Network


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

Hotspot


Dive into the research topics where Sheila Gujrathi is active.

Publication


Featured researches published by Sheila Gujrathi.


The New England Journal of Medicine | 2016

Ozanimod Induction and Maintenance Treatment for Ulcerative Colitis

William J. Sandborn; Brian G. Feagan; Douglas C. Wolf; Geert D’Haens; Severine Vermeire; Stephen B. Hanauer; Subrata Ghosh; Heather Smith; Matthew Cravets; Paul Frohna; Richard Aranda; Sheila Gujrathi; Allan Olson

BACKGROUND Ozanimod (RPC1063) is an oral agonist of the sphingosine-1-phosphate receptor subtypes 1 and 5 that induces peripheral lymphocyte sequestration, potentially decreasing the number of activated lymphocytes circulating to the gastrointestinal tract. METHODS We conducted a double-blind, placebo-controlled phase 2 trial of ozanimod in 197 adults with moderate-to-severe ulcerative colitis. Patients were randomly assigned, in a 1:1:1 ratio, to receive ozanimod at a dose of 0.5 mg or 1 mg or placebo daily for up to 32 weeks. The Mayo Clinic score was used to measure disease activity on a scale from 0 to 12, with higher scores indicating more severe disease; subscores range from 0 to 3, with higher scores indicating more severe disease. The primary outcome was clinical remission (Mayo Clinic score ≤2, with no subscore >1) at 8 weeks. RESULTS The primary outcome occurred in 16% of the patients who received 1 mg of ozanimod and in 14% of those who received 0.5 mg of ozanimod, as compared with 6% of those who received placebo (P=0.048 and P=0.14, respectively, for the comparison of the two doses of ozanimod with placebo). Differences in the primary outcome between the group that received 0.5 mg of ozanimod and the placebo group were not significant; therefore, the hierarchical testing plan deemed the analyses of secondary outcomes exploratory. Clinical response (decrease in Mayo Clinic score of ≥3 points and ≥30% and decrease in rectal-bleeding subscore of ≥1 point or a subscore ≤1) at 8 weeks occurred in 57% of those receiving 1 mg of ozanimod and 54% of those receiving 0.5 mg, as compared with 37% of those receiving placebo. At week 32, the rate of clinical remission was 21% in the group that received 1 mg of ozanimod, 26% in the group that received 0.5 mg of ozanimod, and 6% in the group that received placebo; the rate of clinical response was 51%, 35%, and 20%, respectively. At week 8, absolute lymphocyte counts declined 49% from baseline in the group that received 1 mg of ozanimod and 32% from baseline in the group that received 0.5 mg. The most common adverse events overall were anemia and headache. CONCLUSIONS In this preliminary trial, ozanimod at a daily dose of 1 mg resulted in a slightly higher rate of clinical remission of ulcerative colitis than placebo. The trial was not large enough or of sufficiently long duration to establish clinical efficacy or assess safety. (Funded by Receptos; TOUCHSTONE ClinicalTrials.gov number, NCT01647516.).


Gastroenterology | 2012

Abatacept for Crohn's Disease and Ulcerative Colitis

William J. Sandborn; Jean-Frederic Colombel; Bruce E. Sands; Paul Rutgeerts; Stephan R. Targan; Remo Panaccione; Brian Bressler; K. Geboes; Stefan Schreiber; Richard Aranda; Sheila Gujrathi; Allison Luo; Yun Peng; Luisa Salter–Cid; Stephen B. Hanauer

BACKGROUND & AIMS The efficacy of abatacept, a selective costimulation modulator, in Crohns disease (CD) and ulcerative colitis (UC) is unknown. METHODS Four placebo-controlled trials evaluated the efficacy and safety of abatacept as induction (IP) and maintenance (MP) therapy in adults with active, moderate-to-severe CD (CD-IP; CD-MP) and UC (UC-IP1; UC-MP). In CD-IP and UC-IP1, 451 patients with CD and 490 patients with UC were randomized to abatacept 30, 10, or 3 mg/kg (according to body weight) or placebo, and dosed at weeks 0, 2, 4, and 8. In MP, 90 patients with CD and 131 patients with UC who responded to abatacept at week 12 in the induction trials were randomized to abatacept 10 mg/kg or placebo every 4 weeks through week 52. RESULTS In CD-IP, 17.2%, 10.2%, and 15.5% of patients receiving abatacept 30, 10, and 3 mg/kg achieved a clinical response at weeks 8 and 12, vs 14.4% receiving placebo (P = .611, P = .311, and P = .812, respectively). In UC-IP1, 21.4%, 19.0%, and 20.3% of patients receiving abatacept 30, 10, and 3 mg/kg achieved a clinical response at week 12, vs 29.5% receiving placebo (P = .124, P = .043, and P = .158, respectively). In CD-MP, 23.8% vs 11.1% of abatacept vs placebo patients were in remission at week 52. In UC-MP, 12.5% vs 14.1% of patients receiving abatacept vs placebo were in remission at week 52. Safety generally was comparable between groups. CONCLUSIONS The studies showed that abatacept is not efficacious for the treatment of moderate-to-severe CD or UC.


Gut | 2014

Anti-IP-10 antibody (BMS-936557) for ulcerative colitis: a phase II randomised study

Lloyd Mayer; William J. Sandborn; Yuriy Stepanov; Karel Geboes; Robert Hardi; Michael Yellin; Xiaolu Tao; Li An Xu; Luisa Salter-Cid; Sheila Gujrathi; Richard Aranda; Allison Luo

Objective Interferon-γ-inducible protein-10 (IP-10 or CXCL10) plays a role in inflammatory cell migration and epithelial cell survival and migration. It is expressed in higher levels in the colonic tissue and plasma of patients with ulcerative colitis (UC). This phase II study assessed the efficacy and safety of BMS-936557, a fully human, monoclonal antibody to IP-10, in the treatment of moderately-to-severely active UC. Design In this 8-week, phase II, double-blind, multicentre, randomised study, patients with active UC received placebo or BMS-936557 (10 mg/kg) intravenously every other week. The primary endpoint was the rate of clinical response at Day 57; clinical remission and mucosal healing rates were secondary endpoints. Post hoc analyses evaluated the drug exposure–response relationship and histological improvement. Results 109 patients were included (BMS-936557: n=55; placebo: n=54). Prespecified primary and secondary endpoints were not met; clinical response rate at Day 57 was 52.7% versus 35.2% for BMS-936557 versus placebo (p=0.083), and clinical remission and mucosal healing rates were 18.2% versus 16.7% (p=1.00) and 41.8% versus 35.2% (p=0.556), respectively. However, higher BMS-936557 steady-state trough concentration (Cminss) was associated with increased clinical response (87.5% vs 37.0% (p<0.001) for patients with Cminss 108–235 μg/ml vs placebo) and histological improvements (73.0% vs 41.0%; p=0.004). Infections occurred in 7 (12.7%) BMS-936557-treated patients and 3 (5.8%) placebo-treated patients. 2 (3.6%) BMS-936557 patients discontinued due to adverse events. Conclusions Anti-IP-10 antibody, BMS-936557, is a potentially effective therapy for moderately-to-severely active UC. Higher drug exposure correlated with increasing clinical response and histological improvement. Further dose–response studies are warranted. Clinical Trial Registration Number: ClinicalTrials.gov NCT00656890.


The Journal of Rheumatology | 2013

Safety of Abatacept Administered Intravenously in Treatment of Rheumatoid Arthritis: Integrated Analyses of up to 8 Years of Treatment from the Abatacept Clinical Trial Program

Michael E. Weinblatt; Larry W. Moreland; Rene Westhovens; Roger B. Cohen; S. Kelly; Nader Khan; Ramesh Pappu; Ingrid Delaet; Allison Luo; Sheila Gujrathi; Marc C. Hochberg

Objective. To assess the overall safety, including rare events, of intravenous (IV) abatacept treatment in rheumatoid arthritis (RA). Methods. Data from 8 clinical trials of IV abatacept in RA were pooled. Safety events were assessed during the short-term (duration ≤ 12 months) and cumulative (short-term plus longterm extensions) abatacept treatment periods. Incidence rates per 100 patient-years were calculated. Standardized incidence ratios (SIR) for hospitalized infections and malignancies were compared with external RA cohorts and, for malignancies, with the US general population. Results. There were 3173 IV abatacept-treated patients with 2331 patient-years of exposure in the short-term periods, and 4149 IV abatacept-treated patients with 12,132 patient-years of exposure in the cumulative period. Incidence rates for serious infections were low and consistent over time (3.68 for abatacept vs 2.60 for placebo during the short-term, and 2.87 for abatacept during the cumulative period). Hospitalized infections were generally similar to external RA patient cohorts and were consistent over time. Incidence rates of malignancies were similar for abatacept- and placebo-treated patients during the short-term period (0.73 vs 0.59) and remained low during the abatacept cumulative period (0.73). SIR of some tissue-specific malignancies (e.g., colorectal and breast) in the cumulative period tended to be lower, while others (lymphoma and lung) tended to be higher, compared with the general population; however, incidence rates were comparable with RA cohorts. Autoimmune events were rare and infusion reactions uncommon. Conclusion. Longterm safety of IV abatacept was consistent with the short-term, with no unexpected events and low incidence rates of serious infections, malignancies, and autoimmune events.


Annals of the Rheumatic Diseases | 2012

Abatacept in subjects who switch from intravenous to subcutaneous therapy: results from the phase IIIb ATTUNE study

Edward C. Keystone; Joel M. Kremer; Anthony H. Russell; Jane H. Box; Carlos Abud-Mendoza; Mario Garza Elizondo; Allison Luo; Richard Aranda; Ingrid Delaet; Rene Swanink; Sheila Gujrathi; Michael E. Luggen

Objective To assess safety, immunogenicity and efficacy in rheumatoid arthritis (RA) patients switched from long-term intravenous to subcutaneous (SC) abatacept. Methods In this phase IIIb, open-label, single-arm trial, patients who completed ≥4 years of intravenous abatacept (in long-term extensions of two phase III studies) were enrolled to receive SC abatacept (125 mg/week). The primary objective was safety during the first 3 months after switching from intravenous therapy. Results 123 patients entered the study (mean Disease Activity Score 28 (based on C reactive protein) and HAQ-DI of 3.4 and 0.94, respectively). At month 3, 120 (97.6%) patients were continuing to receive SC abatacept; no patients discontinued due to lack of efficacy. Adverse events (AEs) were reported in 49 (39.8%) patients through month 3. One patient (0.8%) discontinued due to an AE and one patient (0.8%) experienced a serious AE. Two (1.6%) patients had SC injection site reactions (erythema, pain), both with mild intensity. Clinical efficacy was maintained throughout. Limited impact on immunogenicity was observed when switching routes of administration. Conclusion These data demonstrate that patients can switch from long-term monthly intravenous abatacept to a weekly fixed dose of 125 mg SC abatacept with no increased safety concerns. This study further supports SC abatacept as an alternative treatment option for patients with RA.


Lancet Neurology | 2016

Safety and efficacy of the selective sphingosine 1-phosphate receptor modulator ozanimod in relapsing multiple sclerosis (RADIANCE): a randomised, placebo-controlled, phase 2 trial.

Jeffrey Cohen; Douglas L. Arnold; Giancarlo Comi; Amit Bar-Or; Sheila Gujrathi; Jeffrey Hartung; Matt Cravets; Allan Olson; Paul Frohna; Krzysztof Selmaj

BACKGROUND Modulation of sphingosine 1-phosphate (S1P) receptors in a non-selective manner decreases disease activity in patients with multiple sclerosis but has potential safety concerns. We assessed the safety and efficacy of the oral selective S1P receptor modulator ozanimod in patients with relapsing multiple sclerosis. METHODS RADIANCE is a combined phase 2/3 trial. Patients with relapsing multiple sclerosis were recruited from 55 academic and private multiple sclerosis clinics in 13 countries across Europe and the USA. Eligible participants were aged 18-55 years, had an Expanded Disability Status Scale (EDSS) score of 0-5·0, and had either one or more relapses in the previous 12 months, or one or more relapses in the past 24 months and one or more gadolinium-enhancing lesions on MRI in the previous 12 months before screening. Participants were assigned by a computer-generated randomisation sequence in a 1:1:1 ratio to ozanimod (0·5 mg or 1 mg) or matching placebo once daily for 24 weeks by an independent, unmasked, statistical team. Trial participants, study site personnel, MRI assessors, steering committee members, and the study statistician were masked to treatment assignment. To attenuate first-dose cardiac effects, ozanimod was up-titrated from 0·25 mg to 0·5 mg or 1 mg over 8 days. The primary endpoint was the cumulative number of total gadolinium-enhancing MRI lesions measured by an independent MRI analysis centre at weeks 12-24 after treatment initiation. Analysis was by intention to treat. Here, we report results from the 24-week phase 2 trial. This trial is registered with ClinicalTrials.gov, number NCT01628393. The 2-year phase 3 trial is ongoing. FINDINGS The first patient was randomised on Oct 18, 2012, and the final visit of the last randomised patient was on May 11, 2014. The intention-to-treat and safety population consisted of 258 participants, 88 were assigned placebo, 87 ozanimod 0·5 mg, and 83 ozanimod 1 mg; 252 (98%) patients completed the assigned treatment. The mean cumulative number of gadolinium-enhancing lesions at weeks 12-24 was 11·1 (SD 29·9) with placebo compared with 1·5 (3·7) with ozanimod 0·5 mg (odds ratio 0·16, 95% CI 0·08-0·30; p<0·0001) and 1·5 (3·4) with ozanimod 1 mg (odds ratio 0·11, 95% CI 0·06-0·21; p<0·0001). Three serious adverse events unrelated to treatment were reported in patients assigned ozanimod 0·5 mg: optic neuritis, somatoform autonomic dysfunction, and cervical squamous metaplasia (HPV-related). No serious infectious or cardiac adverse events were reported, and no cases of macular oedema arose. The most common adverse events in the ozanimod 0·5 mg and 1 mg groups compared with placebo were nasopharyngitis (11 and five vs 12), headache (five and three vs eight), and urinary-tract infections (six and two vs two). The maximum reduction in mean heart rate by Holter monitoring during the first 6 h in ozanimod-treated participants was less than 2 beats per min (bpm) compared with baseline, with no patient having a minimum hourly heart rate less than 45 bpm. Electrocardiograms and 24-h Holter monitoring showed no increased incidence of atrioventricular block or sinus pause with ozanimod. INTERPRETATION Ozanimod significantly reduced MRI lesion activity in participants with relapsing multiple sclerosis, with a favourable safety profile over a period of 24 weeks. These findings warrant phase 3 trials, which are ongoing. FUNDING Receptos, Inc.


Autoimmunity | 2010

Targeting lymphocyte co-stimulation: From bench to bedside

Nathan J. Felix; Anish Suri; Luisa Salter-Cid; Steven G. Nadler; Sheila Gujrathi; Michael Corbo; Richard Aranda

T and B lymphocytes are central regulators and effectors of immune responses and are believed to have a key role in many autoimmune diseases. Targeting the activation or effector function of lymphocytes is a potentially effective approach to treat autoimmunity. Typically, T-cell activation occurs after engagement of the T-cell receptor with its cognate peptide-major histocompatibility complex (signal 1) and subsequent engagement of co-stimulatory molecules (signal 2). This “second signal” contributes to T-cell activation by promoting proliferation, survival, and effector function. In general, activation in the absence of co-stimulation leads to a reduced immune response, anergy, or even tolerance. B-cell activation similarly requires co-stimulation for the development of complete effector function. The most potent co-stimulatory molecules identified to date are CD28 for T-cells and CD40 for B-cells. Both molecules are recognized for their potential as immune modulators; however, thus far neither molecule has been successfully targeted directly for the treatment of autoimmune disease. The only current therapy to target either of these pathways is cytotoxic T-lymphocyte antigen-4 (CTLA-4-Ig), which indirectly blocks CD28 signaling and has proven efficacy in rheumatoid arthritis and juvenile idiopathic arthritis patients. In addition to CD28 and CD40, an array of other co-stimulatory as well as inhibitory pathways has recently been identified and scientists are just beginning to understand how these different signaling pathways interact to regulate lymphocyte activation. In the more than two decades since the discovery of the first co-stimulatory molecule, the full clinical potential of these pathways is yet to be realized. In this review, we will primarily focus on CD28 and CD40 which are the most clinically validated co-stimulatory pathways, and briefly summarize and discuss some of the other T-cell co-stimulatory molecules.


Inflammatory Bowel Diseases | 2016

P-106 Examination of an Alternative Definition for Clinical Remission in UC: Results from TOUCHSTONE, a Randomized, Double-Blind, Placebo-Controlled Trial of Ozanimod

William J. Sandborn; Brian G. Feagan; Geert DʼHaens; Doug Wolf; Severine Vermeire; Stephen B. Hanauer; Subrata Ghosh; Heather M. Smith; Matthew Cravets; Paul Frohna; Richard Aranda; Sheila Gujrathi; Allan Olson

Background:The Mayo score has been used to assess UC disease activity and to establish the efficacy of medications approved for the treatment of UC for over 2 decades. The definition of clinical remission used for approval of biologic agents since infliximab has been a Mayo score ⩽2 with no subscore >1, which is being scrutinized by the FDA due to its lack of formal validation and inclusion of the poorly-defined physician global assessment subscore (PGA). An alternative definition of clinical remission using the Mayo scoring system without the PGA, has been proposed that requires a rectal bleeding subscore (RBS) = 0, an endoscopy subscore (ES) of ⩽1 and a stool frequency subscore (SFS) ⩽1 with improvement ≥1. We have explored this alternative definition of remission in a post hoc analysis of the data from the TOUCHSTONE study. Methods:TOUCHSTONE was a randomized, double-blind, placebo-controlled trial designed to evaluate efficacy and safety of 0.5 mg (low dose, LD) and 1 mg (high dose, HD) ozanimod, an oral, selective sphingosine 1-phosphate (S1P) 1 and 5 receptor modulator, in comparison to placebo (PBO), in patients with moderate to severe UC. A total of 197 patients were randomized (1:1:1) and treated once daily with PBO (n = 65), LD (n = 65) or HD (n = 67). The patients who achieved clinical response at week 8 continued into the maintenance period (MP) with their original treatment for an additional 24 weeks. Using the data from TOUCHSTONE, we calculated clinical remission rates at weeks 8 and 32 applying the alternative definition of clinical remission, which excludes the PGA. Results:Of 197 patients in the IP, 103 (52.3%) continued in the MP and 91/103 (88.3%) completed. At week 8 using the original definition (Mayo score ⩽2 with no subscore >1) clinical remission occurred in 16.4% for HD (P = 0.0482 versus PBO), 13.8% for LD (P = 0.1422), and 6.2% for PBO while the alternate definition (RBS = 0, SFS ⩽1 with improvement ≥1, ES ⩽1), clinical remission occurred in 23.9%, HD (P = 0.0154 versus PBO), 16.9%, LD (P = 0.2099), and 9.2%, PBO. At week 32, using the original definition, clinical remission occurred in 20.9%, HD (P = 0.0108 versus PBO), 26.2%, LD (P = 0.0021), and 6.2%, PBO while using the alternative definition clinical remission occurred in 26.9%, HD (P = 0.0025 versus PBO), 26.2%, LD (P = 0.0053), and 7.7%, PBO. Conclusions:Using this alternative definition of clinical remission, a greater difference between HD and PBO was observed. An endpoint that excludes the PGA but maintains the other components of the Mayo score (RBS, ES, SFS) is able to demonstrate a treatment effect and could be one of the endpoints in clinical trials of UC. Further, the analysis confirms that patients with moderate to severe UC treated with ozanimod HD were more likely to both achieve and maintain clinical remission.


Gastroenterology | 2010

A Randomized, Placebo-Controlled Trial of MDX-1100, an Anti-IP-10 Antibody, for Moderately to Severely Active Ulcerative Colitis

Lloyd Mayer; William J. Sandborn; Yuriy Stepanov; Judith Maccarone; Xiaolu Tao; Li-An Lu; Sheila Gujrathi; Richard Aranda; Allison Luo


Gastroenterology | 2015

445 The TOUCHSTONE Study: A Randomized, Double-Blind, Placebo-Controlled Induction Trial of an Oral S1P Receptor Modulator (RPC1063) in Moderate to Severe Ulcerative Colitis

William J. Sandborn; Brian G. Feagan; Douglas C. Wolf; Geert R. D'Haens; Severine Vermeire; Stephen B. Hanauer; Subrata Ghosh; Heather M. Smith; Matt Cravets; Paul Frohna; Sheila Gujrathi; Allan Olson

Collaboration


Dive into the Sheila Gujrathi's collaboration.

Top Co-Authors

Avatar

Allan Olson

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard Aranda

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian G. Feagan

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Severine Vermeire

Katholieke Universiteit Leuven

View shared research outputs
Researchain Logo
Decentralizing Knowledge