AGILE (Early Phase Platform Trial for COVID-19)

November 21, 2023 updated by: University of Liverpool

AGILE: Seamless Phase I/IIa Platform for the Rapid Evaluation of Candidates for COVID-19 Treatment

The AGILE platform master protocol allows incorporation of a range of identified and yet-to-be-identified candidates as potential treatments for adults with COVID-19 into the trial. Candidates will be added into the trial via candidate-specific trial (CST) protocols of this master protocol as appendices. Having one master protocol ensures different candidates are evaluated in the same consistent manor and opening up new trials for new candidates is more efficient. Inclusion of new candidates will be determined by the AGILE Scientific Advisory Board based on pre-clinical data, evidence in the clinical setting and GMP capabilities.

Study Overview

Detailed Description

AGILE is a multicentre, multi-arm, multi-dose, multi-stage open-label, adaptive, seamless phase I/II Bayesian randomised platform trial to determine the optimal dose, activity and safety of multiple candidate agents for the treatment of COVID-19.

This study allows for the assessment of many candidates at different doses, with the ability to add candidates as they are identified or drop them as their evaluation is completed. Promising candidates will move to the an external trial for further evaluation in the phase II/III setting.

Each candidate will be evaluated in its own trial, randomising between candidate and control with 2:1 allocation in favour of the candidate. Each dose will be assessed for safety sequentially in cohorts of 6 patients. Once a phase II dose has been identified we will assess efficacy by seamlessly expanding into a larger cohort.

AGILE is completely flexible in that the core design in the master protocol (as has been explained above) can be adapted for each candidate based on prior knowledge of the candidate - i.e. population, primary endpoint and sample size can be amended. This will be detailed in each candidate-specific trial protocol of the master protocol.

Candidate-Specific Trial 2 (CST-2): Open-label 2:1 randomised controlled phase I of EIDD-2801 versus standard of care followed by a 1:1 blinded controlled parallel group Phase II trial of EIDD-2801 versus placebo. A phase I will be carried out to confirm the optimal dose in this group. Following a safety review, EIDD-2801 will be tested for efficacy in a blinded placebo controlled randomised phase II trial.

Candidate-Specific Trial 3 (CST-3A): Multicentre, Adaptive, Phase I trial to Determine the optimal dose, Safety and Efficacy of Nitazoxanide for the Treatment of COVID-19

Candidate-Specific Trial 3 (CST-3B): A Randomized, Multicentre, Seamless, Adaptive, Phase I/II trial to Determine the optimal dose, Safety and Efficacy of Nitazoxanide for the Treatment of COVID-19

Candidate-Specific Trial 5 (CST-5): Randomized, Multicentre, Seamless, Adaptive, Phase I/II Platform Study to Determine the Phase II dose of VIR-7832, and Evaluate the Safety and Efficacy of VIR-7831 and VIR-7832 for the Treatment of COVID-19

Candidate-Specific Trial 6 (CST-6): A Randomized, Multicentre, Seamless, Adaptive, Phase I/II Platform Study to Determine the Phase II dose and to Evaluate the Safety and Efficacy of intravenous Favipiravir for the Treatment of COVID-19

Candidate-Specific Trial 8 (CST-8): A Randomised, Multicentre, Seamless, Adaptive, Phase I Platform Study to Determine the recommended Phase II dose and Evaluate the Safety and Efficacy of antiviral combination of Molnupiravir and Paxlovid® for the Treatment of COVID-19

Study Type

Interventional

Enrollment (Estimated)

600

Phase

  • Phase 2
  • Phase 1

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Contact

Study Locations

      • Cape Town, South Africa
        • Desmond Tutu Health Foundation
      • Johannesburg, South Africa
        • Ezintsha
      • Liverpool, United Kingdom, L7 8XP
        • Liverpool University Hospitals NHS Foundation Trust
      • London, United Kingdom
        • Kings College Hospital NHS Foundation Trust
      • Manchester, United Kingdom
        • Manchester University NHS Foundation Trust
      • Southampton, United Kingdom, SO16 6YD
        • University Hospital Southampton NHS Foundation Trust

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

18 years and older (Adult, Older Adult)

Accepts Healthy Volunteers

No

Description

Master Protocol Inclusion Criteria:

  1. Adults (≥18 years) with laboratory-confirmed* SARS-CoV-2 infection (PCR)
  2. Ability to provide informed consent signed by study patient or legally acceptable representative
  3. Women of childbearing potential (WOCBP) and male patients who are sexually active with WOCBP must agree to use a highly effective method of contraception (as outlined in the protocol) from the first administration of trial treatment, throughout trial treatment and for the duration outlined in the candidate-specific trial protocol after the last dose of trial treatment

    • If any CSTs are included in the community setting, the CST protocol will clarify whether patients with suspected SARS-CoV-2 infection are also eligible.

Standard additional criteria that may be applied per CST protocol:

Group A (severe disease) 4a. Patients with clinical status of Grades 4 (hospitalised, oxygen by mask or nasal prongs), 5 (hospitalised, on non-invasive ventilation, or high flow oxygen), 6 (hospitalised, intubation and mechanical ventilation) or 7 (ventilation and additional organ support - pressors, renal replacement therapy (RRT), extracorporeal membrane oxygenation (ECMO)), as defined by the WHO clinical severity score, 9-point ordinal scale.

Group B (mild-moderate disease) 4b. Ambulant or hospitalised patients with the following characteristics peripheral capillary oxygen saturation (SpO2) >94% RA N.B. The CST protocol inclusion criteria will take precedence over the master protocol inclusion criteria.

CST-2 Inclusion Criteria:

For the purpose of the EIDD-2801 candidate-specific trial the following inclusion criteria have been amended from the Master protocol to:

1. Male or female ≥ 60 years old or ≥50 years old with at least one well controlled comorbidity: cardiovascular disease, chronic lung disease (e.g. COPD, or pulmonary hypertension), immune deficiency (taking the equivalent of 20 mg prednisone daily, chemotherapy, or immune modulating biologic therapies), diabetes (treated with insulin or oral medications), BMI≥30, or hypertension requiring medication with laboratory confirmed SARS-CoV-2 infection (PCR) .

3. Women of childbearing potential (WOCBP) and male patients who are sexually active with WOCBP must agree to use two effective methods of contraception, one of which should be highly effective (as outlined in the protocol). For women, from the first administration of trial treatment, throughout trial and up to 50 days after the last follow up visit (50 days after day 29) and for men with female partners of child bearing potential, from the first administration until 100 days after last follow up visit (100 days after day 29).

4. Group B (mild-moderate disease): Ambulant with the following characteristics peripheral capillary oxygen saturation (SpO2) >94% RA (NB this differs to the Master Protocol which also includes hospitalised patients in this group).

Additional criteria specific to this candidate are:

5. Has signs or symptoms of COVID-19 that began within 5 days of the planned first dose of study drug.

6. Is in generally good health (except for current respiratory infection) and is free of uncontrolled chronic conditions.

7. Is willing and able to comply with all study procedures and attending clinic visits through the 4th week.

8. Has someone, aged ≥ 16 living in the same household during the dosing period.

CST-6 Additional inclusion criteria:

  1. Group A (severe disease). Patients with clinical status of Grades 5 (hospitalised, oxygen by mask or nasal prongs), 6 (hospitalised, on non-invasive ventilation, or high flow oxygen as defined by the WHO Clinical Progression Scale (WHO, 2020)).
  2. Less than or equal to 14 days from onset of COVID-19 symptoms

CST-8 Inclusion Criteria:

  1. For the purpose of CST-8, criteria 1 has been amended from the Master Protocol to:

    Adults (≥18 years) outpatients positive lateral flow test at screening or baseline Day 1, who are within 5 days of symptom onset prior to the planned first dose of study drug.

  2. Criteria 3 has been amended from the Master Protocol to:

Women of childbearing potential (WOCBP) and male participants who are sexually active with WOCBP must agree to use a highly effective method of contraception (as outlined in section 5.5 of the Master Protocol) for the duration of the treatment and for six weeks following the last dose.

Additional criteria specific to CST-8 are:

  • Initial onset of COVID-19 signs/symptoms within 5 days prior to the day of randomisation and at least 1 of the current specified COVID-19 signs/symptoms (listed on the NHS website) present on the day of randomisation
  • Is willing and able to comply with all study procedures and attending clinic visits

Master Protocol Exclusion Criteria:

  1. Alanine aminotransferase (ALT) and/or aspartate aminotransferase (AST) >5 times the upper limit of normal (ULN)
  2. Stage 4 severe chronic kidney disease or requiring dialysis (i.e., estimated glomerular filtration rate <30 mL/min/1.73 m^2)
  3. Pregnant or breast feeding
  4. Anticipated transfer to another hospital which is not a study site within 72 hours
  5. Allergy to any study medication
  6. Patients taking other prohibited drugs (as outline in CST protocol) within 30 days or 5 times the half-life (whichever is longer) of enrolment
  7. Patients participating in another CTIMP trial

    N.B. The CST protocol exclusion criteria will take precedence over the master protocol exclusion criteria.

    CST-2 Exclusion Criteria:

    Additional criteria specific to this candidate are:

  8. Has a febrile respiratory illness that includes signs of pneumonia, or requires hospitalization, oxygenation, mechanical ventilation, or other supportive modalities.
  9. Has a platelet count less than 50x10^9/L, or lymphocytes less than 0.2x10^9/L, haemoglobin less than 10 g/dL, or has a disorder of the hematologic system including anaemic disorder or other blood dyscrasia, cancer of the hematologic system, history of bone marrow transplant, or other significant hematologic disease at screening.
  10. Is experiencing adverse events or laboratory abnormalities that are Grade 3 or above based on the CTCAE scale.
  11. Has clinically significant liver dysfunction or renal impairment.
  12. Has history of Hepatitis C infection or concurrent bacterial pneumonia.
  13. Has received an experimental agent (vaccine, drug, biologic, device, blood product, or medication) within 30 days prior to the first dose of study drug.
  14. In the opinion of the investigator, has significant end-organ disease as a result of relevant comorbidities: chronic kidney disease, congestive heart failure, peripheral vascular disease including diabetic ulcers.
  15. Has a SaO2<95% by oximetry or has lung disease that requires supplemental oxygen.
  16. Has any condition that would, in the opinion of the investigator, put the patient at increased risk for participation in a clinical study.

CST-8 Exclusion Criteria:

For the purpose of the combination CST8 candidate-specific trial the following exclusion criteria also apply:

  1. Swallowing difficulties
  2. Known medical history of active liver disease
  3. Receiving dialysis or have known moderate to severe renal impairments (defined as CKD stage 4 or 5 or current acute kidney injury or most recent eGFR in the past 6 months <30 ml/min/1.73m2)
  4. Currently taking Paxlovid® or molnupiravir at time of screening
  5. Oxygen saturation of <92% on room air, or on their standard home oxygen supplementation
  6. Taking a drug which would put subject at unacceptable risk due to interaction or is contraindicated as per SPC for each IMP

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

  • Primary Purpose: Treatment
  • Allocation: Randomized
  • Interventional Model: Sequential Assignment
  • Masking: Quadruple

Arms and Interventions

Participant Group / Arm
Intervention / Treatment
No Intervention: CST-2 Control
Phase 1b only (standard of care)
Placebo Comparator: CST-2 Placebo
Phase II placebo blinded controlled
CST-2 Phase II: Placebo will be administered orally, twice daily (BID) for 10 doses (5 or 6 days).
Other Names:
  • Placebo
Experimental: CST-3A Nitazoxanide
Phase Ia Nitazoxanide will be administered orally, initially twice daily (BID) for 14 doses (7 days). The starting dose will be 1500mg BID based on existing dose information, but dose adaptations may occur

CST3A & CST3B Phase I: Nitazoxanide will be administered orally, initially twice daily (BID) for 14 doses (7 days). The starting dose will be 1500mg BID based on existing dose information, but dose adaptations may occur.

Phase II: As per Phase Ib, with the dose determined by the recommended phase II dose.

No Intervention: CST3B Control
Standard of care
Experimental: CST6 IV Favipiravir
IV Favipiravir twice daily for 7 days. Starting dose 600 mg twice daily. Dose escalation to 1200 mg twice daily, 1800 twice daily, 2400 twice daily.
CST-6: Multiple doses of IV Favipiravir will be administered by intravenous (IV) infusion over 1 hour. Dosing regimen will be every 12 hours for 7 days duration. The starting dose will be 600mg (BID), and dose escalations to 1200mg (BID), 1800mg (BID) and 2400mg (BID) are anticipated as well as a de-escalation dose of 300mg (BID) if necessary, with de-escalation and escalation guided by emerging safety data and decision by the Safety Review Committee (SRC).
No Intervention: CST6 Control
Standard of care
Experimental: CST-2 EIDD-2801 Phase Ib
EIDD-2801 (also known as MK-4482, molnupiravir). Phase Ib: EIDD-2801 will be administered orally, twice daily (BID) for 10 doses (5 or 6 days). The starting dose will be established based on safety and pharmacokinetics from the EIDD-2801-1001-US/UK study, and dose escalations may occur as described in this CST.

CST-2 Phase Ib: EIDD-2801 will be administered orally, twice daily (BID) for 10 doses (5 or 6 days). The starting dose will be established based on safety and pharmacokinetics from the EIDD-2801-1001-US/UK study, and dose escalations may occur as described in this CST.

Phase II: As per Phase Ib, with the dose determined by the recommended phase II dose.

Other Names:
  • MK-4482
  • Molnupiravir
Experimental: CST-5 VIR-7832 Phase I
Phase I: Single doses of VIR-7832 will be administered by intravenous (IV) infusion. The starting dose will be 50 mg, and dose escalations of 150 and 500 mg are anticipated.

CST-5: Phase I, Single doses of VIR-7832 will be administered by intravenous (IV) infusion over 1 hour. The starting dose will be 50 mg, and dose escalations of 150 and 500 mg are anticipated, with escalation guided by emerging safety data and decision by the SRC.

Phase II: As per Phase I, with the dose determined by the recommended phase II dose.

Active Comparator: CST-5 VIR-7831 Phase II
Phase II: 500 mg dose of VIR-7831 will be given by IV infusion.
CST-5 Phase II: A 500 mg dose of VIR-7831 will also be given by IV infusion over 1 hour.
Other Names:
  • Sotrovimab
Placebo Comparator: CST-5 Placebo Phase I
Phase I: placebo blinded controlled
CST-5 Phase 1, Phase II: Placebo given by intravenous infusion over 1 hour
Other Names:
  • Placebo
Experimental: CST3B Nitazoxanide
Phase II experimental arm.

CST3A & CST3B Phase I: Nitazoxanide will be administered orally, initially twice daily (BID) for 14 doses (7 days). The starting dose will be 1500mg BID based on existing dose information, but dose adaptations may occur.

Phase II: As per Phase Ib, with the dose determined by the recommended phase II dose.

Experimental: CST-2 EIDD-2801 Phase II

EIDD-2801 (also known as MK-4482, molnupiravir).

Phase II: As per Phase Ib, with the dose determined by the recommended phase II dose.

CST-2 Phase Ib: EIDD-2801 will be administered orally, twice daily (BID) for 10 doses (5 or 6 days). The starting dose will be established based on safety and pharmacokinetics from the EIDD-2801-1001-US/UK study, and dose escalations may occur as described in this CST.

Phase II: As per Phase Ib, with the dose determined by the recommended phase II dose.

Other Names:
  • MK-4482
  • Molnupiravir
Experimental: CST-8 Phase I Molnupiravir + Paxlovid®
Molnupiravir 800mg Twice a day (BD) in combination with Paxlovid® (300mg nirmatrelvir + ritonavir 100mg) twice a day (BD) for 5 days as starting dose, with a de-escalation protocol reducing in increments of molnupiravir to 600mg BD, then 400mg BD if required. The dose of Paxlovid® will be fixed for all cohorts.
Molnupiravir 800mg Twice a day (BD) for 5 days as starting dose, with a de-escalation protocol reducing in increments of molnupiravir to 600mg BD, then 400mg BD if required.
Other Names:
  • Lagevrio
Paxlovid® (300mg nirmatrelvir + ritonavir 100mg) twice a day (BD) for 5 days. The dose of Paxlovid® will be fixed for all cohorts.
Other Names:
  • nirmatrelvir and ritonavir
No Intervention: CST-8 Phase I Molnupiravir + Paxlovid® Control
Standard of care
Active Comparator: CST-5 VIR-7832
Phase II: 500 mg dose of VIR-7832 will be given by IV infusion.

CST-5: Phase I, Single doses of VIR-7832 will be administered by intravenous (IV) infusion over 1 hour. The starting dose will be 50 mg, and dose escalations of 150 and 500 mg are anticipated, with escalation guided by emerging safety data and decision by the SRC.

Phase II: As per Phase I, with the dose determined by the recommended phase II dose.

Placebo Comparator: CST-5 Placebo Phase II
Phase II: placebo blinded controlled
CST-5 Phase 1, Phase II: Placebo given by intravenous infusion over 1 hour
Other Names:
  • Placebo

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Master Protocol: Dose-finding/Phase I
Time Frame: 29 days from randomisation
Determination of a dose(s) for efficacy evaluation. Dose limiting toxicities (Safety and Tolerability of drug under study - CTCAE v5 Grade ≥3 adverse events)
29 days from randomisation
Master Protocol: Efficacy evaluation/Phase II - Severe patients (Group A)
Time Frame: 29 days from randomisation

Determination of activity and safety.

In severe patients (Group A): time to clinical improvement. Improvement will be determined according to the WHO Clinical Progression Scale; improvement is defined as a minimum 2-step change from randomisation in the scale up to day 29 post-randomisation.

29 days from randomisation
Master Protocol: Efficacy evaluation/Phase II - Mild to moderate patients (Group B)
Time Frame: 15 days from randomisation

Determination of activity and safety.

In mild to moderate patients (Group B): pharmacodynamics of drug under study, defined as time to negative viral titres in nose and/or throat swab, measured up to 15 days post-randomisation.

15 days from randomisation
CST-2 Phase I: To determine the safety and tolerability of multiple ascending doses of EIDD-2801 to recommend dose for phase II.
Time Frame: 7 days from randomisation

Dose limiting toxicity (DLT) using CTCAE version 5 (grades 3 and above) over 7 days.

CTCAE grading related to platelets and/or lymphocytes

7 days from randomisation
CST-2 Phase II: To determine the ability of EIDD-2801 to reduce serious complications of COVID-19 including hospitalization, reduction in SAO2<92%, or death.
Time Frame: 29 days from randomisation
Progression of disease (SpO2<92% based on at least 2 consecutive recordings on the same day) or hospitalization or death up to day 29
29 days from randomisation
CST6 Phase I: To determine the safety and tolerability of multiple doses of IV Favipiravir in patients with COVID-19
Time Frame: 29 days from randomisation
Adverse events and serious adverse events
29 days from randomisation
CST6 Phase I: To determine the maximum safe dose of IV Favipiravir for efficacy evaluation in phase II
Time Frame: 8 days from randomisation
Dose limiting toxicities (Safety and Tolerability of IV Favipiravir- CTCAE v5 Grade ≥3 adverse events)
8 days from randomisation
CST-8 Phase I: Dose Limiting Toxicities up to and including Day 11
Time Frame: 11 days from randomisation
Dose limiting toxicities (Safety and Tolerability of molnupiravir and Paxlovid® combination - CTCAE v5 Grade ≥3 adverse events) up to and including Day 11
11 days from randomisation

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Master Protocol: Safety assessed by rate of adverse events
Time Frame: Up to 29 days from randomisation
Adverse event rate according to CTCAE v5
Up to 29 days from randomisation
Master Protocol: To evaluate clinical improvement
Time Frame: From randomisation to day 29
Proportion of patients with clinical improvement (as defined above) at day 8, 15 and day 29.
From randomisation to day 29
Master Protocol: To evaluate clinical improvement using WHO clinical progression scale
Time Frame: From randomisation to day 15
Change at day 8 and 15 from randomisation in the WHO Clinical Progression Scale
From randomisation to day 15
Master Protocol: To evaluate clinical improvement using WHO clinical progression scale
Time Frame: From randomisation to day 29
Time to a one point change on the WHO Clinical Progression Scale
From randomisation to day 29
Master Protocol: To evaluate clinical improvement using SpO2/FiO2
Time Frame: From randomisation to day 29
The ratio of the oxygen saturation to fractional inspired oxygen concentration (SpO2/FiO2)
From randomisation to day 29
Master Protocol: To evaluate discharge
Time Frame: From randomisation to day 29
Proportion of patient discharged at days 8, 15 and 29
From randomisation to day 29
Master Protocol: To evaluate admission to ICU
Time Frame: From randomisation to day 29
Admission rate to ICU
From randomisation to day 29
Master Protocol: To evaluate safety further (WCC)
Time Frame: From randomisation to day 29
White cell count on day 1, 3, 5, 8, 11 (while hospitalised); and Day 15 and 29
From randomisation to day 29
Master Protocol: To evaluate safety further (Hg)
Time Frame: From randomisation to day 29
Haemoglobin on day 1, 3, 5, 8, 11 (while hospitalised); and Day 15 and 29
From randomisation to day 29
Master Protocol: To evaluate safety further (platelets)
Time Frame: From randomisation to day 29
Platelets on day 1, 3, 5, 8, 11 (while hospitalised); and Day 15 and 29
From randomisation to day 29
Master Protocol: To evaluate safety further (creatinine)
Time Frame: From randomisation to day 29
Creatinine on day 1, 3, 5, 8, 11 (while hospitalised); and Day 15 and 29
From randomisation to day 29
Master Protocol: To evaluate safety further (ALT)
Time Frame: From randomisation to day 29
ALT on day 1, 3, 5, 8, 11 (while hospitalised); and Day 15 and 29
From randomisation to day 29
Master Protocol: To evaluate overall mortality
Time Frame: From randomisation to day 29
Mortality at Days 8, 15 and 29. Time to death from randomisation
From randomisation to day 29
Master Protocol: To evaluate the number of oxygen-free days
Time Frame: From randomisation to day 29
Duration (days) of oxygen use and oxygen-free days
From randomisation to day 29
Master Protocol: To evaluate ventilator-free days
Time Frame: From randomisation to day 29
Duration (days) of mechanical ventilation and mechanical ventilation-free days
From randomisation to day 29
Master Protocol: To evaluate incidence of new mechanical ventilation use
Time Frame: From randomisation to day 29
Incidence of new mechanical ventilation use
From randomisation to day 29
Master Protocol: To evaluate National Early Warning Score (NEWS)2/qSOFA
Time Frame: From randomisation to day 29
NEWS2/qSOFA assessed daily while hospitalised
From randomisation to day 29
Master Protocol: To evaluate translational outcomes (Viral Load)
Time Frame: From randomisation to day 29
Change in viral load over time
From randomisation to day 29
Master Protocol: To evaluate translational outcomes (Baseline SARS-COV-2)
Time Frame: From randomisation to day 29
Change in viral load over time
From randomisation to day 29
CST-2 Additional: Pharmacokinetic Objective: To define PK of EIDD-2801 and EIDD-1931 in plasma following multiple doses administered to patients with COVID-19.
Time Frame: Samples collected on Day 1 and Day 5 post-randomisation
Concentrations of EIDD-2801 and -1931 in plasma
Samples collected on Day 1 and Day 5 post-randomisation
CST-2 Additional: Virologic Objective: To assess the difference in viral clearance (time to negative PCR) between EIDD-2801 and control.
Time Frame: Swabs taken on Day 1 (day of randomisation), 3, 5, 8, 11, 15, 22 and 29
Qualitative (and quantitative when possible) PCR for SARS-CoV-2 by nasal swab.
Swabs taken on Day 1 (day of randomisation), 3, 5, 8, 11, 15, 22 and 29
CST-2 Additional: Clinical Objective: To determine the ability of EIDD-2801 to reduce the duration of signs and symptoms of COVID-19 in patients (FLU-PRO)
Time Frame: From randomisation to Day 29
Patient Reported Outcome Measures (FLU-PRO).
From randomisation to Day 29
CST-2 Additional: Clinical Objective: To determine the ability of EIDD-2801 to reduce the duration of signs and symptoms of COVID-19 in patients (WHO Scale).
Time Frame: From randomisation to Day 29
WHO Progression Scale at day 15 and 29
From randomisation to Day 29
CST-2 Additional: Clinical Objective: To determine the ability of EIDD-2801 to reduce the duration of signs and symptoms of COVID-19 in patients (NEWS2)
Time Frame: From randomisation to Day 29
NEWS2 (National Early Warning Score2) assessed during study clinic visit on days 15 and 29.
From randomisation to Day 29
CST-2 Additional: Clinical Objective: To determine the ability of EIDD-2801 to reduce the duration of signs and symptoms of COVID-19 in patients (mortality)
Time Frame: From randomisation to Day 29
Mortality at Days 15 and 29
From randomisation to Day 29
CST-2 Additional: Clinical Objective: To determine the ability of EIDD-2801 to reduce the duration of signs and symptoms of COVID-19 in patients (death)
Time Frame: From randomisation to Day 29
Time from randomisation to death
From randomisation to Day 29
CST-6 Additional: To characterise the pharmacokinetics (PK) of multiple doses of IV Favipiravir
Time Frame: From randomisation to Day 8
Plasma PK parameters of IV Favipiravir
From randomisation to Day 8
CST-6 Additional: To investigate the ability of IV Favipiravir to reduce the duration of signs and symptoms of COVID-19 in-patients
Time Frame: Randomisation to Day 15 and Day 29
WHO Progression Scale (WHO, 2020)
Randomisation to Day 15 and Day 29
CST-6 Additional: To investigate the effect of IV Favipiravir on SARS-CoV-2 viral load
Time Frame: From randomisation to Day 29
Viral load change from baseline over time
From randomisation to Day 29
CST-8: Assess feasibility for late phase study by reviewing any recorded AEs and SAEs
Time Frame: From randomisation to Day 29
Review of any adverse events
From randomisation to Day 29
CST-8: Assess feasibility for late phase study by reviewing hospitalisation or death up to Day 29
Time Frame: From randomisation to Day 29
Death and hospitalisation up to Day 29
From randomisation to Day 29
CST-8: Measure concentrations of IMP re evidence of virological efficacy
Time Frame: From randomisation to Day 11
PK concentrations of both IMPs and their circulating metabolites in plasma.
From randomisation to Day 11
CST-8: Measure PK of each drug within the combination
Time Frame: From randomisation to Day 11
PK concentrations of both IMPs and their circulating metabolites in plasma.
From randomisation to Day 11
CST8: Review evidence of virological efficacy via viral elimination slopes
Time Frame: From baseline to Day 11
Qualitative (and quantitative when possible) PCR for SARS-CoV-2 by nose and throat swab
From baseline to Day 11
CST8: Vital signs (Heart Rate) at Screening, Baseline/Day 1, Day 3, Day 5 and Day 11
Time Frame: From randomisation to Day 11
Vital sign measure 1 heart rate (beats/min) - to be part of aggregated review of pt vitals to assess safety and tolerability.
From randomisation to Day 11
CST-8: Vital signs (Blood Pressure) at Screening, Baseline/Day 1, Day 3, Day 5 and Day 11
Time Frame: From randomisation to Day 11
Vital sign measure 2 Blood Pressure (mmHG) - to be part of aggregated review of pt vitals to assess safety and tolerability.
From randomisation to Day 11
CST-8: Vital signs (Respiratory Rate) at Screening, Baseline/Day 1, Day 3, Day 5 and Day 11
Time Frame: From randomisation to Day 11
Vital sign measure 3 respiratory rate (breaths/min) - to be part of aggregated review of pt vitals to assess safety and tolerability.
From randomisation to Day 11
CST-8: Vital signs (Temperature) at Screening, Baseline/Day 1, Day 3, Day 5 and Day 11
Time Frame: From randomisation to Day 11
Vital sign measure 4 temperature (degrees c) - to be part of aggregated review of pt vitals to assess safety and tolerability.
From randomisation to Day 11
CST-8: Vital signs (Oxygen Saturation) at Screening, Baseline/Day 1, Day 3, Day 5 and Day 11
Time Frame: From randomisation to Day 11
Vital sign measure 5 oxygen saturation (FiO2 as %) - to be part of aggregated review of pt vitals to assess safety and tolerability.
From randomisation to Day 11

Other Outcome Measures

Outcome Measure
Measure Description
Time Frame
CST-8: Measure PK of nirmatrelvir and ritonavir in tears, saliva and nasal secretions
Time Frame: From randomisation to Day 5
Concentration of nirmatrelvir and ritonavir in non-plasma
From randomisation to Day 5
CST-8:To characterise genetic variability in SARS-CoV-2 before and during treatment via PCR analysis
Time Frame: From randomisation to Day 11
PCR analysis re Baseline and treatment emergent genetic mutations in SARS-CoV-2
From randomisation to Day 11

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

General Publications

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Actual)

July 3, 2020

Primary Completion (Estimated)

December 18, 2023

Study Completion (Estimated)

April 30, 2024

Study Registration Dates

First Submitted

October 13, 2020

First Submitted That Met QC Criteria

February 8, 2021

First Posted (Actual)

February 9, 2021

Study Record Updates

Last Update Posted (Actual)

November 28, 2023

Last Update Submitted That Met QC Criteria

November 21, 2023

Last Verified

November 1, 2023

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

YES

IPD Plan Description

Master protocol publication. Data sets will be registered on CSDR

IPD Sharing Time Frame

Master protocol published 19 June 2020

IPD Sharing Access Criteria

Master protocol available from https://trialsjournal.biomedcentral.com/articles/10.1186/s13063-020-04473-1

IPD Sharing Supporting Information Type

  • STUDY_PROTOCOL

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

No

Studies a U.S. FDA-regulated device product

No

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Covid19

Clinical Trials on CST-2: EIDD-2801

3
Subscribe