28 Retrospective pooled analysis of epacadostat clinical studies identifies doses required for maximal pharmacodynamic effect in anti-PD-1 combination studies

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A27-A27
Author(s):  
Michael Smith ◽  
Robert Newton ◽  
Sherry Owens ◽  
Xiaohua Gong ◽  
Chuan Tian ◽  
...  

BackgroundIDO1 is the initial rate-limiting enzyme in one breakdown pathway of tryptophan. It reduces tryptophan levels and generates metabolites (e.g., kynurenine [KYN]) that contribute to tumor-associated immune suppression. Epacadostat (EPA) is a novel, potent, selective, reversible inhibitor of IDO1 studied in clinical trials in combination with anti-PD-1 antibodies. Epacadostat-induced decreases in plasma KYN have been used as a pharmacodynamic measure of drug activity and have aided in dose selection for clinical studies. Despite encouraging signs of efficacy in combination with pembrolizumab (PMB) in the ECHO-202 study, a large phase 3 study in melanoma (ECHO-301) failed to reproduce this outcome.1MethodsLongitudinal plasma samples were obtained from participants in EPA clinical studies. Plasma KYN and EPA concentrations were measured by validated liquid chromatography tandem mass spectrometry. Quantitative mass spectrometry imaging (qMSI) of intratumoral tryptophan metabolites was also performed.ResultsAnalysis of plasma KYN levels demonstrated that PMB monotherapy significantly elevated KYN. While blocking the PMB-induced increase, EPA (100 mg BID) in combination with PMB failed to normalize KYN to healthy control levels as was reported for EPA monotherapy.2 Because anti-PD-1 treatment can induce interferon gamma (IFN-γ) production and IDO1 expression is IFNg inducible,3 we hypothesize that PMB-induced IFN-γ may be responsible for the observed increase of plasma KYN levels. Combined analysis of plasma KYN from additional EPA/anti-PD-1 combination (ECHO-202; EPA/PMB, ECHO-204; EPA/nivolumab) and monotherapy (ECHO-210) studies, with EPA doses ranging from 50 to 600 mg BID, suggested that higher EPA doses (≥600 mg BID) may be necessary to overcome the anti-PD-1–associated KYN elevation. Doses ≥600 mg BID are projected to cover the EPA IC90 value for 24h. The POD1UM-102 study is currently evaluating the combination of a novel anti-PD-1 monoclonal antibody (retifanlimab) plus EPA at doses up to 900 mg BID. Preliminary results from this study indicate that 600 mg BID is the maximally tolerated dose and is capable of maintaining suppression of KYN to healthy control levels through treatment cycle 4. Additionally, qMSI of paired pre-treatment and on-treatment biopsies demonstrated intratumoral suppression of KYN production with EPA 600 mg BID.ConclusionsUsing suppression of plasma KYN as a pharmacodynamic marker of EPA activity, we demonstrated that maximal blockade of IDO1 activity in the context of anti-PD-1 treatment requires doses of EPA substantially higher than those tested in prior clinical studies. These findings are now informing additional proof of concept clinical studies.AcknowledgementsThese studies were sponsored by Incyte Corporation (ECHO-210, POD1UM-102) and in collaboration with MSD (ECHO-301, ECHO-202) and Bristol Myers Squibb (ECHO-204).Trial RegistrationECHO-202 [NCT NCT02178722]; ECHO-204 [NCT02327078]; ECHO-210 [NCT01685255]; ECHO-301 [NCT02752074]; POD1UM-102 [NCT03589651]Ethics ApprovalThese studies were each approved by the institutional review board or independent ethics committee of participating institutions.ReferencesLong GV, Dummer R, Hamid O, et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomized, double-blind study. Lancet Oncol 2019;20:1083–1097Beatty GL, O’Dwyer PJ, Clark J, et al. First-in-human phase I study of the oral inhibitor of indoleamine 2,3-dioxygenase-1 epacadostat (INCB024360) in patients with advanced solid malignancies. Clin Cancer Res 2017;23:3269–3276.Munn, DH, Mellor AL. IDO in the tumor microenvironment: inflammation, counter-regulation, and tolerance. Trends Immunol 2016;37:193–207.

2015 ◽  
Vol 86 (11) ◽  
pp. e4.26-e4
Author(s):  
David Barnes ◽  
Thomas Leist ◽  
Mark Freedman ◽  
Tomas Olsson ◽  
Aaron Miller ◽  
...  

IntroductionTeriflunomide, approved for the treatment of relapsing-remitting multiple sclerosis, has a well-characterized safety profile based on individual clinical studies. We report pooled safety and tolerability data from four, double-blind, placebo-controlled trials of teriflunomide. Post-approval updates on hair thinning and pregnancy outcomes, sometimes concerns for patients initiating teriflunomide, are reported.MethodsData were pooled from phase 2 (NCT01487096) and phase 3 TEMSO (NCT00134563), TOWER (NCT00751881), and TOPIC (NCT00622700) studies. Patients were randomized to receive teriflunomide 14 mg, 7 mg, or placebo. Safety analyses were performed for all patients exposed to teriflunomide.ResultsThe pooled dataset included 3044 patients. Commonly reported adverse events (AEs) were in accordance with individual clinical studies, most being transient and mild-to-moderate in intensity. Incidence of hepatic AEs was higher in teriflunomide groups; however, serious hepatic AEs were similar across groups (∼2–3%). Hair thinning was higher in teriflunomide than placebo groups, but typically resolved on treatment without intervention and led to discontinuation in <2% of patients. No structural or functional abnormalities were reported in 42 newborns from teriflunomide-exposed parents.ConclusionsThese data from >6800 patient-years of teriflunomide exposure were consistent with individual studies and no new, unexpected safety signals were observed. (Study supported by Genzyme, a Sanofi company).


2017 ◽  
Vol 1 ◽  
pp. s82
Author(s):  
Steven Dayan ◽  
Patricia Ogilvie ◽  
Alexander Z Rivkin ◽  
Steven G Yoelin ◽  
Julie K Garcia ◽  
...  

Abstract Not Available Disclosures: Study supported by Allergan.


BMJ Open ◽  
2021 ◽  
Vol 11 (5) ◽  
pp. e047993
Author(s):  
Nirosen Vijiaratnam ◽  
Christine Girges ◽  
Grace Auld ◽  
Marisa Chau ◽  
Kate Maclagan ◽  
...  

IntroductionParkinson’s disease (PD) is a common neurodegenerative disorder with substantial morbidity. No disease-modifying treatments currently exist. The glucagon like peptide-1 receptor agonist exenatide has been associated in single-centre studies with reduced motor deterioration over 1 year. The aim of this multicentre UK trial is to confirm whether these previous positive results are maintained in a larger number of participants over 2 years and if effects accumulate with prolonged drug exposure.Methods and analysisThis is a phase 3, multicentre, double-blind, randomised, placebo-controlled trial of exenatide at a dose of 2 mg weekly in 200 participants with mild to moderate PD. Treatment duration is 96 weeks. Randomisation is 1:1, drug to placebo. Assessments are performed at baseline, week 12, 24, 36, 48, 60, 72, 84 and 96 weeks.The primary outcome is the comparison of Movement Disorders Society Unified Parkinson’s Disease Rating Scale part 3 motor subscore in the practically defined OFF medication state at 96 weeks between participants according to treatment allocation. Secondary outcomes will compare the change between groups among other motor, non-motor and cognitive scores. The primary outcome will be reported using descriptive statistics and comparisons between treatment groups using a mixed model, adjusting for baseline scores. Secondary outcomes will be summarised between treatment groups using summary statistics and appropriate statistical tests to assess for significant differences.Ethics and disseminationThis trial has been approved by the South Central-Berkshire Research Ethics Committee and the Health Research Authority. Results will be disseminated in peer-reviewed journals, presented at scientific meetings and to patients in lay-summary format.Trial registration numbersNCT04232969, ISRCTN14552789.


2021 ◽  
Vol 22 (1) ◽  
Author(s):  
Michael Ament ◽  
Kathleen Day ◽  
Virginia L Stauffer ◽  
Vladimir Skljarevski ◽  
Mallikarjuna Rettiganti ◽  
...  

Abstract Background Galcanezumab, a humanized monoclonal antibody that binds calcitonin gene-related peptide, has demonstrated a significant reduction in monthly migraine headache days compared with placebo. Here, we analyze data from 3 randomized clinical trials (2 episodic trials [EVOLVE-1, EVOLVE-2] and 1 chronic trial [REGAIN]), to examine if galcanezumab also alleviates the severity and symptoms of migraine. Methods The episodic migraine trials were 6-month, double-blind studies in patients with episodic migraine (4–14 monthly migraine headache days). The chronic migraine trial was a 3-month, double-blind study in patients with chronic migraine (≥ 15 headache days per month, where ≥ 8 met criteria for migraine). Patients (18–65 years) were randomized to placebo or galcanezumab 120 mg with a 240-mg loading dose or 240 mg. Patients recorded headache characteristics, duration, severity, and presence of associated symptoms with each headache. The outcomes analyzed were changes from baseline in number of monthly migraine headache days with nausea and/or vomiting, photophobia and phonophobia, aura, and prodromal symptoms other than aura. Additional outcomes analyzed included the number of moderate-to-severe monthly migraine headache days, number of severe migraine headache days, and mean severity of remaining migraine headache days. Change from baseline in the proportion of days with nausea and/or vomiting and the proportion of days with photophobia and phonophobia among the remaining monthly migraine headache days were also analyzed. Results Galcanezumab was superior to placebo in reducing the frequency of migraine headache days with associated symptoms of migraine such as nausea and/or vomiting, photophobia and phonophobia, and prodromal symptoms. Galcanezumab reduced the frequency of migraine headache days with aura in the episodic migraine studies. There was a significant reduction in the proportion of remaining migraine headache days with nausea and/or vomiting for the episodic and chronic migraine studies, and with photophobia and phonophobia for the episodic migraine studies. Galcanezumab was superior to placebo in reducing the number of monthly moderate-to-severe migraine headache days and the overall and monthly severe migraine headache days. Conclusions Galcanezumab reduces the frequency of migraine headache days and can alleviate potentially disabling non-pain symptoms on days when migraine is present in patients with episodic or chronic migraine. Trial registration NCT, NCT02614183 (EVOLVE-1), registered 25 November 2015; NCT, NCT02614196, (EVOLVE-2), registered 25 November 2015; NCT, NCT02614261 (REGAIN), registered 25 November 2015.


Author(s):  
Richard G Wunderink ◽  
Antoine Roquilly ◽  
Martin Croce ◽  
Daniel Rodriguez Gonzalez ◽  
Satoshi Fujimi ◽  
...  

Abstract Background Hospital-acquired bacterial pneumonia (HABP) and ventilator-associated bacterial pneumonia (VABP) are associated with high mortality rates. We evaluated the efficacy and safety of tedizolid (administered as tedizolid phosphate) for treatment of gram-positive ventilated HABP/VABP. Methods In this randomized, noninferiority, double-blind, double-dummy, global phase 3 trial, patients were randomized 1:1 to receive intravenous tedizolid phosphate 200 mg once daily for 7 days or intravenous linezolid 600 mg every 12 hours for 10 days. Treatment was 14 days in patients with concurrent gram-positive bacteremia. The primary efficacy end points were day 28 all-cause mortality (ACM; noninferiority margin, 10%) and investigator-assessed clinical response at test of cure (TOC; noninferiority margin, 12.5%) in the intention-to-treat population. Results Overall, 726 patients were randomized (tedizolid, n = 366; linezolid, n = 360). Baseline characteristics, including incidence of methicillin-resistant Staphylococcus aureus (31.3% overall), were well balanced. Tedizolid was noninferior to linezolid for day 28 ACM rate: 28.1% and 26.4%, respectively (difference, –1.8%; 95% confidence interval [CI]: –8.2 to 4.7). Noninferiority of tedizolid was not demonstrated for investigator-assessed clinical cure at TOC (tedizolid, 56.3% vs linezolid, 63.9%; difference, –7.6%; 97.5% CI: –15.7 to 0.5). In post hoc analyses, no single factor accounted for the difference in clinical response between treatment groups. Drug-related adverse events occurred in 8.1% and 11.9% of patients who received tedizolid and linezolid, respectively. Conclusions Tedizolid was noninferior to linezolid for day 28 ACM in the treatment of gram-positive ventilated HABP/VABP. Noninferiority of tedizolid for investigator-assessed clinical response at TOC was not demonstrated. Both drugs were well tolerated. Clinical Trials Registration NCT02019420.


Blood ◽  
2021 ◽  
Author(s):  
Glen Kennedy ◽  
Siok-Keen Tey ◽  
Luke Buizen ◽  
Antiopi Varelias ◽  
Kate H. Gartlan ◽  
...  

We determined the efficacy of tocilizumab (TCZ) in preventing grade II-IV acute GVHD (aGVHD) in patients with acute-leukemia or myelodysplasia undertaking matched-related-sibling (MSD) or volunteer-unrelated-donor (VUD) allogeneic-SCT after myeloablative or reduced-intensity conditioning across five Australian centers. 145 patients (50 MSD, 95 VUD) were randomly assigned to placebo or TCZ on day-1. All patients received T-cell-replete PBSC grafts and GVHD-prophylaxis with cyclosporin/methotrexate. A planned sub-study analyzed the VUD cohort. With a median follow up of 746 days, the incidence of grade II-IV aGVHD at day 100 for the entire cohort was 36% versus 27% for placebo versus TCZ (HR 0.69; CI:0.38-1.26, p=0.23) and 45% versus 32% (HR 0.61; CI:0.31-1.22, p=0.16) for the VUD subgroup. The incidence of grade II-IV aGVHD at day 180 for the entire cohort was 40% versus 29% for placebo versus TCZ (HR 0.68; CI:0.38-1.22, p=0.19) and 48% versus 32% (HR 0.59; CI:0.30-1.16, p=0.13) for the VUD subgroup. Reductions in aGVHD were predominantly in grade II disease. A trend to improved aGVHD-free survival (aGVHD-FS) was noted in the TCZ-treated VUD subgroup, 52% versus 68% for placebo versus TCZ (p=0.13). For the entire cohort, transplant-related-mortality occurred in 8% versus 11% of placebo versus TCZ-treated patients respectively (p=0.56) and overall-survival was 79% versus 71% (p=0.27). Median day to neutrophil and platelet engraftment was delayed by 2-3 days in TCZ-treated patients while liver toxicity and infectious complications were similar between groups. In this phase-3 randomized, double-blind trial (ACTRN12614000266662), tocilizumab showed non-significant trends to reduced incidence of grade II-IV aGVHD and improved aGVHD-FS in recipients of HLA-matched VUD donors, but no improvements in long term-survival.


2020 ◽  
Vol 15 (1) ◽  
Author(s):  
Amy S. Paller ◽  
◽  
John Browning ◽  
Milos Nikolic ◽  
Christine Bodemer ◽  
...  

Abstract Background Epidermolysis bullosa (EB) is a rare genetic disorder that manifests as blistering and/or skin erosion. There is no approved treatment for EB; current standard of care consists of wound and pain management. SD-101 6% is a topical cream containing 6% allantoin that was developed for treating skin lesions in patients with EB. The aim of this phase 3, multicenter, randomized, double-blind, vehicle-controlled study was to assess the efficacy and safety of SD-101 6% cream versus vehicle (0% allantoin) on lesions in patients with EB. Methods Eligible patients were ≥1 month old, had a diagnosis of EB (simplex, recessive dystrophic, or intermediate junctional) and a target wound 10–50 cm2 in size that was present for ≥21 days. Patients were randomly assigned to SD-101 6% cream or vehicle, which was applied topically once a day to the entire body for 3 months. Primary efficacy endpoints were time to complete target wound closure within 3 months and the proportion of patients who experienced complete target wound closure within 3 months. Post hoc subgroup analyses were conducted by patient age and in those with body surface area index of total body wound burden ≥5% at baseline. Results In total, 169 patients were enrolled and randomly assigned to SD-101 6% (n = 82) or vehicle (n = 87). Baseline demographics and disease characteristics were similar between treatment groups. There were no statistically significant differences between treatment groups in time to target wound closure (hazard ratio, 1.004; 95% confidence interval [CI] 0.651, 1.549; P = 0.985) or proportion of patients with complete target wound closure within 3 months (odds ratio [95% CI], 0.733 [0.365, 1.474]; nominal P = 0.390). A positive trend toward faster wound closure with SD-101 6% versus vehicle was observed in patients aged 2 to <12 years and those with total body wound burden ≥5% at baseline. SD-101 6% cream was well tolerated. Conclusions SD-101 6% cream for treatment of EB-associated lesions was not more effective than vehicle in shortening the time to complete target wound closure or achieving complete target wound closure within 3 months. Trial registration ClinicalTrials.gov, NCT02384460; Date of trial registration, February 13, 2015; First participant enrolled, March 11, 2015.


Sign in / Sign up

Export Citation Format

Share Document