search
Back to results

Safety, Tolerance, Pharmacokinetic and Antiviral Study of Amdoxovir in Combination With Zidovudine in Adults With HIV

Primary Purpose

HIV Infections

Status
Completed
Phase
Phase 1
Locations
Argentina
Study Type
Interventional
Intervention
zidovudine
amdoxovir
pharmacokinetic sampling
Sponsored by
RFS Pharma, LLC
About
Eligibility
Locations
Outcomes
Full info

About this trial

This is an interventional treatment trial for HIV Infections focused on measuring HIV, pharmacokinetics, zidovudine, amdoxovir, DAPD, AMDX, Treatment Naive

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

  • HIV-infection
  • Antiretroviral therapy naïve, or if experienced, no treatment within 90 days of study screening and plan to remain off therapy for the duration of the screening and study treatment period
  • HIV-1 RNA ≥ 5,000 copies/mL within 30 days of study Day 1
  • CD4+ count ≥ 200 cells/mm³ within 30 days of study Day 1
  • Agree to the use of two forms of adequate contraception for women, one form for men
  • Estimated creatinine clearance > 80 mL/min
  • Serum creatinine < 1.5 g/dL
  • Able to give written informed consent prior to study start and adhere to study requirements

Exclusion Criteria:

  • Active alcohol or drug use which in the opinion of the Investigator will likely compromise adherence to the study requirements
  • A positive urine test for amphetamines, cocaine, and/or opioids
  • Currently has any active AIDS defining illness (Category C condition according to the CDC Classification System for HIV Infection 1993)
  • Any active clinically significant disease or findings during screening of medical history or physical examination that in the Investigator's opinion would compromise the outcome of the study
  • Receiving oral concomitant antiviral or prophylactic drugs for opportunistic infections within 30 days prior to study entry
  • Receiving concomitant treatment with nephrotoxic drugs (e.g., aminoglycosides [tobramycin, gentamicin, and amikacin], amphotericin B, vancomycin, cidofovir, foscarnet, cis-platinum, pentamidine), or competitors of renal excretion (e.g., probenecid) within 30 days of study entry
  • Visual abnormalities (e.g. cataracts, macular degeneration) other than non-organic decreased visual acuity
  • Receiving concomitant treatment with immunosuppressive drugs within 30 days prior to study entry
  • Diabetes mellitus Type 1 or 2 which is being treated or not with any anti-diabetes agents
  • Current significant gastrointestinal, renal, hepatic, bronchopulmonary, biliary, neurological, cardiovascular, oncologic, allergic, or ophthalmologic diseases including history of cataracts/lens opacities
  • The following laboratory values performed within 30 days prior to study Day 1:

    • Hemoglobin < 9.0 g/dL for men; 8.0 g/dL for women
    • Platelet count < 75,000 cells per mL
    • Absolute neutrophil count < 1,000 cells per mL
    • AST, ALT, alkaline phosphatase, and amylase > 3 x ULN
    • Random or fasting glucose > 121 mg/dL
    • Serum lipase > 1.5 x ULN
    • Urinalysis ≥ 2+ proteinuria and ≥ 2 cellular casts per high powered field (HPF)
  • Active acute hepatitis A and/or chronic hepatitis B or C with detectable viremia
  • Subjects with clinical or laboratory evidence of significantly decreased hepatic function or decompensation, irrespective of liver enzyme levels (INR > 1.3 or albumin < 30 g/l or bilirubin > 2.5 x ULN)
  • Pregnant women
  • Breastfeeding women

Sites / Locations

  • Hospital Privado Modelo

Outcomes

Primary Outcome Measures

Proportion of subjects in each arm with Grade 3 or greater treatment emergent adverse events (AE)
Plasma Cmax, Cmin, tmax, AUC0→τ, AUC0→∞, t1/2, CL/F of DAPD and AZT in each group at Days 1 and 10
Intracellular Cmax, Cmin, tmax, AUC0→τ, AUC0→∞, and t1/2 of DXG-TP, -MP and -DP and AZT-TP, -MP, and -DP at Days 1 and 10

Secondary Outcome Measures

Change in viral load (plasma HIV-1 RNA) from Baseline through Day 10
Correlate intracellular DXG-TP and AZT-TP with viral response, as measured by plasma HIV-1 RNA
Quantitate DXG, AZT, and GAZT in urine with and without DAPD
Characterize viral rebound (plasma HIV-1 RNA) following drug discontinuation for 48 hr
Measure CD4+ count changes from Baseline to Day 10

Full Information

First Posted
February 5, 2007
Last Updated
May 10, 2007
Sponsor
RFS Pharma, LLC
search

1. Study Identification

Unique Protocol Identification Number
NCT00432016
Brief Title
Safety, Tolerance, Pharmacokinetic and Antiviral Study of Amdoxovir in Combination With Zidovudine in Adults With HIV
Official Title
A Randomized, Placebo-Controlled Study to Evaluate Safety, Tolerance, Pharmacokinetics and Antiviral Activity of Amdoxovir and Zidovudine in Untreated HIV-1 Infected Subjects Currently Untreated
Study Type
Interventional

2. Study Status

Record Verification Date
May 2007
Overall Recruitment Status
Completed
Study Start Date
February 2007 (undefined)
Primary Completion Date
undefined (undefined)
Study Completion Date
May 2007 (Actual)

3. Sponsor/Collaborators

Name of the Sponsor
RFS Pharma, LLC

4. Oversight

Data Monitoring Committee
No

5. Study Description

Brief Summary
The purpose of this study is to determine the short term safety, tolerance, and antiviral effect of zidovudine (AZT) and amdoxovir (AMDX, DAPD) in combination, and whether the dosage for AZT can be reduced, potentially decreasing side effects, while maintaining antiviral effects. Study hypothesis: DADP in combination with AZT is safe and effective, and AZT dosing may be reduced, resulting in lower levels of AZT-monophosphate associated with toxicity and maintaining levels of AZT-triphosphate associated with efficacy.
Detailed Description
By 2008, market data suggest that the most commonly prescribed initial treatment regimen for HIV-1 will consist of Truvada® (FTC and tenofovir disoproxil fumarate (TDF)) and Sustiva® (efavirenz (EFV)). A newly formulated drug called Atripla™ containing all three active ingredient has now been approved by the US FDA. Therefore, second line treatments that are currently in development should provide activity against resultant mutations, primarily M184V/I (17%) and much less commonly K65R (0 to 5%), and ideally prevent or be effective against mutations that may occur during second line therapy. The goal of our program is to identify an AZT/DAPD co-formulation with reduced resistance development and an improved safety profile for the treatment of HIV infections. DAPD has increased sensitivity to M184V/I strains and is active against thymidine analog mutations (TAMs) that may have occurred during previous antiretroviral regimens. AZT offers anti-K65R activity which is believed to be conferred by the 3'-azido moiety containing pseudo-sugar structure and base components of AZT. Hence, AZT could potentially be incorporated to prevent the emergence of the K65R mutation that could limit the long-term benefit of DAPD. Since nucleoside reverse transcriptase inhibitors (NRTIs) require intracellular phosphorylation to form their active triphosphates, it is preferable to combine NRTIs with different critical kinases. Thymidine kinase-1 is the critical enzyme for the phosphorylation of AZT to its monophosphates. The enzyme involved in the initial phosphorylation of the active metabolite of DAPD, DXG, is guanosine kinase. Nucleotide competition studies conducted using activated human peripheral blood mononuclear (PBM) cells performed with DAPD and AZT with concentrations between 0.1-10 µM demonstrated no competitive inhibition of DXG-triphosphate formation. The approved dose for AZT is 300 mg bid, and AZT is available as a 300 mg tablet or 100 mg capsule. A previous study in 10 HIV seropositive individuals comparing cellular AZT-mono, di- and tri-phosphate nucleotides at normal and reduced doses, 100 mg tid versus 300 mg bid, demonstrated a significant decrease in plasma AZT and intracellular AZT-monophosphate (AZT-MP) levels, (AZT-MP is associated with toxicity), without significant changes in AZT-triphosphate (AZT-TP) levels in activated PBM cells or antiviral activity. These findings on the effect of AZT dose on intracellular AZT-TP are supported by computer simulations. Maximal predicted cellular levels of AZT-MP and AZT-TP at steady-state, following 200 mg bid and 300 mg bid dosing in 3,000 simulated individuals demonstrated a high overlap between the AZT-TP histograms (> 85 %), suggesting similar efficacies for the 200 and 300 mg bid doses. The low degree of overlap between the AZT-MP histograms (< 8 %) for the two dose regimens, suggests that there may be fewer toxicities with the 200 mg bid dose, supporting our hypothesis that a zidovudine dose of 200 mg bid could reduce the AZT-MP levels without compromising the AZT-TP levels that would be obtained with a 300 mg bid dose. This 10 day, proof of principle, pharmacokinetic study will provide important information about reduced AZT dosing to support the development of an AZT/DAPD co-formulation which may prevent the emergence of K65R mutations.

6. Conditions and Keywords

Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
HIV Infections
Keywords
HIV, pharmacokinetics, zidovudine, amdoxovir, DAPD, AMDX, Treatment Naive

7. Study Design

Primary Purpose
Treatment
Study Phase
Phase 1, Phase 2
Interventional Study Model
Single Group Assignment
Masking
Double
Allocation
Randomized
Enrollment
24 (false)

8. Arms, Groups, and Interventions

Intervention Type
Drug
Intervention Name(s)
zidovudine
Intervention Type
Drug
Intervention Name(s)
amdoxovir
Intervention Type
Procedure
Intervention Name(s)
pharmacokinetic sampling
Primary Outcome Measure Information:
Title
Proportion of subjects in each arm with Grade 3 or greater treatment emergent adverse events (AE)
Title
Plasma Cmax, Cmin, tmax, AUC0→τ, AUC0→∞, t1/2, CL/F of DAPD and AZT in each group at Days 1 and 10
Title
Intracellular Cmax, Cmin, tmax, AUC0→τ, AUC0→∞, and t1/2 of DXG-TP, -MP and -DP and AZT-TP, -MP, and -DP at Days 1 and 10
Secondary Outcome Measure Information:
Title
Change in viral load (plasma HIV-1 RNA) from Baseline through Day 10
Title
Correlate intracellular DXG-TP and AZT-TP with viral response, as measured by plasma HIV-1 RNA
Title
Quantitate DXG, AZT, and GAZT in urine with and without DAPD
Title
Characterize viral rebound (plasma HIV-1 RNA) following drug discontinuation for 48 hr
Title
Measure CD4+ count changes from Baseline to Day 10

10. Eligibility

Sex
All
Minimum Age & Unit of Time
18 Years
Accepts Healthy Volunteers
No
Eligibility Criteria
Inclusion Criteria: HIV-infection Antiretroviral therapy naïve, or if experienced, no treatment within 90 days of study screening and plan to remain off therapy for the duration of the screening and study treatment period HIV-1 RNA ≥ 5,000 copies/mL within 30 days of study Day 1 CD4+ count ≥ 200 cells/mm³ within 30 days of study Day 1 Agree to the use of two forms of adequate contraception for women, one form for men Estimated creatinine clearance > 80 mL/min Serum creatinine < 1.5 g/dL Able to give written informed consent prior to study start and adhere to study requirements Exclusion Criteria: Active alcohol or drug use which in the opinion of the Investigator will likely compromise adherence to the study requirements A positive urine test for amphetamines, cocaine, and/or opioids Currently has any active AIDS defining illness (Category C condition according to the CDC Classification System for HIV Infection 1993) Any active clinically significant disease or findings during screening of medical history or physical examination that in the Investigator's opinion would compromise the outcome of the study Receiving oral concomitant antiviral or prophylactic drugs for opportunistic infections within 30 days prior to study entry Receiving concomitant treatment with nephrotoxic drugs (e.g., aminoglycosides [tobramycin, gentamicin, and amikacin], amphotericin B, vancomycin, cidofovir, foscarnet, cis-platinum, pentamidine), or competitors of renal excretion (e.g., probenecid) within 30 days of study entry Visual abnormalities (e.g. cataracts, macular degeneration) other than non-organic decreased visual acuity Receiving concomitant treatment with immunosuppressive drugs within 30 days prior to study entry Diabetes mellitus Type 1 or 2 which is being treated or not with any anti-diabetes agents Current significant gastrointestinal, renal, hepatic, bronchopulmonary, biliary, neurological, cardiovascular, oncologic, allergic, or ophthalmologic diseases including history of cataracts/lens opacities The following laboratory values performed within 30 days prior to study Day 1: Hemoglobin < 9.0 g/dL for men; 8.0 g/dL for women Platelet count < 75,000 cells per mL Absolute neutrophil count < 1,000 cells per mL AST, ALT, alkaline phosphatase, and amylase > 3 x ULN Random or fasting glucose > 121 mg/dL Serum lipase > 1.5 x ULN Urinalysis ≥ 2+ proteinuria and ≥ 2 cellular casts per high powered field (HPF) Active acute hepatitis A and/or chronic hepatitis B or C with detectable viremia Subjects with clinical or laboratory evidence of significantly decreased hepatic function or decompensation, irrespective of liver enzyme levels (INR > 1.3 or albumin < 30 g/l or bilirubin > 2.5 x ULN) Pregnant women Breastfeeding women
Overall Study Officials:
First Name & Middle Initial & Last Name & Degree
Carlos Zala, MD
Organizational Affiliation
Hospital Privado Modelo
Official's Role
Principal Investigator
Facility Information:
Facility Name
Hospital Privado Modelo
City
Buenos Aires
ZIP/Postal Code
B1602DBG
Country
Argentina

12. IPD Sharing Statement

Citations:
PubMed Identifier
8902065
Citation
Barry MG, Khoo SH, Veal GJ, Hoggard PG, Gibbons SE, Wilkins EG, Williams O, Breckenridge AM, Back DJ. The effect of zidovudine dose on the formation of intracellular phosphorylated metabolites. AIDS. 1996 Oct;10(12):1361-7. doi: 10.1097/00002030-199610000-00008.
Results Reference
background
PubMed Identifier
10858331
Citation
Bazmi HZ, Hammond JL, Cavalcanti SC, Chu CK, Schinazi RF, Mellors JW. In vitro selection of mutations in the human immunodeficiency virus type 1 reverse transcriptase that decrease susceptibility to (-)-beta-D-dioxolane-guanosine and suppress resistance to 3'-azido-3'-deoxythymidine. Antimicrob Agents Chemother. 2000 Jul;44(7):1783-8. doi: 10.1128/AAC.44.7.1783-1788.2000.
Results Reference
background
PubMed Identifier
16641288
Citation
Parikh UM, Bacheler L, Koontz D, Mellors JW. The K65R mutation in human immunodeficiency virus type 1 reverse transcriptase exhibits bidirectional phenotypic antagonism with thymidine analog mutations. J Virol. 2006 May;80(10):4971-7. doi: 10.1128/JVI.80.10.4971-4977.2006.
Results Reference
background
PubMed Identifier
16184030
Citation
Thompson MA, Kessler HA, Eron JJ Jr, Jacobson JM, Adda N, Shen G, Zong J, Harris J, Moxham C, Rousseau FS; DAPD-101 Study Group. Short-term safety and pharmacodynamics of amdoxovir in HIV-infected patients. AIDS. 2005 Oct 14;19(15):1607-15. doi: 10.1097/01.aids.0000186822.68606.05.
Results Reference
background
PubMed Identifier
20386073
Citation
Murphy RL, Kivel NM, Zala C, Ochoa C, Tharnish P, Mathew J, Pascual ML, Schinazi RF. Antiviral activity and tolerability of amdoxovir with zidovudine in a randomized double-blind placebo-controlled study in HIV-1-infected individuals. Antivir Ther. 2010;15(2):185-92. doi: 10.3851/IMP1514.
Results Reference
derived

Learn more about this trial

Safety, Tolerance, Pharmacokinetic and Antiviral Study of Amdoxovir in Combination With Zidovudine in Adults With HIV

We'll reach out to this number within 24 hrs