HIGHLIGHTS OF 40th ICAAC:

< back to main page <   

Experimental Treatments for HIV

Protease Inhibitor Drugs BMS-232632, DMP-450, Tipranavir

The interim results of a phase II study of BMS-232632 were presented by I. Sanne, MD of Johannesburg Hospital in South Africa.  This drug is an experimental protease (PI) inhibitor that has a once daily dosing, without ìboostingî from a second PI drug.  A total of 422 HIV positive patients were enrolled; 32% were women, 40% were non-Caucasian and 13% had a history of injection drug use.  Less than four weeks of previous NRTI drug experience and less than one week of previous NNRTI or PI drug experience was allowed.  The median baseline CD4 count was 318 cells per microliter, with an HIV RNA viral load of 4.7 log (50,118) copies per milliliter.  In this double-blind study (medication type and dose not know by patient or physician), patients took BMS-232632 as the only drug for two weeks.  They were randomized to a dose of 200, 400 or 500 mg once daily.  A control arm took nelfinavir (Viracept, PI drug) 750 mg 3-times daily.  After two weeks, stavudine (d4T, Zerit) and didanosine (Videx, ddI, both NRTI drugs) were added.  A sub-group of 98 patients have completed 24 of the planned 46 weeks, while the remaining have completed only 16 weeks. 

The interim results in the first sub-group after 24 weeks showed that up to 35% had an undetectable viral load, with a lower limit of 50 copies per milliliter (strict ìintent-to-treatî analysis, all patients included).  In the larger subgroup, the three doses and their respective percentages with an undetectable viral load after 12 weeks were: 200 mg (39%), 400 mg (31%) and 500 mg (50%) (Editorial comment: the larger subgroup was started after the first sub-group, and was a larger study (n=300 or more; about 80 per arm), and patients were recommended to take BMS-232632 with food which enhances drug exposure and reduces drug level variability significantly).  In general, the viral load decreased by approximately 2.5 log (316-fold) at 12-16 weeks.  In the larger subgroup, the CD4 count increased by approximately 75-100 cells per microliter after 12 weeks.  The most common side effects were nausea, diarrhea, stomach area (gastrointestinal) pain, infection and headache.  As has been previously reported, the blood bilirubin (bile pigment) increased in all three dosing armsóthis was treated by reducing the dose of BMS-232632.  In seven of these cases, the patients had jaundice (yellow appearance to skin and whites of eyes).  There were five cases of life-threatening (grade 4) increases.  On the other hand, there were only minimal increases or no changes in total blood cholesterol, ìLDLî (ìbadî or low density lipoprotein) cholesterol or triglycerides (blood fats).  Whereas, the nelfinavir control arm did have increases in those measurements of cholesterol and triglycerides.

In a second presentation about BMS-232632, Edward OíMara MD of Bristol-Myers Squibb showed that there were no significant changes in blood levels of BMS-232632 when it is taken at a 400 mg dose with 200 mg of ketoconazole (Nizoral, anti-fungal antibiotic).  In a third presentation about BMS-232632, Dr. OíMara reported initial results suggesting a genetic marker increases the risk of increased bilirubin (see side effects above) from the drug.  The marker is called ìUDP-GT 1A1î (see reference list).  This marker also should be tested for the same side effect that occurs among some patients when they take the PI drug indinavir (Crixivan).  In a fourth presentation about BMS-232632, Richard Colonno, PhD of Bristol-Myers showed that the drug has ìno obvious mutational patternÖother than a requirement of several [PI drug] mutations to be present.î

See NATAP ICAAC Report on  BMS-232632: once a day protease inhibitor in early human development

References
Colonno RJ and others.  Efficacy of BMS-232632 against a panel of HIV-1 clinical isolates resistance to currently used protease inhibitors. Abstract 2114.

OíMara E and others.  Relationship between uridine diphosphate-glucuronosyl transferase (UDP-GT) 1A1 genotype and total bilirubin elevations in healthy subjects receiving BMS-232632 and saquinavir.  Abstract and poster 1645.

OíMara E and others.  Steady-state pharmacokinetic interaction study between BMS-232632 and ketoconazole in healthy subjects.  Abstract and poster 1646.

Sanne I and others.  Safety and antiviral efficacy of a once-daily HIV-1 protease inhibitor BMS-232632: 24 weeks results from a phase II clinical study.  Abstract 691.

There were three presentations about DMP-450, an experimental PI drug under development by Triangle Pharmaceuticals.  A phase I/II study included 40 HIV positive patients.  Women represented 13%; race-ethnic background was not reported.  None had previously taken anti-HIV drugs.  The median baseline viral load was 4.6-5.3 (39,810-177,827) log copies per milliliter with a median CD4 count of 234-285 cells per microliter.  The dose of DMP-450 was 750 mg 3-times daily, 1250 mg 3-times daily or 1,250 mg twice daily.  The two NRTI drugs also taken by all patients were stavudine (d4T, Zerit) and lamivudine (3TC, Epivir). 

The results showed that after up to 48 weeks, 87% had an undetectable viral load using a lower limit of 50 copies per milliliter (strict ìintent-to-treatî analysis, all patients included).  Side effects included diarrhea, nausea, muscle aches, headache and rash.  There were mild increases in blood cholesterol.  The authors concluded that DMP-450 was tolerated well, had significant anti-HIV benefits with few differences when comparing the three doses.  Interestingly, the 1,250 mg 3-times daily dosing led to equal or marginally better drug concentrations than the 1,250 mg twice daily dosing.  This included the maximal, minimal and total (ìarea-under-the-curveî) concentrations.  This suggests that DMP-450 might be dosed twice daily, which is associated with better adherence to dosing in other studies.  Dr. G. Moralles of Triangle Pharmaceuticals has reported previously that this PI drug has a relatively lower cost of production that could lead to marketing in resource-poor countries.

In a second presentation about DMP-450, Dr. Laurene Wang of Triangle Pharmaceuticals reported drug interactions when it was added to either indinavir (Crixivan) or saquinavir (Fortovase, both PI drugs).  Healthy HIV negative volunteers were enrolled.  Neither of those two PI drugs affected the blood concentrations of DMP-450.  However, 1000 mg of DMP increased the minimal and total (ìarea-under-the-curveî or ìAUCî) concentration of indinavir by approximately 40%.  Yet, the maximal concentration and ìhalf-lifeî (time until an original amount is metabolized by half) were not changed.  When DMP-450 was added to saquinavir, the maximal concentration increased 5-fold, the ìAUCî concentration 10-fold and the ìhalf-lifeî by 2.5-fold.  This information is important before clinical studies could be devised that would combine DMP with either indinavir or saquinavir.

There was a third presentation about side effects of DMP-450 authored by Dr. Gregory Chitick of Triangle Pharmaceuticals.  The results were a lesser increase in total blood cholesterol when compared to indinavir (Crixivan, PI drug) and similar increases in triglycerides (fats) and glucose (sugar).  Dr. Chitick concluded that 2,000 mg twice daily of DMP-450 ìappears to be the maximum tolerated dose.

References
Chittick GE and others.  Effects of a novel HIV-1 protease inhibitor, DMP 450, on cardiac tracing, serum lipids and glucose tolerance, as compared to indinavir.  Abstract and presentation 1648.

Sierra J and others.  Preliminary profile of the antiviral activity, metabolic effects, safety and pharmacokinetics of DMP-450, a novel cyclic urea protease inhibitor.  Abstract and presentation 540.

Wang LH and others.  Effects of a novel HIV-1 protease inhibitor, DMP 450, on the pharmacokinetics of indinavir and saquinavir.  Abstract and presentation 1647.

Information about resistance to tipranavir was presented by Dr. Sharon Kemp of Virco.  This experimental PI drug has activity against numerous HIV strains (isolates) that are resistant to PI drugs currently available by prescription.  Dr. Kemp concluded, ìThere does not appear to be any obvious common combinations of the known PI mutations that clearly confer tipranavir resistance.î  She was able to generate moderate resistance with eleven primary and secondary mutations seen with other PI drugs.

Reference
Kemp S and others.  Site-directed mutagenesis and in vitro drug selection studies have failed to reveal a consistent genotypic resistance pattern for tipranavir.  Abstract 2113.

A more detailed report was shown by Sharon Kemp at the Resistance Workshop. Here's the link to NATAP Report: Tipranavir: in vitro resistance data on a protease inhibitor for PI resistance

NNRTI Drugs Capravirine, R165335

The results of a phase I study of capravirine (AG1549) were presented by Dr. S. Hayashi of Agouron Pharmaceuticals.  This drug is an experimental NNRTI drug with activity against the common ìK103Nî mutation that causes resistance to each of the three NNRTI drugs currently available.  A total of 45 HIV negative volunteers included 53% women, 22% African-American and 16% Hispanics.  The results showed that, when compared to fasting (empty stomach), blood concentrations of capravirine were increased 2-fold when taken with a meal that was either low-fat, low-calorie or high-fat, high-calorie.  Those concentrations included minimal, maximal and total (ìarea-under-the-curveî).  The ìhalf-lifeî (time until an original amount is metabolized by half) was not changed.  The dose was two 700 mg tablets. 

Reference  
Hayashi S and others.  Effects of fasting, high-fat and low-fat meals on the pharmacokinetics of a single dose of capravirine (AG1549), an NNRTI, in healthy volunteers.  Abstract and presentation 1667.

Laboratory testing of a ìthird generationî NNRTI drug called ìR165335î were presented by researchers from Janssen Research Foundation and Tibotec.  This compound had anti-HIV activity at ìnanomolarî (extremely low) concentrations.  It also retained activity against several HIV strains (isolates) with one or two resistance mutations that renders any of the three FDA-approved NNRTI drugs ineffective.  Also, its activity is not affected by human proteins albumin and ìalpha 1 acid glycoproteinî that can bind several of the marketed anti-HIV drugs.  Further development is proceeding.

References
Andries K and others.  R165335-TMC125, a novel non nucleoside reverse transcriptase inhibitor (NNRTI) with nanomolar activity against NNRTI resistance HIV strains.  Abstract 1840.

De Bethune MP and others.  R165335-TMC125, a third generation non nucleoside reverse transcriptase inhibitor (NNRTI) inhibits 98% of more than 2,000 recombinant HIV clinical isolates at 100nM.  Abstract 1841.

NtRTI Drug Tenofovir

There were three presentations about tenofovir disoproxil (PMPA).  This drug is an experimental nucleotide reverse transcriptase inhibitor drug (NtRTI) under development by Gilead Sciences.  It is dosed once daily and has activity against HIV strains (isolates) with specific resistance to several NRTI drugs (see 2nd next paragraph).  The NtRTI drugs differ from the NRTI drugs in that they only require two steps for activation rather than three that are required for the NRTI drugs.  An update of tenofovir in patients with advanced disease was presented by Robert Schooley, MD of the University of Colorado at Denver.  The study was ìdouble blindî (medication type not known by patient or physician) and ìplacebo-controlledî (one arm takes a ìdummy,î inactive drug or sugar pill).  Enrolled patients must have had a detectable viral load on a stable anti-HIV regimen for at least two months.  Tenofovir was added at a dose of 75 mg, 150 mg, or 300 mg.  All patients, including those randomized to placebo took 300 mg of tenofovir after week 24.  The 189 enrolled patients had taken anti-HIV medication for a mean of 4.6 years with a mean baseline viral load (when starting this study) of 375 cells per microliter.  At that time, 97% had ìgenotypeî resistance mutations to NRTI drugs.

The results after 48 weeks showed the following viral load reductions: 300 mg (decrease of 0.7 log copies per milliliter or 5-fold); 150 mg (decrease of 0.6 log copies per milliliter or 4-fold); 75 mg (decrease of 0.4 log or 2.5-fold); and placebo changed to 300 mg (decrease of 0.7 log or 5-fold).  The CD4 count increases were: 300 mg (11 cells per microliter); 150 mg (16 cells per microliter); 75 mg (11 cells per microliter); and placebo changed to 300 mg (25 cells per microliter).  At 48 weeks, 20% had stopped tenofovir, compared to 25% in the 300 mg arm up to 24 weeks.  Dr. Schooley reported that there was no dose-related toxicity.  The percentage of patients with an undetectable viral load was not presented.

In a separate presentation, Michael Miller, PhD of Gilead presented information about tenofovir resistance.  HIV strains (isolates) with the common lamivudine (3TC)-induced mutation ìM184Vî are ìhypersensitiveî (less drug needed to achieve the same effect as with ìwild-typeî virus without mutations) to tenofovir.  Almost all strains with high-level resistance to zidovudine (AZT, Retrovir) also were sensitive to tenofovir.  Adding ìM184Vî to those strains increased tenofovir sensitivity somewhat.  A combination of NRTI mutations (ìQ151Mî) that lead to multi-drug NRTI resistance did not decrease tenofovirís effectiveness, with or without ìM184V.î  However, another group of multi-drug resistance mutations (ì69 insertionsî) were resistant to tenofovir, but became more sensitive (ìintermediate resistance, 6-fold decreaseî) when ìM184Vî was added.  The ìK65Rî resistance mutation (due to abacavir, didanosine or zalcitabine) decreased sensitivity to tenofovir somewhat, but this was nearly reversed when ìM184Vî was added.  These results indicate the unique effectiveness of tenofovir against HIV with several types and groupings of mutations that cause NRTI drug resistance.

In a third presentation, Dr. L.K. Naeger of Gilead presented other information that might explain part of the reason for tenofovirís effectiveness against HIV strains with various NRTI mutations.  Resistance to zidovudine and stavudine (d4T, Zerit) has been partly explained by two different mechanisms.  They are called ìpyrophosphorolysisî and ìdinucleotide polyphosphate synthesis.î  In a series of experiments, Dr. Naeger showed that tenofovir is much less susceptible to these resistance mechanisms than zidovudine is.

References
Miller MD and others.  Antiviral activity of tenofovir (PMPA) against nucleoside-resistant HIV samples.  Abstract and presentation 2115.

Naeger LK and others.  Tenofovir (PMPA) is less efficiently removed through pyrophosphorolysis and dinucleotide polyphosphate synthesis than zidovudine by HIV-1 wild-type RT and RT mutations.  Abstract and presentation 1265.

Schooley R and others.  Tenofovir disoproxil fumarate (TDF) for the treatment of antiretroviral experienced patients, a double blind, placebo-controlled study.  Abstract and presentation 692.

NRTI Drugs: emtricitabine (Coviracil), DAPD

Emtricitabine (FTC, Coviracil) is a NRTI drug with activity against HIV and HBV (hepatitis B virus).  Safety results were presented by Dr. T.B. Grizzle of Triangle Pharmaceuticals.  In various tests in the laboratory, including small animals, essentially no toxicity (including cancers and birth defects) was detected.  At a dose more than 100-times that used in humans, anemia (low red cell count) occurred in female monkeys and resolved after the drug was stopped.  At a dose almost 1,000-times that used in humans, the same side effect occurred in mice.  In several laboratory tests for ìmitochondrialî toxicity, none was found.  (Mitochondria are the energy producers of cells and several NRTI drugs are toxic to them.)  In a small study of 40 patients that combined FTC with didanosine (ddI, Videx) and efavirenz (Sustiva) in a once-daily regimen, 95% achieved an undetectable HIV viral load (lower limit 400 copies per milliliter) after 48 weeks (see reference).  None had had prior treatment for HIV.  This strongly suggests very high potency of that triple drug combination.  A second report at ICAAC presented DAPD results in children when testing safety and drug metabolism.

References
Grizzle TB and others.  Emtricitabine: summary of toxicology and nonclinical pharmacology evaluations.  Abstract and poster 1631.

Molina J-M and others. Once-daily therapy with emtricitabine, didanosine and efavirenz in treatment naÔve HIV-infected adults: 48-week follow-up of The ANRS 091 Trial. Late Breaker abstract 648 at the 38th Annual Meeting of the IDSA (Infectious Diseases Society of America); September 7-10, 2000; New Orleans, Louisiana.

Wiznia AA and others.  An evaluation of the pharmacokinetics and safety of single oral doses of emtricitabine (Coviracil) in HIV-infected or exposed children.  Abstract and poster 1665.

The results of a phase I/II study of DAPD were presented by Joseph Eron, MD of the University of North Carolina at Chapel Hill.  DAPD is a NRTI drug and has activity against HIV and HBV (hepatitis B virus).  It also has activity against HIV strains with selected resistance to zidovudine (AZT, Retrovir), lamivudine (3TC, Epivir) and/or abacavir (Ziagen).  HIV positive patients without previous treatment for HIV were enrolled.  The DAPD dose was twice daily of 25 mg, 100 mg, 200 mg, 300 mg, or 500 mg.  The maximum HIV RNA viral load reduction after 15 days of the drug was 1.5 log (31-fold) copies per milliliter at the 300 mg dose. The drug was tolerated well, without any discontinuations due to drug adverse effects (side effects).  No ìgenotypeî resistance associated with NRTI drugs was detected at the end of the 15-day study. In a treatment experienced group, a 1.1 log reduction in viral load has been seen in small 15 day study using the 500 mg dose.

Other Drugs in Pre-Human Testing

There were a few other drugs or ìcompoundsî presented at ICAAC that are in ìpreclinicalî (before use in humans) development.  The first group of integrase inhibitors called ìbutanoic acidsî was described by Dr. John S. Wai from Merck.  PRO 140, an entry inhibitor drug, is an antibody (type of protein) to the ìCCR5 receptorî on human immune cells to which HIV binds.  In lab tests, ìlow nanomolarî (extremely low) concentrations of PRO 140 blocked infection of blood ìmononuclear cellsî by HIV strains that use ìCCR5î to enter.  Dr. W. C. Olson of Progenics presented the study.  In a separate presentation, Dr. Olson found that combining the two HIV entry inhibitor drugs PRO 542 and T-20 led to ìsynergisticî (enhanced effects by combining) benefits in the laboratory.  (T-20 is already in phase III human studies and is also called an HIV ìfusionî inhibitor.)  In yet another presentation, Dr. C. L. Tremblay of Massachusetts General Hospital in Boston reported anti-HIV ìsynergyî when T-20 was combined with the ìCCR5î receptor inhibitor TAK-779.  Previously, Dr. Tremblay has reported a similar synergistic effect in the laboratory when T-20 was combined with AMD-3100, an entry inhibitor of the CXCR4 receptor on human T cells that HIV uses to enter.  AMD 3100 was reported to be absorbed into the blood of rabbits when they took the experimental medication by mouth.  Dr. R. T. MacFarland of AnorMED Inc. presented the report at ICAAC.  Two experimental ìribonucleotide reductaseî inhibitors showed anti-AIDS benefits in an animal (ìmurineî) AIDS model, either alone or when combined with didanosine (ddI, Videx).  Both Trimidox and Didox are similar to hydroxyurea (Hydrea), but showed much less toxicity than hydroxyurea.  The report was presented by Dr. C. N. Mayhew of the University of Wolverhampton in the UK.

References
MacFarland RT and others.  An orally bioavailable CXCR4 antagonist for inhibition of HIV replication.  Abstract 1845.

Mayhew CN and others.  Comparison of novel ribonucleotide reductase inhibitors, Didox and Trimidox, to hydroxyurea regarding antiretroviral activity and toxicity in murine AIDS.  Abstract 553.

Olson WC and others.  Potent, broad spectrum inhibition of HIV-1 by the CCR5 antibody PRO 140.  Abstract 550.

Olson WC and others.  Potent, synergistic inhibition of HIV-1 by combinations of the viral entry inhibitors PRO 542 and T-20.  Abstract 549.

Tremblay CL and others. In vitro synergy observed between the fusion inhibitor T-20 and a CCR5 inhibitor TAK-779.  Abstract 1164.

Wai JS and others.  4-aryl-2,4-dioxobutanoic acid inhibitors of HIV-1 integrase and viral replication in cells.  Abstract and presentation 1844.

 < back to main page <