: AZD3293 was generally well tolerated and potently reduced plasma and CSFAb peptides in Japanese adult subjects. Reductions in CSF Ab peptides were similar to those previously reported in non-Japanese subjects with AD.


Neurology | 2018

ABBY: A phase 2 randomized trial of crenezumab in mild to moderate Alzheimer disease

Jeffrey L. Cummings; Sharon Cohen; Christopher H. van Dyck; Mark Brody; Craig Curtis; William Cho; Michael Ward; Michel Friesenhahn; Christina Rabe; Flavia Brunstein; Angelica Quartino; Lee Honigberg; Reina N. Fuji; David Clayton; Deborah L. Mortensen; Carole Ho; Robert H. Paul

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 Alzheimers 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 β-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. Clinicaltrials.gov 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.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 Alzheimers 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 &bgr;-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. Clinicaltrials.gov 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.


JAMA Ophthalmology | 2018

Efficacy and Safety of Lampalizumab for Geographic Atrophy Due to Age-Related Macular Degeneration: Chroma and Spectri Phase 3 Randomized Clinical Trials

Frank G. Holz; Srinivas R Sadda; Brandon G. Busbee; Emily Y. Chew; Paul Mitchell; Adnan Tufail; Christopher Brittain; Daniela Ferrara; Sarah Gray; Lee Honigberg; Jillian Martin; Barbara Tong; Jason S. Ehrlich; Neil M. Bressler

Importance Geographic atrophy (GA) secondary to age-related macular degeneration is a leading cause of visual disability in older individuals. A phase 2 trial suggested that lampalizumab, a selective complement factor D inhibitor, reduced the rate of GA enlargement, warranting phase 3 trials. Objective To assess the safety and efficacy of lampalizumab vs sham procedure on enlargement of GA. Design, Setting, and Participants Two identically designed phase 3 double-masked, randomized, sham-controlled clinical trials, Chroma and Spectri, enrolled participants from August 28, 2014, to October 6, 2016, at 275 sites in 23 countries. Participants were aged 50 years or older, with bilateral GA and no prior or active choroidal neovascularization in either eye and GA lesions in the study eye measuring 2.54 to 17.78 mm2 with diffuse or banded fundus autofluorescence patterns. Interventions Participants were randomized 2:1:2:1 to receive 10 mg of intravitreous lampalizumab every 4 weeks, sham procedure every 4 weeks, 10 mg of lampalizumab every 6 weeks, or sham procedure every 6 weeks, through 96 weeks. Main Outcomes and Measures Safety and efficacy assessed as mean change from baseline in GA lesion area at week 48 from centrally read fundus autofluorescence images of the lampalizumab arms vs pooled sham arms, in the intent-to-treat population and by complement factor I–profile genetic biomarker. Results A total of 906 participants (553 women and 353 men; mean [SD] age, 78.1 [8.1] years) were enrolled in Chroma and 975 participants (578 women and 397 men; mean [SD] age, 77.9 [8.1] years) were enrolled in Spectri; 1733 of the 1881 participants (92.1%) completed the studies through 48 weeks. The adjusted mean increases in GA lesion area from baseline at week 48 were 1.93 to 2.09 mm2 across all groups in both studies. Differences in adjusted mean change in GA lesion area (lampalizumab minus sham) were −0.02 mm2 (95% CI, −0.21 to 0.16 mm2; P = .80) for lampalizumab every 4 weeks in Chroma, 0.16 mm2 (95% CI, 0.00-0.31 mm2; P = .048) for lampalizumab every 4 weeks in Spectri, 0.05 mm2 (95% CI, −0.13 to 0.24 mm2; P = .59) for lampalizumab every 6 weeks in Chroma, and 0.09 mm2 (95% CI, −0.07 to 0.24 mm2; P = .27) for lampalizumab every 6 weeks in Spectri. No benefit of lampalizumab was observed across prespecified subgroups, including by complement factor I–profile biomarker. Endophthalmitis occurred after 5 of 12 447 injections (0.04%) or in 5 of 1252 treated participants (0.4%) through week 48. Conclusions and Relevance In Chroma and Spectri, the largest studies of GA conducted to date, lampalizumab did not reduce GA enlargement vs sham during 48 weeks of treatment. Results highlight the substantial and consistent enlargement of GA, at a mean of approximately 2 mm2 per year. Trial Registration ClinicalTrials.gov Identifier: NCT02247479 and NCT02247531


Alzheimer's & Dementia: Translational Research & Clinical Interventions | 2018

The Alzheimer's Prevention Initiative Autosomal-Dominant Alzheimer's Disease Trial: A study of crenezumab versus placebo in preclinical PSEN1 E280A mutation carriers to evaluate efficacy and safety in the treatment of autosomal-dominant Alzheimer's disease, including a placebo-treated noncarrier cohort

Pierre N. Tariot; Francisco Lopera; Jessica B. Langbaum; Ronald G. Thomas; Suzanne Hendrix; Lon S. Schneider; Silvia Rios-Romenets; Margarita Giraldo; Natalia Acosta; Carlos Tobón; Claudia Ramos; Alejandro Espinosa; William Cho; Michael Ward; David Clayton; Michael Friesenhahn; Howard Mackey; Lee Honigberg; Sandra Sanabria Bohorquez; Kewei Chen; Trisha Walsh; Carolyn Langlois; Eric M. Reiman; Alzheimer's Prevention Initiative

Autosomal‐dominant Alzheimers disease (ADAD) represents a crucial population for identifying prevention strategies that might modify disease course for cognitively unimpaired individuals at high imminent risk for developing symptoms due to Alzheimers disease (AD), that is, who have “preclinical” AD. Crenezumab is an antiamyloid monoclonal antibody that binds monomeric and aggregated forms of amyloid β, with highest affinity for oligomers; it is in development for early stages of sporadic AD and for ADAD.


The Journal of Allergy and Clinical Immunology | 2018

Elevated IgE M1 prime transcripts in nasal tissues in patients with nasal polyps and asthma

Heleen Scheerens; Ashley Smith; Olga Li; Lee Honigberg; Jeffrey M. Harris; Gabriele Holtappels; Claus Bachert

A novel PCR assay detected IgE M1 prime transcripts in nasal polyps of patients with CRSwNP and asthma, confirming that IgE switching occurred and membrane IgE-expressing B cells were resident at mucosal sites.


Alzheimers & Dementia | 2018

BASELINE RECOVERY AND LONGITUDINAL STABILITY OF CEREBROSPINAL FLUID AMYLOID-β PEPTIDES: AN EVALUATION OF COLLECTION METHODS, LONGITUDINAL ASSAY PERFORMANCE, AND BIOMARKER STABILITY OVER A 2-YEAR PERIOD

Stephen Schauer; Kristin R. Wildsmith; Christina Rabe; Lee Honigberg

was shown to be < 0.5 pg/mL and the LoQ < 5 pg/mL. No high dose hook effect was observed for samples containing up to 30000 pg/mL of phospho-Tau181. Linearity was shown across the clinical relevant range. A method comparison study with the INNOTEST PHOSPHO-TAU-(181P) assay demonstrated a good correlation (r>0.90). Conclusions:Automation, the mono test cartridge principle, short throughput times, and instrument flexibility are key attributes of the LUMIPULSE G instrument series making it the ideal platform to fulfill today’s needs for rapid and accurate quantification of CSF biomarkers. The novel Lumipulse G pTau 181 assay (under development) shows good sensitivity and precision, correlates well with the INNOTEST assay and completes the routine CSF biomarker panel for AD on LUMIPULSE.

Collaboration


Dive into the Lee Honigberg's collaboration.

Researchain Logo
Decentralizing Knowledge