Network


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

Hotspot


Dive into the research topics where Pinky Agarwal is active.

Publication


Featured researches published by Pinky Agarwal.


Movement Disorders | 2008

Movement Disorder Society-Sponsored Revision of the Unified Parkinson's Disease Rating Scale (MDS-UPDRS): Scale Presentation and Clinimetric Testing Results

Christopher G. Goetz; Barbara C. Tilley; Stephanie R. Shaftman; Glenn T. Stebbins; Stanley Fahn; Pablo Martinez-Martin; Werner Poewe; Cristina Sampaio; Matthew B. Stern; Richard Dodel; Bruno Dubois; Robert G. Holloway; Joseph Jankovic; Jaime Kulisevsky; Anthony E. Lang; Andrew J. Lees; Sue Leurgans; Peter A. LeWitt; David L. Nyenhuis; C. Warren Olanow; Olivier Rascol; Anette Schrag; Jeanne A. Teresi; Jacobus J. van Hilten; Nancy R. LaPelle; Pinky Agarwal; Saima Athar; Yvette Bordelan; Helen Bronte-Stewart; Richard Camicioli

We present a clinimetric assessment of the Movement Disorder Society (MDS)‐sponsored revision of the Unified Parkinsons Disease Rating Scale (MDS‐UPDRS). The MDS‐UDPRS Task Force revised and expanded the UPDRS using recommendations from a published critique. The MDS‐UPDRS has four parts, namely, I: Non‐motor Experiences of Daily Living; II: Motor Experiences of Daily Living; III: Motor Examination; IV: Motor Complications. Twenty questions are completed by the patient/caregiver. Item‐specific instructions and an appendix of complementary additional scales are provided. Movement disorder specialists and study coordinators administered the UPDRS (55 items) and MDS‐UPDRS (65 items) to 877 English speaking (78% non‐Latino Caucasian) patients with Parkinsons disease from 39 sites. We compared the two scales using correlative techniques and factor analysis. The MDS‐UPDRS showed high internal consistency (Cronbachs alpha = 0.79–0.93 across parts) and correlated with the original UPDRS (ρ = 0.96). MDS‐UPDRS across‐part correlations ranged from 0.22 to 0.66. Reliable factor structures for each part were obtained (comparative fit index > 0.90 for each part), which support the use of sum scores for each part in preference to a total score of all parts. The combined clinimetric results of this study support the validity of the MDS‐UPDRS for rating PD.


PLOS Genetics | 2011

Genome-Wide Gene-Environment Study Identifies Glutamate Receptor Gene GRIN2A as a Parkinson's Disease Modifier Gene via Interaction with Coffee

Taye H. Hamza; Honglei Chen; Erin M. Hill-Burns; Shannon L. Rhodes; Jennifer S. Montimurro; Denise M. Kay; Albert Tenesa; Victoria I. Kusel; Patricia Sheehan; Muthukrishnan Eaaswarkhanth; Dora Yearout; Ali Samii; John W. Roberts; Pinky Agarwal; Yikyung Park; Liyong Wang; Jianjun Gao; Jeffery M. Vance; Kenneth S. Kendler; Silviu Alin Bacanu; William K. Scott; Beate Ritz; John G. Nutt; Stewart A. Factor; Cyrus P. Zabetian; Haydeh Payami

Our aim was to identify genes that influence the inverse association of coffee with the risk of developing Parkinsons disease (PD). We used genome-wide genotype data and lifetime caffeinated-coffee-consumption data on 1,458 persons with PD and 931 without PD from the NeuroGenetics Research Consortium (NGRC), and we performed a genome-wide association and interaction study (GWAIS), testing each SNPs main-effect plus its interaction with coffee, adjusting for sex, age, and two principal components. We then stratified subjects as heavy or light coffee-drinkers and performed genome-wide association study (GWAS) in each group. We replicated the most significant SNP. Finally, we imputed the NGRC dataset, increasing genomic coverage to examine the region of interest in detail. The primary analyses (GWAIS, GWAS, Replication) were performed using genotyped data. In GWAIS, the most significant signal came from rs4998386 and the neighboring SNPs in GRIN2A. GRIN2A encodes an NMDA-glutamate-receptor subunit and regulates excitatory neurotransmission in the brain. Achieving P2df = 10−6, GRIN2A surpassed all known PD susceptibility genes in significance in the GWAIS. In stratified GWAS, the GRIN2A signal was present in heavy coffee-drinkers (OR = 0.43; P = 6×10−7) but not in light coffee-drinkers. The a priori Replication hypothesis that “Among heavy coffee-drinkers, rs4998386_T carriers have lower PD risk than rs4998386_CC carriers” was confirmed: ORReplication = 0.59, PReplication = 10−3; ORPooled = 0.51, PPooled = 7×10−8. Compared to light coffee-drinkers with rs4998386_CC genotype, heavy coffee-drinkers with rs4998386_CC genotype had 18% lower risk (P = 3×10−3), whereas heavy coffee-drinkers with rs4998386_TC genotype had 59% lower risk (P = 6×10−13). Imputation revealed a block of SNPs that achieved P2df<5×10−8 in GWAIS, and OR = 0.41, P = 3×10−8 in heavy coffee-drinkers. This study is proof of concept that inclusion of environmental factors can help identify genes that are missed in GWAS. Both adenosine antagonists (caffeine-like) and glutamate antagonists (GRIN2A-related) are being tested in clinical trials for treatment of PD. GRIN2A may be a useful pharmacogenetic marker for subdividing individuals in clinical trials to determine which medications might work best for which patients.


JAMA Neurology | 2010

SNCA Variant Associated With Parkinson Disease and Plasma α-Synuclein Level

Ignacio F. Mata; Min Shi; Pinky Agarwal; Kathryn A. Chung; Karen L. Edwards; Stewart A. Factor; Douglas Galasko; Carmen Ginghina; Alida Griffith; Donald S. Higgins; Denise M. Kay; Hojoong Kim; James B. Leverenz; Joseph F. Quinn; John W. Roberts; Ali Samii; Katherine W. Snapinn; Debby W. Tsuang; Dora Yearout; Jing Zhang; Haydeh Payami; Cyrus P. Zabetian

BACKGROUND A functional repeat polymorphism in the SNCA promoter (REP1) conveys susceptibility for Parkinson disease (PD). There is also increasing evidence that single-nucleotide polymorphisms (SNPs) elsewhere in the gene are associated with PD risk. OBJECTIVES To further explore the association of common SNCA SNPs with PD susceptibility, to determine whether evidence of allelic heterogeneity exists, and to examine the correlation between PD-associated variants and plasma α-synuclein levels. DESIGN Two-tiered analysis. SETTING Academic research. PATIENTS Patients and control subjects from the NeuroGenetics Research Consortium. MAIN OUTCOME MEASURES We performed a 2-tiered analysis of 1956 patients with PD and 2112 controls from the NeuroGenetics Research Consortium using a comprehensive tag SNP approach. Previously published REP1 genotypes were also included. Plasma α-synuclein was assayed in 86 patients with PD and 78 controls using a highly sensitive Luminex assay. RESULTS Five of 15 SNPs genotyped were associated with PD under an additive model in tier 1 (α = .05). Of these, 4 were successfully replicated in tier 2. In the combined sample, the most significant marker was rs356219 (odds ratio, 1.41; 95% confidence interval, 1.28-1.55; P = 1.6 × 10(-12)), located approximately 9 kilobases downstream from the gene. A regression model containing rs356219 alone best fit the data. The linkage disequilibrium correlation coefficient between this SNP and REP1 was low (r(2) = 0.09). The risk-associated C allele of rs356219 was also correlated with higher transformed plasma α-synuclein levels in patients under an adjusted additive model (P = .005). CONCLUSIONS Our data suggest that 1 or more unidentified functional SNCA variants modify risk for PD and that the effect is larger than and independent of REP1. This variant(s), tagged by rs356219, might act by upregulating SNCA expression in a dose-dependent manner.


Movement Disorders | 2013

A randomized, double-blind, placebo-controlled trial of pridopidine in Huntington's disease

Kieburtz Karl; Andrew McGarry; Michael P. McDermott; Elise Kayson; Francis O. Walker; Jody Goldstein; Christopher Hyson; Pinky Agarwal; Patricia Deppen; Jess G. Fiedorowicz; Sandra K. Kostyk; Allison Wright; Blair R. Leavitt; Martha Nance; Mark S. LeDoux; Kathleen M. Shannon; Andrew Siderowf; Merit Cudkowicz; Karen Rabinowitz; Victoria Ross; Arthur Watts; Joakim Tedroff

We examined the effects of 3 dosages of pridopidine, a dopamine‐stabilizing compound, on motor function and other features of Huntingtons disease, with additional evaluation of its safety and tolerability. This was a randomized, double‐blind, placebo‐controlled trial in outpatient neurology clinics at 27 sites in the United States and Canada. Two hundred twenty‐seven subjects enrolled from October 24, 2009, to May 10, 2010. The intervention was pridopidine, either 20 (n=56), 45 (n=55), or 90 (n=58) mg daily for 12 weeks or matching placebo (n=58). The primary outcome measure was the change from baseline to week 12 in the Modified Motor Score, a subset of the Unified Huntingtons Disease Rating Scale Total Motor Score. Measures of safety and tolerability included adverse events and trial completion on the assigned dosage. After 12 weeks, the treatment effect (relative to placebo, where negative values indicate improvement) of pridopidine 90 mg/day on the Modified Motor Score was −1.2 points (95% confidence interval [CI], −2.5 to 0.1 points; P = .08). The effect on the Total Motor Score was −2.8 points (95% CI, −5.4 to −0.1 points; nominal P = .04). No significant effects were seen in secondary outcome measures with any of the active dosages. Pridopidine was generally well tolerated. Although the primary analysis did not demonstrate a statistically significant treatment effect, the overall results suggest that pridopidine may improve motor function in Huntingtons disease. The 90 mg/day dosage appears worthy of further study. Pridopidine was well tolerated.


JAMA | 2016

Effect of Deutetrabenazine on Chorea Among Patients With Huntington Disease: A Randomized Clinical Trial

Samuel Frank; Claudia M. Testa; David Stamler; Elise Kayson; Charles E. Davis; Mary C. Edmondson; Shari Kinel; Blair R. Leavitt; David Oakes; Christine O'Neill; Christina Vaughan; Jody Goldstein; Margaret Herzog; Victoria Snively; Jacquelyn Whaley; Cynthia Wong; Greg Suter; Joseph Jankovic; Joohi Jimenez-Shahed; Christine Hunter; Daniel O. Claassen; Olivia C. Roman; Victor W. Sung; Jenna Smith; Sarah Janicki; Ronda Clouse; Marie Saint-Hilaire; Anna Hohler; Denyse Turpin; Raymond C. James

IMPORTANCE Deutetrabenazine is a novel molecule containing deuterium, which attenuates CYP2D6 metabolism and increases active metabolite half-lives and may therefore lead to stable systemic exposure while preserving key pharmacological activity. OBJECTIVE To evaluate efficacy and safety of deutetrabenazine treatment to control chorea associated with Huntington disease. DESIGN, SETTING, AND PARTICIPANTS Ninety ambulatory adults diagnosed with manifest Huntington disease and a baseline total maximal chorea score of 8 or higher (range, 0-28; lower score indicates less chorea) were enrolled from August 2013 to August 2014 and randomized to receive deutetrabenazine (n = 45) or placebo (n = 45) in a double-blind fashion at 34 Huntington Study Group sites. INTERVENTIONS Deutetrabenazine or placebo was titrated to optimal dose level over 8 weeks and maintained for 4 weeks, followed by a 1-week washout. MAIN OUTCOMES AND MEASURES Primary end point was the total maximal chorea score change from baseline (the average of values from the screening and day-0 visits) to maintenance therapy (the average of values from the week 9 and 12 visits) obtained by in-person visits. This study was designed to detect a 2.7-unit treatment difference in scores. The secondary end points, assessed hierarchically, were the proportion of patients who achieved treatment success on the Patient Global Impression of Change (PGIC) and on the Clinical Global Impression of Change (CGIC), the change in 36-Item Short Form- physical functioning subscale score (SF-36), and the change in the Berg Balance Test. RESULTS Ninety patients with Huntington disease (mean age, 53.7 years; 40 women [44.4%]) were enrolled. In the deutetrabenazine group, the mean total maximal chorea scores improved from 12.1 (95% CI, 11.2-12.9) to 7.7 (95% CI, 6.5-8.9), whereas in the placebo group, scores improved from 13.2 (95% CI, 12.2-14.3) to 11.3 (95% CI, 10.0-12.5); the mean between-group difference was -2.5 units (95% CI, -3.7 to -1.3) (P < .001). Treatment success, as measured by the PGIC, occurred in 23 patients (51%) in the deutetrabenazine group vs 9 (20%) in the placebo group (P = .002). As measured by the CGIC, treatment success occurred in 19 patients (42%) in the deutetrabenazine group vs 6 (13%) in the placebo group (P = .002). In the deutetrabenazine group, the mean SF-36 physical functioning subscale scores decreased from 47.5 (95% CI, 44.3-50.8) to 47.4 (44.3-50.5), whereas in the placebo group, scores decreased from 43.2 (95% CI, 40.2-46.3) to 39.9 (95% CI, 36.2-43.6), for a treatment benefit of 4.3 (95% CI, 0.4 to 8.3) (P = .03). There was no difference between groups (mean difference of 1.0 unit; 95% CI, -0.3 to 2.3; P = .14), for improvement in the Berg Balance Test, which improved by 2.2 units (95% CI, 1.3-3.1) in the deutetrabenazine group and by 1.3 units (95% CI, 0.4-2.2) in the placebo group. Adverse event rates were similar for deutetrabenazine and placebo, including depression, anxiety, and akathisia. CONCLUSIONS AND RELEVANCE Among patients with chorea associated with Huntington disease, the use of deutetrabenazine compared with placebo resulted in improved motor signs at 12 weeks. Further research is needed to assess the clinical importance of the effect size and to determine longer-term efficacy and safety. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT01795859.


Movement Disorders | 2005

Diagnosis and management of pergolide-induced fibrosis

Pinky Agarwal; Stanley Fahn; Steven J. Frucht

We report on 2 patients treated with pergolide, 1 of whom developed pleural fibrosis and the other retroperitoneal fibrosis. In both cases, an extensive diagnostic evaluation and surgical intervention were required to reach a diagnosis. Based on our experience with these patients and a review of cases of pergolide‐induced fibrosis in the English‐language literature, we propose guidelines for the diagnosis and management of this rare complication.


Movement Disorders | 2008

Adult-onset Alexander disease with progressive ataxia and palatal tremor

Katherine L. Howard; Deborah A. Hall; Michelle Moon; Pinky Agarwal; Elizabeth Newman; Michael Brenner

A novel glial fibrillary acidic protein (GFAP) mutation, Y257C, is reported in a patient with adult‐onset Alexander disease. This is the oldest reported case with confirmation of a GFAP mutation. Onset was late in the sixth decade. Genetic analysis of the GFAP gene is recommended in cases of progressive ataxia and palatal tremor.


JAMA Neurology | 2017

ADS-5102 (Amantadine) Extended-Release Capsules for Levodopa-Induced Dyskinesia in Parkinson Disease (EASE LID Study): A Randomized Clinical Trial

Rajesh Pahwa; Caroline M. Tanner; Robert A. Hauser; Stuart Isaacson; Paul A. Nausieda; Daniel D. Truong; Pinky Agarwal; Keith L. Hull; Kelly E. Lyons; Reed Johnson; Mary Jean Stempien

Importance Medical treatment of levodopa-induced dyskinesia (LID) in Parkinson disease (PD) is an unmet need. Objective To evaluate the efficacy and safety of ADS-5102 (amantadine) extended-release 274-mg capsules for treatment of LID in patients with PD. Design, Setting, and Participants A randomized, double-blind, placebo-controlled clinical trial was conducted between May 7, 2014, and July 22, 2015, at 44 North American sites among patients with PD treated with levodopa who experienced at least 1 hour of troublesome dyskinesia per day with at least mild functional impact. Interventions Patients were randomized to receive placebo or 274 mg of ADS-5102 administered orally at bedtime for up to 25 weeks. Main Outcomes and Measures The primary efficacy analysis was the change from baseline to week 12 in the Unified Dyskinesia Rating Scale total score for ADS-5102 vs placebo in the modified intent-to-treat population. OFF time (amount of time the PD medication is not controlling motor symptoms) was a key secondary end point. Safety analyses included all patients who received the study drug (ADS-5102 or placebo). Results A total of 189 patients were screened, and 126 were randomized; the modified intent-to-treat population included 121 patients (51 women and 70 men; mean [SD] age, 64.7 [9.1] years). At week 12, the least-squares mean (SE) change in the Unified Dyskinesia Rating Scale score was –15.9 (1.6) for ADS-5102 (n = 63) and –8.0 (1.6) for placebo (n = 58) (treatment difference, –7.9; 95% CI, –12.5 to –3.3; P < .001). OFF time decreased by a mean (SE) of 0.6 (0.3) hours for ADS-5102 and increased by 0.3 (0.3) hours for placebo (treatment difference, –0.9 hours; 95% CI, –1.6 to –0.2; P = .02). Common adverse events for ADS-5102 vs placebo included visual hallucinations (15 [23.8%] vs 1 [1.7%]), peripheral edema (15 [23.8%] vs 0), and dizziness (14 [22.2%] vs 0). Adverse events led to treatment discontinuation for 13 patients receiving ADS-5102 (20.6%) vs 4 patients receiving placebo (6.9%). Conclusions and Relevance ADS-5102, 274 mg at bedtime, may be an effective treatment for LID. An additional benefit is reduced OFF time. To our knowledge, this is the first demonstration of an oral treatment reducing both LID and OFF time in patients with PD with dyskinesia. Trial Registration clinicaltrials.gov Identifier: NCT02136914


International Journal of Neuroscience | 2009

MOVEMENT DISORDERS ASSOCIATED WITH ARIPIPRAZOLE USE: A CASE SERIES

Deborah A. Hall; Pinky Agarwal; Alida Griffith; Vicki Segro; Lauren C. Seeberger

Aripiprazole is an atypical antipsychotic that is a partial agonist at the D2 and 5HT1a receptors and an antagonist at 5HT2a receptors. Despite previous hypotheses that it would be less likely to cause movement disorders, recent reports suggest it actually may be more likely to cause movement disorders than other atypical antipsychotics. This case series illustrates the variety of movement disorders associated with aripiprazole use at three movement disorder clinics. It also suggests that aripiprazole be used with caution in patients with a prior history of dystonia, parkinsonism, or previous tardive dyskinesia.


Neuropsychiatric Disease and Treatment | 2010

Gabapentin enacarbil - clinical efficacy in restless legs syndrome.

Pinky Agarwal; Alida Griffith; Henry R Costantino; Narendra Vaish

Restless legs syndrome (RLS) is a sleep-related movement disorder commonly involving an unpleasant urge to move the limbs, typically the legs. Dopaminergic agents represent the first-line therapy for RLS; however, long-term use of such drugs results in worsening symptoms due to “augmentation” or other adverse events. Gabapentin, an analog of the inhibitory neurotransmitter gamma-aminobutyric acid (GABA), is an anticonvulsant/analgesic agent. Gabapentin is only mildly effective in relieving RLS symptoms, perhaps a result of its poor absorption from the gastrointestinal (GI) tract. Gabapentin enacarbil is a prodrug of gabapentin specifically designed to enhance absorption via the GI tract, and hence provide improved circulating levels of gabapentin on metabolism. Clinical trials to date have demonstrated favorable safety and (compared to traditional gabapentin) improved pharmacokinetics and efficacy in treating RLS symptoms. Thus, gabapentin enacarbil may prove to be a useful drug in treating RLS. An application of gabapentin enacarbil for treatment of RLS is currently pending with FDA for approval.

Collaboration


Dive into the Pinky Agarwal's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ali Samii

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Dora Yearout

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John W. Roberts

Virginia Mason Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge