Clinical Trials Logo

Clinical Trial Details — Status: Completed

Administrative data

NCT number NCT01992900
Other study ID # ST3073-ST3074-DM-12-002
Secondary ID 2013-002255-15
Status Completed
Phase Phase 2
First received November 11, 2013
Last updated November 4, 2016
Start date November 2013
Est. completion date January 2016

Study information

Verified date November 2016
Source sigma-tau i.f.r. S.p.A.
Contact n/a
Is FDA regulated No
Health authority Burkina Faso: Ministry of HealthGambia: Department of State for Health and Social WelfareMozambique: Ministry of Health (MISAU)Tanzania: Food & Drug AdministrationCongo, Democratic Republic of the: Ministry of Health
Study type Interventional

Clinical Trial Summary

There is a need for paediatric formulations that permit accurate dosing and enhance patient compliance. However, for the treatment of malaria, scarce paediatric-friendly formulations are available on the market. Thus, a new water dispersible formulation of eurartesim has been developed for oral administration.

Aim of this study is to provide data on pharmacokinetic profile, safety and efficacy of this new paediatric formulation and compare it with the crushed film coated tablet in infant patients (6 to ≤12 months of age) suffering from uncomplicated Plasmodium falciparum malaria.

Furthermore, a Pharmacokinetic/Pharmacodynamic(PK/PD) modelling will be built up to establish PK/PD relationship in adult and paediatric populations.


Description:

Although the significant advances made during the last decades in controlling malaria in Africa, morbidity and mortality in sub-Saharan countries remain substantial. It is estimated that around 655.000 deaths a year still occur due to malaria infection and the majority of such deaths occur among young African children.

In response to the emergence and spread of classical drug-resistant Plasmodia strains, the WHO recommends since 2004 the use of artemisinin-based combination therapies (ACTs) in the treatment of uncomplicated malaria episodes.

The artemisinin derivatives are currently the most rapidly acting and potent antimalarial drugs.

Eurartesim is a fixed-dose combination product composed of dihydroartemisinin (DHA) and piperaquine phosphate (PQP). This second compound assures the long-term efficacy of eurartesim completing the whole body cleaning from the parasites. Eurartesim appears to offer benefits over existing licensed malaria treatments and is in line with current WHO treatment policy recommendations.

Eurartesim obtained a centralized marketing authorization by the European Union as film coated tablets containing 160 mg PQP/20 mg DHA and 320 mg PQP/40 mg DHA. The drug, licensed for its use in children (above 6 months of age) and adults has been administered in infants (above 6 months) and young children by crushing the tablets and administering them with a small amount of water.

According to the Guidelines on Clinical Investigation of Medicinal Products in the Paediatric Population (EMA ICH Topic E 11), there is a need for paediatric formulations that permit accurate dosing and enhance patient compliance.

However, for the treatment of malaria, scarce paediatric-friendly formulations are available on the market, and this is a particularly blatant problem as young children carry the brunt of the malaria burden. Thus, a new water dispersible formulation of eurartesim has been developed for oral administration, since liquid formulations may be needed or desirable for paediatric patients of smaller ages due to their inability to swallow tablets. Moreover, in order to increase paediatric compliance to treatment, the new formulation is prepared with acceptable flavour and sweetener for children.

Eurartesim is a promising effective ACT treatment for malaria. It provides a simple dosing scheme (a single daily dose over 3 days) and it does not need any concomitant administration of food to improve its absorption. Moreover, eurartesim offers an interesting post-treatment prophylactic effect following therapy, reducing the risk of new infection, an issue of particular relevance in highly endemic malaria countries.


Recruitment information / eligibility

Status Completed
Enrollment 300
Est. completion date January 2016
Est. primary completion date July 2015
Accepts healthy volunteers No
Gender Both
Age group 6 Months to 12 Months
Eligibility Inclusion Criteria:

- Male and Female infants aged from 6 months to = 12 months included.

- Ability to swallow oral suspension.

- Body weight >5 kg.

- Uncomplicated malaria, with microscopically confirmed mono-infection by P. falciparum (parasitaemia =1000/microL and <200000/microL).

- History of fever anytime during the preceding 48 hours or presence of fever (axillary temperature =37.5 °C or =38.0 °C rectally).

- Ability of parents or guardians to understand the nature of the trial and providing signed informed consent.

- Stable residence in the study area during the two months after recruitment and willingness to comply with the study protocol and the study visit schedule.

Exclusion Criteria:

- Antimalarial treatment with amodiaquine, chloroquine, quinine or lumefantrine-based compounds within the previous 6 weeks, with piperaquine-based compound, or mefloquine, or sulphadoxine pyrimethamine within the previous 3 months and with halofantrine within the 30 days prior to screening.

- Any other antimalarial treatment or antibiotics with antimalarial activity (including cotrimoxazol) and any herbal products, within the 7 days prior to screening.

- Severe malnutrition (defined as weight for height <70% of the median National Center for Health Statistics(NCHS)/WHO reference).

- Severe vomiting or dehydration.

- Presence of jaundice.

- Known hypersensitivity to the artemisinin-based therapy or piperaquine.

- History of relevant clinical allergic reaction of any origin.

- Clinical and/or laboratory features of severe malaria.

- Known moderate/ severe renal or liver insufficiency.

- Evidence of clinically relevant haematological, pulmonary, metabolic-endocrine, neurological, urogenital diseases as judged by the investigator.

- Already diagnosed HIV infection, hepatitis B virus (HBV) or hepatitis C virus (HCV) infection.

- Previous admission for, or evidence of symptomatic cardiac arrhythmias or with clinically relevant bradycardia at screening (bpm < 90).

- Family history of sudden death, or known congenital prolongation of the QT interval, or any clinical condition known to prolong the QT interval.

- ECG abnormality that requires urgent management.

- Any treatment which can induce a lengthening of QT interval.

- Gastrointestinal dysfunction that could alter absorption or motility (i.e. malabsorption syndromes, intestinal sub-occlusion or previous major gastrointestinal surgery).

- Any contraindication to blood sampling.

- Moderate and severe anaemia (Hb < 7 g/dL).

- Patients who have used any drugs or substances known to be strong inhibitors of Cytochrome P enzymes (formerly known as cytochrome P450 enzymes) within 10 days prior to screening.

- Patients who have used any drugs or substances known to be strong inducers of CYP enzymes (formerly known as cytochrome P450 enzymes)within 28 days prior to screening.

- Children lactated by HIV positive women who are undergoing treatment with antiretroviral drugs.

- Participation in any investigational drug study during the 30 days prior to screening or previously randomised in the present trial.

Study Design

Allocation: Randomized, Endpoint Classification: Pharmacokinetics/Dynamics Study, Intervention Model: Parallel Assignment, Masking: Open Label, Primary Purpose: Treatment


Related Conditions & MeSH terms


Intervention

Drug:
Eurartesim dispersible oral tablet
The first dose of Eurartesim dispersible oral tablet will be administered immediately after randomization. the other two doses will be administered with an interval of 24 hours. Eurartesim tablet will be dispersed in about 10 mL of water.
eurartesim film coated tablet
The first dose of Eurartesim film coated tablet will be administered immediately after randomization. the other two doses will be administered with an interval of 24 hours. tablet will be crushed and dispersed in a few amount of water (about 10 ml).

Locations

Country Name City State
Burkina Faso Centre Muraz Bobo Dioulasso
Burkina Faso Centre National de Recherche et de Formation en Paludisme Ouagadougou
Congo, The Democratic Republic of the Kinshasa School of Public Health, School of Medicine, University of Kinshasa Kinshasa
Gambia Medical Research Council Fajara
Mozambique Manica's Health Research Centre Manica
Tanzania Bagamoyo Research center, Ifakara Heath Institute Bagamoyo
Tanzania National Insititute for Medical Research Tanga

Sponsors (1)

Lead Sponsor Collaborator
sigma-tau i.f.r. S.p.A.

Countries where clinical trial is conducted

Burkina Faso,  Congo, The Democratic Republic of the,  Gambia,  Mozambique,  Tanzania, 

Outcome

Type Measure Description Time frame Safety issue
Primary Comparison of peak plasma concentration of Dihydroartemisinin in the two studied formulations In order to minimize the blood sampling in infants, patients will be divided into 10 blood sampling groups for both the treatments (water dispersible and crushed film coated tablet) and only 2 blood draws per group will be taken within the prespecified timepoints.
Optimal sampling design was performed by using the appropriate software. The D-optimal design option was used to determine the optimal sampling design.
DHA plasma samples will be collected on the first day of study drug administration at 20, 30, 40, 60, 100, 200 and 300 minutes post-dose No
Primary Comparison of area under the plasma concentration versus time curve of Dihydroartemisinin in the two studied formulations. In order to minimize the blood sampling in infants, patients will be divided into 10 blood sampling groups for both the treatments (water dispersible and crushed film coated tablet) and only 2 blood draws per group will be taken within the pre-specified timepoints.
Optimal sampling design was performed by using the appropriate software. The D-optimal design option was used to determine the optimal sampling design.
DHA plasma samples will be collected on the first day of study drug administration at 20, 30, 40, 60, 100, 200 and 300 minutes post-dose No
Primary Comparison of peak plasma concentration of Piperaquine in the two studied formulations In order to minimize the blood sampling in infants, patients will be divided into 10 blood sampling groups for both the treatments (water dispersible and crushed film coated tablet) and only 2 blood draws per group will be taken within the pre-specified timepoints.
Optimal sampling design was performed by using the appropriate software. The D-optimal design option was used to determine the optimal sampling design.
PQP plasma samples will be collected on day 0 of treatment at 40, 80, 280, 720 minutes, then on day 1 at 24 hours and on day 2 at 40, 80, 280, 720 minutes after the last drug administration and then 24, 120, 288 and 456 hours No
Primary Comparison of area under the plasma concentration versus time curve of Piperaquine in the two studied formulations. In order to minimize the blood sampling in infants, patients will be divided into 10 blood sampling groups for both the treatments (water dispersible and crushed film coated tablet) and only 2 blood draws per group will be taken within the pre-specified timepoints.
Optimal sampling design was performed by using the appropriate software. The D-optimal design option was used to determine the optimal sampling design.
PQP plasma samples will be collected on day 0 of treatment at 40, 80, 280, 720 minutes, then on day 1 at 24 hours and on day 2 at 40, 80, 280, 720 minutes after the last drug administration and then 24, 120, 288 and 456 hours No
Secondary Parasite clearance time Blood film for parasite count will be read by two independent microscopists. At screening and then about every 12 hours until the time of the first negative result, then confirmed by a second negative result or up to three days No
Secondary Fever clearance time Body temperature will be recorded to collect information about the fever clearance time At screening and then about every 12 hours until the time on which body temperature falls down below 37.5 °C, or up to three days No
Secondary Change from baseline of electrocardiographic QT interval at 4-6 hours after the last study drug intake Triplicate ECGs will be undertaken on screening (before study drug administration)as well as 4-6 hours after the last drug administration.
The triplicate ECG values will be averaged in order to obtain one single value per patient and time point. These averages will be used for the statistical analysis.
From the collected values the heart rate corrected QT intervals will be derived according to Fridericia's correction (QTcF)
Before randomization and then at 4-6 hours after the last dose of study drug. No
Secondary Blood chemistry: proportion of patients with deterioration of parameters at day 7 respect to screening. The evaluated analytes are: Blood Urea Nitrogen, Creatinine, Glucose, Alanine aminotransferase, aspartate aminotransferase, Total Bilirubin, electrolytes (Na+, K+ and Cl-) screening and Day 7 No
Secondary Hematology: proportion of patients with deterioration of parameters at day 7 respect to screening the following parameters are evaluated: Hemoglobin, hematocrit and full blood counts including Red Blood Cell and differential White Blood Cells, Platelet Count screening and day 7 No
Secondary Adverse Events occurrence to calculate percentage of patients experiencing Adverse Events and Serious Adverse Events during all the study period from randomization and up to day 42 No
See also
  Status Clinical Trial Phase
Terminated NCT04130282 - VAC077: Safety and Immunogenicity of the Pfs25-IMX313/Matrix-M Vaccine Phase 1
Completed NCT04049916 - Pyronaridine-artesunate With Low Dose Primaquine for Preventing P. Falciparum Transmission Phase 2/Phase 3
Active, not recruiting NCT03814616 - Pyramax in Asymptomatic Carriers of P. Falciparum Monoinfections Phase 2
Active, not recruiting NCT04079621 - Short Course Radical Cure of P. Vivax Malaria in Nepal Phase 4
Completed NCT05135273 - Study of the Transmission-Blocking Vaccine Pfs230D1-EPA/Matrix-M Against Malaria in Adults in Mali Phase 1
Not yet recruiting NCT06083688 - Preventing Malaria in School Children to Protect the Whole Community in Rural Blantyre District, Malawi Phase 4
Recruiting NCT03511443 - Evaluation of the Performance of a hsRDT Versus cRDT in Reactive Case Detection of Malaria Infections N/A
Completed NCT05550909 - Gametocytocidal and Transmission-blocking Efficacy of ASAQ and ALAQ With or Without PQ in Mali Phase 2
Recruiting NCT05306067 - Plasmodium Falciparum Genomic Intelligence in Mozambique
Completed NCT05081089 - Gametocytocidal and Transmission-blocking Efficacy of PQ in Combination With AL and TQ in Combination With SPAQ in Mali Phase 2
Recruiting NCT05150808 - Vectron T500 (Broflanilide 50WP) for IRS in Tanzania Tanzania Phase 3
Recruiting NCT05757167 - Improving Neonatal Health Through Rapid Malaria Testing in Early Pregnancy With High-Sensitivity Diagnostics Phase 4
Completed NCT04565184 - Effectiveness and Safety of Artesunate-Amodiaquine and Artemether-Lumefantrine for the Treatment of Malaria in Yaounde Phase 4
Completed NCT03896724 - Safety, Immunogenicity and Efficacy of R21 Matrix-M in 5-17 Month Old Children in Nanoro, Burkina Faso Phase 1/Phase 2
Completed NCT03454048 - Controlled Human Malaria Infection Model for Evaluation of Transmission-blocking Interventions - Study 2 N/A
Recruiting NCT04844905 - Adjunctive Ivermectin Mass Drug Administration for Malaria Control Phase 3
Completed NCT03138096 - Safety and Protective Efficacy of Pb(PfCS@UIS4) Phase 1/Phase 2
Recruiting NCT04271306 - Safety, Immunogenicity and ex Vivo Efficacy of Pfs25-IMX313/Matrix-M in Healthy Volunteers in Bagamoyo, Tanzania. Phase 1
Recruiting NCT05058885 - Plasmodium Vivax Among Duffy Negative Population in Cameroon.
Completed NCT04862416 - Safety and Efficacy of R0.6C Vaccine Phase 1