Clinical Trials Logo

Clinical Trial Details — Status: Enrolling by invitation

Administrative data

NCT number NCT04056325
Other study ID # 3
Secondary ID
Status Enrolling by invitation
Phase Phase 2
First received
Last updated
Start date November 27, 2019
Est. completion date December 1, 2021

Study information

Verified date March 2021
Source Swiss Tropical & Public Health Institute
Contact n/a
Is FDA regulated No
Health authority
Study type Interventional

Clinical Trial Summary

This study is a phase 2, blinded and randomized clinical trial. The phase 2a trial is single blinded and conducted in Lao, while the phase 2b trial is double-blinded and conducted in Lao and Cambodia. The study aims at providing evidence on effective doses and safety of moxidectin in adults against infection with S. stercoralis in Laos (trial 2a) and efficacy and safety of moxidectin compared to ivermectin in adults against infection with S. stercoralis in Laos and Cambodia (trial 2b). The efficacy of the treatment will be assessed by collecting three stool samples once pre-treatment and once 21 days post-treatment. The stool samples will be analyzed by a quantitative Baermann assay.


Description:

This is a phase 2a single-blinded and a phase 2b double-blinded randomized clinical trial, which aims to determine efficacy and safety of (2a) seven ascending oral moxidectin dosages in 210 adults infected with S. stercoralis, namely placebo, 2 mg, 4 mg, 6 mg, 8 mg, 10 mg, 12 mg in Lao and (2b) the recommended dose moxidectin (i.e. the most promising dosage identified in trial A; between 2-12 mg) in comparison to the standard treatment dose of ivermectin (200 µg/kg) in 245 adults infected with S. stercoralis in Lao and Cambodia. Embedded in the trial is a pharmacokinetic/-dynamic study with the goal to measure moxidectin disposition in adults and to determine population pharmacokinetic (PK) parameters of the optimal dose of moxidectin in the treatment of S. stercoralis. The primary objective is to determine the dose-response of moxidectin based on cure rates (CR) against S. stercoralis and to quantify the efficacy of the recommended dose to the standard treatment (ivermectin) in adults. The secondary objectives of the trial are: Evaluation of the safety and tolerability of the dose-dependent treatment regimes, evaluation of the safety and tolerability of moxidectin compared to ivermectin, comparison of the larvae reduction rate (LRR) of the different treatment regimens against S. stercoralis (trial 2a,b), determination of an exposure- (including Cmax, area under curve (AUC) and tmax) -response correlation of moxidectin in adults, comparison of the exposure-response of moxidectin using venous and capillary blood, evaluation of the cure rate of the different moxidectin treatment regimens against co-infection and the determination of the population PK parameters of the optimal dose of moxidectin in the treatment of S. stercoralis. After obtaining informed consent from each individual, the medical history of the participants will be assessed with a standardized questionnaire, in addition to a clinical examination carried out by the study physician before treatment. Enrollment will be based on collection and analysis by a quantitative Baermann method (in duplicates) of three stool samples. Randomization of participants into the different treatment arms will be stratified according to intensity of infection. The adults will also be interviewed before treatment, 3 and 24 hours as well as 21 days after treatment about the occurrence of adverse events (AE). The efficacy of the treatment will be determined 21 days post-treatment by collecting another three stool samples. All stool samples will be examined with duplicate Baermann assays recorded quantitatively. Co-infection with T. trichiura, A. lumbricoides and hookworm infection will be identified using duplicate Kato-Katz thick smears on stool samples. A subsample of adults will further be sampled using finger pricking for micro sampling at 0, 2, 4, 8, 24, and 72 hours, 7 and 21 days post treatment to evaluate pharmacokinetic parameters (trial phase 2a) and at defined time windows, that are based on the PK model earned from trial 2a (trial phase 2b). For validation of the analytical method the subsample of one study arm (8 mg, trial phase 2a) will undergo venous blood sampling in addition to finger pricking. An available case analysis (full analysis set according to the intention to treat principle) will be performed, including all subjects with primary end point data. Supplementary, a per-protocol analysis will be conducted. CRs will be calculated as the percentage of larvae-positive subjects at baseline who become larvae-negative after treatment. Larvae per gram (LPG) stool sample will be assessed by calculating the mean of the larvae counts from the three duplicate Baermann assays and divided by the mean weighted amount of these stool samples. The LRR will be calculated following: (LRR = (1-(mean at follow-up/mean at baseline))*100) Geometric and arithmetic mean larvae counts will be calculated for the different treatment arms before and after treatment to assess the corresponding LRRs. Bootstrap resampling method with 2,000 replicates will be used to calculate 95% confidence intervals (CIs) for LRRs. Emax models using the dose finding package of the statistical software environment R will be implemented to predict the dose-response curves in terms of CRs and LRRs.


Recruitment information / eligibility

Status Enrolling by invitation
Enrollment 210
Est. completion date December 1, 2021
Est. primary completion date September 15, 2021
Accepts healthy volunteers No
Gender All
Age group 18 Years to 60 Years
Eligibility Inclusion Criteria: - Adults (= 18 years) infected with S. stercoralis - Absence of major systemic illnesses - Written informed consent signed by individual Exclusion Criteria: - Any abnormal medical conditions or chronic disease - Negative diagnostic result for S. stercoralis - No written informed consent by individual. - Pregnant and lactating women. - Recent use of anthelmintic drug (within past 4 weeks), attending other clinical trials during the study - Known allergy to study medications (i.e. moxidectin, ivermectin) - Currently taking medications with known interaction (i.e. for warfarin)

Study Design


Related Conditions & MeSH terms


Intervention

Drug:
Moxidectin
Monotherapy, oral administration, single dose, fixed dose
Ivermectin
Monotherapy, oral administration, single dose, weight dependent
Placebo oral tablet
Monotherapy, oral administration, single dose, matching number of tablets

Locations

Country Name City State
Cambodia National Centre for Parasitology, Entomology and Malaria Control Phnom Penh
Lao People's Democratic Republic National Institute of Public Health Vientiane

Sponsors (3)

Lead Sponsor Collaborator
Jennifer Keiser National Centre for Parasitology, Entomology and Malaria Control, Cambodia, National Institute of Public Health, Vientiane, Laos

Countries where clinical trial is conducted

Cambodia,  Lao People's Democratic Republic, 

Outcome

Type Measure Description Time frame Safety issue
Primary Cure rate against Strongyloidiasis stercoralis The conversion from being larvae positive pre-treatment to larvae negative post-treatment, or cure rate (CR). 14-21 days after treatment
Secondary Larvae-reduction rate (LRR) against Strongyloidiasis stercoralis Larvae per gram (LPG) stool sample will be assessed by calculating the mean of the larvae counts from the three duplicate Baermann assays and divided by the mean weighted amount of these stool samples. The LRR will be calculated following: (LRR = (1-(mean at follow-up/mean at baseline))*100) 14-21 days after treatment
Secondary CRs and LRRs against concomitant soil-transmitted helminth infections CRs and LRRs will be calculated for T. trichiura, A. lumbricoides and hookworm infections as described in primary and secondary outcome. 14-21 days after treatment
Secondary Number of participants reporting adverse events Subjects will be kept for observation for at least 3 hours following treatment for any acute AEs. If there is any abnormal finding, the local study physician will perform a full clinical, physical and biochemical examination and findings will be recorded. An emergency kit will be available on site to treat any medical conditions that warrant urgent medical intervention. In addition patients will also be interviewed 3 and 24 hours and again 3 weeks after treatment about the occurrence of AEs. A standardized symptom questionnaire is used, that includes the recording of headache, abdominal pain, itching, nausea, vomiting, diarrhea, allergic reaction as well as any further mentioned event by the participant. 14-21 days after treatment
Secondary Maximum concentration (Cmax) of moxidectin in adults Upon oral intake moxidectin levels in blood will be quantified with time using a micro sampling device. Moxidectin will be quantified using a validated liquid chromatography tandem mass spectrometry (LC-MS/MS) method. Drug concentrations will be calculated by interpolation from a calibration curve with a foreseen limit of quantification of approximately 5 ng/ml. The PK analysis will be undertaken fitting a structural compartmental PK model. 0, 2, 4, 8, 24, and 72 hours, 7 and 21 days post treatment
Secondary Time to reach Cmax (tmax) of moxidectin in adults Upon oral intake moxidectin levels in blood will be quantified with time using a micro sampling device. Moxidectin will be quantified using a validated liquid chromatography tandem mass spectrometry (LC-MS/MS) method. Drug concentrations will be calculated by interpolation from a calibration curve with a foreseen limit of quantification of approximately 5 ng/ml. The PK analysis will be undertaken fitting a structural compartmental PK model. 0, 2, 4, 8, 24, and 72 hours, 7 and 21 days post treatment
Secondary Area under the curve (AUC) of moxidectin in adults Upon oral intake moxidectin levels in blood will be quantified with time using a micro sampling device. Moxidectin will be quantified using a validated liquid chromatography tandem mass spectrometry (LC-MS/MS) method. Drug concentrations will be calculated by interpolation from a calibration curve with a foreseen limit of quantification of approximately 5 ng/ml. The PK analysis will be undertaken fitting a structural compartmental PK model. 0, 2, 4, 8, 24, and 72 hours, 7 and 21 days post treatment
Secondary Elimination half life (t1/2) of moxidectin in adults Upon oral intake moxidectin levels in blood will be quantified with time using a micro sampling device. Moxidectin will be quantified using a validated liquid chromatography tandem mass spectrometry (LC-MS/MS) method. Drug concentrations will be calculated by interpolation from a calibration curve with a foreseen limit of quantification of approximately 5 ng/ml. The PK analysis will be undertaken fitting a structural compartmental PK model. 0, 2, 4, 8, 24, and 72 hours, 7 and 21 days post treatment
See also
  Status Clinical Trial Phase
Completed NCT04999774 - Evaluation of Strongyloidiasis in Ecuador: a fieLd Laboratory Accuracy Study
Terminated NCT03605758 - Treatment of Strongyloides Infection Phase 3
Not yet recruiting NCT06373835 - Efficacy and Safety of Emodepside in Adults Infected With Strongyloidiasis Stercoralis Phase 2
Completed NCT06308705 - The SsRI/NIE Recombinant Antigen ELISA for the Follow up of Patients Infected by Strongyloides Stercoralis N/A
Completed NCT04848688 - Efficacy and Safety of Moxidectin Versus Ivermectin Against Strongyloides Stercoralis Phase 3
Recruiting NCT01308268 - Management of Soil-transmitted Helminthiasis and Strongyloidiasis N/A
Not yet recruiting NCT06368609 - Preliminary Evaluation of Two Rapid Diagnostic Test Prototypes for Strongyloides Stercoralis Infection N/A