1 / 59

Chicago, Illinois | September 17-20, 2007

Key HIV Research From ICAAC 2007: The 47th Interscience Conference on Antimicrobial Agents and Chemotherapy. Faculty:. Chicago, Illinois | September 17-20, 2007. Cal Cohen, M.D., M.S. Eric Daar, M.D. This activity is supported by an educational grant from:. Faculty for This Activity.

haig
Télécharger la présentation

Chicago, Illinois | September 17-20, 2007

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Key HIV Research From ICAAC 2007: The 47th Interscience Conference on Antimicrobial Agents and Chemotherapy Faculty: Chicago, Illinois | September 17-20, 2007 Cal Cohen, M.D., M.S. Eric Daar, M.D. This activity is supported by an educational grant from:

  2. Faculty for This Activity Dr. Cohen is the research director of the Community Research Initiative of New England and teaches at Harvard Medical School in Boston, Mass. In addition, he works as a HIV clinical management consultant and internist at Harvard Pilgrim Health Care, Boston, Mass., and is affiliated with Harvard Vanguard Medical Associates. Dr. Cohen was co-chair of the Scientific Advisory Committee of amfAR community-based clinical trials network, and served as co-principal investigator of the Harvard/BCH AIDS Clinical Trials Unit, AIDS Clinical Trials Group. He holds appointments at Brigham and Women's Hospital and Beth Israel Hospital, both in Boston, Mass. Cal Cohen, M.D., M.S. Dr. Daar is the chief of HIV medicine at Harbor-UCLA Medical Center in Los Angeles, Calif., and a professor of medicine at the University of California-Los Angeles' David Geffen School of Medicine. He has been an active HIV physician and researcher since the 1980s; during the past three decades, he has led dozens of studies on a vast range of HIV-related issues, with a particular focus on coinfections and other health complications associated with HIV and HIV treatment, including hepatitis C, metabolic complications, cardiovascular disease and psychosocial issues such as depression. Eric Daar, M.D.

  3. ICAAC 2007: Key HIV Research About this slide presentation • This presentation was created to accompany The Body PRO's podcast summary of key research presented at ICAAC 2007, featuring interviews with Cal Cohen, M.D., M.S., and Eric Daar, M.D. For more information about this program, please visit us on the Web at: TheBodyPRO.com/ICAAC2007 • Please feel free to use this slide presentation for personal reference or for your own presentations; however, we ask that you not modify any aspects of the slides contained within this presentation, so proper attribution can be retained. If you would like to publish all or part of this presentation, or repost any of these slides online, permission must first be obtained from Body Health Resources Corporation. • Our gratitude goes out to all who granted permission for their slides to be adapted for this presentation. Disclaimer Knowledge about HIV changes rapidly. Note the date of this presentation's publication, and before treating patients or employing any therapies described in these materials, verify all information independently. If you are a patient, please consult a doctor or other medical professional before acting on any of the information presented in this presentation.

  4. First-Line Antiretroviral Therapy

  5. ARTEMIS: Phase III Study Design DRV/r 800/100mg qd + TDF 300 mg and FTC 200 mg (N=343) 689 ARV-naïve patients VL>5,000; no CD4 entry LPV/r 400/100mg bid or 800/200mg qd + TDF 300 mg and FTC 200 mg (N=346) Dosing was based on regulatory approval; switch was made according to local regulatory approval and drug availability Edwin DeJesus et al. ICAAC 2007; abstract H-718b. Reprinted with permission.

  6. ARTEMIS: Baseline Characteristics DRV/r qd (N=343) LPV/r qd or bid (N=346) 104 (30) 36 (9) 40/23/23 105 (30) 35 (9) 44/21/22 Baseline demographics Female, N (%) Mean (±SD) age (yrs) Caucasian/Black/Hispanic, % Baseline disease characteristics Median HIV-1 RNA (cpm) (range) Median CD4 (cells/mm3 [range]) HBV/HCV co-infected, n (%) 70,800 (835–5,580,000) 228 (4–750) 43 (13) 62,100(667–4,580,000) 218 (2–714) 48 (14) Stratification factors at screening CD4 count <200 cells/mm3 Plasma HIV-1 RNA ≥100,000 cpm 40% 36% 41% 36% Edwin DeJesus et al. ICAAC 2007; abstract H-718b. Reprinted with permission.

  7. ARTEMIS: Viral Load <50 copies/mL to Week 48 (ITT-TLOVR) 100 DRV/r qd (N=343) LPV/r qd or bid(N=346) 90 84% 80 78% 70 60 Patients with VL <50 copies/mL (% [±SE]) 50 40 Estimated difference in response vs LPV/r for non-inferiority: PP = 5.6% (95% CI –0.1;11.3) p<0.001 Estimated difference in response vs LPV/r for superiority: ITT = 5.5% (95% CI –0.3;11.2) p=0.062 Estimated difference in response vs LPV/r for non-inferiority: PP = 5.6% (95% CI –0.1;11.3) p<0.001 30 20 10 0 2 4 8 12 16 24 36 48 Time (weeks) Edwin DeJesus et al. ICAAC 2007; abstract H-718b. Reprinted with permission.

  8. DRV/r qd LPV/r qd or bid ARTEMIS: Confirmed Response by Baseline Viral Load or CD4 at Week 48 (ITT-TLOVR) 100 100 †p<0.05 vs LPV/r 87 86 85 84 n=194 79† n=191 n=28 80 80 80 77 71 67 67 60 60 Patients with VL <50 copies/mL (%) Patients with VL <50 copies/mL (%) 40 40 20 20 0 0 0 <100,000 ≥100,000 <50 50–200 >200 Baseline viral load (copies/mL) Baseline CD4 cell count (cells/mm3) N = 226 226 117 120 N = 30 30 111 118 202 198 †Chi square analysis Edwin DeJesus et al. ICAAC 2007; abstract H-718b. Reprinted with permission.

  9. ARTEMIS: Virologic Failure (VF)and Emergence of Mutations DRV/r qd LPV/r qd or bid (N=343) (N=346) VF (> 50 cpm) 34 (10%) 49 (14%) VF (> 400 cpm) 11 (3%) 18 (5%) 10 18 Paired baseline and VF genotype available IAS-USA PI RAMS 0 1* 1† IAS-USA NRTI mutations 2† †184V *A71T, V77I VF by TLOVR *IAS-USA mutations, Fall 2006; Johnson et al. Topics in HIV Medicine. 2006; 14:125-130 Edwin DeJesus et al. ICAAC 2007; abstract H-718b. Reprinted with permission.

  10. ARTEMIS: Grade 2–4 Adverse Events (AEs) p<0.01 p<0.05 †At least possibly related to study drug, excluding laboratory-related events • No renal SAEs and no treatment discontinuations due to renal AEs Edwin DeJesus et al. ICAAC 2007; abstract H-718b. Reprinted with permission.

  11. ARTEMIS: Conclusions • The use of once-daily DRV/r 800/100mg + TDF/FTC in treatment-naïve patients: • resulted in excellent virologic and immunologic responses • provided suitable exposure in all patients • was well tolerated, with a favorable safety profile • In comparison to the LPV/r arm* in treatment-naïve patients: • For efficacy, DRV/r 800/100mg qd was non-inferior in the overall population, and superior in patients with high VL • DRV/r had lower incidence of common GI toxicities and triglyceride elevations *LPV/r arm included: LPV/r 400/100mg bid or 800/200mg qd, capsule and tablet formulations Edwin DeJesus et al. ICAAC 2007; abstract H-718b. Reprinted with permission.

  12. Antiretrovirals in Development for Treatment-Experienced Patients

  13. Antiretrovirals in Advanced Development

  14. Screening 6 weeks Follow up 4 weeks 600 patients target per trial DUET: Study Design and Major Inclusion Criteria • DUET-1 and -2 differed only in geographical location; pooled analysis was pre-specified • Plasma viral load >5,000 HIV-1 RNA copies/mL and stable therapy for ≥8 weeks • ≥1 NNRTI RAM, at screening or in documented historical genotype • ≥3 primary PI mutations at screening • Patients recruited from Thailand, Australia, Europe and the Americas 48-week treatment period with optional 48-week extension 24-week primary analysis TMC125 + BR* Placebo + BR* *BR = darunavir/ritonavir with optimised NRTIs and optional enfuvirtide BR = background regimen; RAM = resistance-associated mutation Pedro Cahn et al. ICAAC 2007; abstract H-717. Reprinted with permission.

  15. TMC125 + BR (n=599) Placebo + BR (n=604) DUET: Viral Load Reduction From Baseline (ITT NC=F) 0.0 –0.5 –1.0 –1.5 –2.0 –2.5 –3.0 –3.5 p<0.0001 Mean change in viral load from baseline (log10 copies/mL) ± SE –1.7 –2.4 0 4 8 12 16 20 24 Time (weeks) BR = background regimen; SE = standard error; ITT = intent-to-treat population; NC=F = noncompleter equals failureimputation algorithm; changes below the detection limit (<50 copies/mL) were imputed as 49 copies/mL Pedro Cahn et al. ICAAC 2007; abstract H-717. Reprinted with permission.

  16. TMC125 + BR (n=599) Placebo + BR (n=604) DUET: Change in CD4 Cell Count From Baseline (ITT NC=F) 100 75 50 25 0 +86 +67 p<0.0001 Mean change in CD4 cell count from baseline (cells/mm3) ± SE 0 4 8 12 16 20 24 Time (weeks) BR = background regimen; SE = standard error; ITT = intent-to-treat population; NC=F = noncompleter equals failureimputation algorithm Pedro Cahn et al. ICAAC 2007; abstract H-717. Reprinted with permission.

  17. TMC125 + BR (n=599) Placebo + BR (n=604) p=0.427 p<0.05 80 73% 62% 60 56% 34% 40 Adjusted for differences in baseline DRV FC between groups 20 Unadjusted response rates 0 Using de novo ENF DUET: Response (<50 copies/mL) According to Enfuvirtide (ENF) Use (Primary Analysis) p<0.0001 67% 62% Patients with viral load <50 copies/mL at Week 24 (%) DRV = darunavir FC = baseline fold change Re-using or not using ENF Using de novo ENF Pedro Cahn et al. ICAAC 2007; abstract H-717. Reprinted with permission.

  18. TMC125 + BR (n=545) Placebo + BR (n=559) DUET: Response (<50 copies/mL) According to Number of Active Background Antiretrovirals 45% 40/88 8%a 7/91 60% 120/199 Number of fully active background ARVs (PSS) 30% 63/211 74% 191/258 ≥ 67% 171/257 Patients with viral load <50 copies/mL at Week 24 (%) Analysis excludes patients who discontinued except for virological failure; PSS = phenotypic sensitivity score; darunavir and enfuvirtide are counted as fully active if FC<10 or used de novo, respectively Pedro Cahn et al. ICAAC 2007; abstract H-717. Reprinted with permission.

  19. DUET: Conclusions • In treatment-experienced patients, including those with NNRTI resistant virus, TMC125 consistently demonstrated superiority over placebo • 59% of patients achieved confirmed undetectable VL (<50 copies/mL) with TMC125 plus BR at Week 24 • Even in the absence of any other fully active background agents, with TMC125, 45% of patients achieved undetectable (<50 copies/mL)viral load • response rates increased as more active agents were used in the background regimen • 13 TMC125 resistance-associated mutations (TMC125 RAMs) were identified • the greatest added benefit in the TMC125 versus placebo group was seen in patients with <3 TMC125 RAMs • 86% patients had<3 TMC125 RAMs • Except for rash, incidence and severity of AEs with TMC125 were similar to placebo • TMC125 has the ability to extend and enhance the NNRTI class and provide a new treatment option for patients with resistance to other NNRTIs Pedro Cahn et al. ICAAC 2007; abstract H-717. Reprinted with permission.

  20. Interim Analysis of Part I Before Initiating Part II Total Part I Part II Integrase Monotherapy for 10 Days Combination Therapy ~ 30 pts ~ 8 pts ~ 38 pts Raltegravir 600 mg BID Raltegravir 600 mg BID + TDF/3TC ~ 8 pts ~ 30 pts ~ 38 pts Raltegravir 400 mg BID Raltegravir 400 mg BID + TDF/3TC ~ 8 pts ~ 30 pts ~ 38 pts Raltegravir 200 mg BID Raltegravir 200 mg BID + TDF/3TC ~ 8 pts ~ 30 pts ~ 38 pts Raltegravir 100 mg BID Raltegravir 100 mg BID + TDF/3TC ~ 30 pts ~ 8 pts ~ 38 pts Placebo BID Efavirenz 600 mg QD + TDF/3TC Part I cohort: Rx-naive patients stratified and randomized to integrase monotherapy or placebo for 10 days Part II cohort: Rx-naive patients stratified and randomized to combination therapy for 48 weeks Protocol 004: Study Design Adapted from Martin Markowitz et al. IAS 2007; abstract TUAB104.

  21. Protocol 005: Study Design Weeks 1-24 Weeks 25-48 Raltegravir 200 mg BID* (43) Raltegravir 400 mg BID* (178) Raltegravir 400 mg BID* (45) Raltegravir 600 mg BID* (45) Placebo BID* (45) All 178 HIV-infected participants had an HIV RNA > 5,000 copies/mL and documented resistance to three classes of oral ARTs at baseline. *All patients received optimized background therapy. Adapted from Beatrice Grinsztejn et al. ICAAC 2007; abstract H-713.

  22. Protocol 005: Patient and Optimized Background Therapy (OBT) Characteristics *By Phenosense GTNote: Enfuvirtide is not included in the phenotypic sensitivity score since there is no clinical cut-off. Adapted from Beatrice Grinsztejn et al. ICAAC 2007; abstract H-713.

  23. Protocol 005: Time to Loss of Virologic Response to Week 72: Raltegravir (MK-0518) All Doses Combined vs. Placebo 100 80 Raltegravir All Doses Combined 60 % of Patients Remaining HIV RNA <400 copies/mL 40 20 Placebo Placebo 0 0 2 4 8 12 16 24 32 40 48 56 64 72 Week # of Patients at Risk Raltegravir All Doses Combined* 133 122 122 122 120 113 100 95 92 86 69 43 12 Placebo* 45 11 11 11 9 9 8 6 5 5 4 3 1 * Plus optimized background therapy.Patients who never achieved HIV RNA <400 copies/mL were considered failures. Non-responders assigned failure at time 0. Adapted from Beatrice Grinsztejn et al. ICAAC 2007; abstract H-713.

  24. Protocol 005: Analysis of Raltegravir (MK-0518) Resistance • Study found no association between dose and/or drug concentration and resistance. • The factors decreasing likelihood of developing mutations at amino acids 148 alone, 155 alone, and either 148 or 155 were found to be: • Phenotypic sensitivity score > 0. • Lower viral load (≤ 100,000 copies/mL vs. > 100,000 copies/mL). • New use of enfuvirtide (T-20, Fuzeon) in optimized background therapy. From: Hazuda et al, XVI International HIV Drug Resistance Workshop, 2007, Barbados Adapted from Beatrice Grinsztejn et al. ICAAC 2007; abstract H-713.

  25. Protocol 005: Safety During Double-Blind Study Period • Raltegravir (MK-0518) safety profile for all doses similar to placebo • Liver function test abnormalities were uncommon • No grade 4 abnormalities for aspartate aminotransferase (AST), alanine transaminase (ALT) or alkaline phosphatase (ALP) • Grade 3 AST: 2/133 patients (1.5%) in all MK-0518 groups • Grade 3 ALT: 1/133 patients (0.8%) in all MK-0518 groups • Most clinical adverse events (AEs) were mild to moderate • 4 serious, drug-related clinical AEs • Acute pancreatitis after 2 doses, considered 2º to optimized background therapy (200 mg group) • Metabolic acidosis and renal insufficiency; sepsis; death(600 mg group) • Lacunar infarction by CT (placebo) • Worsening lipoatrophy (placebo) • 2 discontinuations due to drug-related AE • Elevated AST/ALT, considered 2º to optimized background therapy (200 mg group) • Lipoatrophy (placebo) Adapted from Beatrice Grinsztejn et al. ICAAC 2007; abstract H-713.

  26. Elvitegravir (EVG) Phase 2 Study Schema Andrew Zolopa et al. ICAAC 2007; abstract H-714. Reprinted with permission.

  27. Elvitegravir Study: HIV RNA < 50 copies/mL at Week 16 for Patients With First Use of T-20 Andrew Zolopa et al. ICAAC 2007; abstract H-714. Reprinted with permission.

  28. Week 16 HIV RNA < 50 copies/mL for Patients Receiving EVG/r 125 mg: Impact of Gradations in Activity of Optimized Background Therapy Andrew Zolopa et al. ICAAC 2007; abstract H-714. Reprinted with permission.

  29. OBT alone (N=209) MVC QD + OBT (N=414) MVC BID + OBT (N=426) MOTIVATE 1 and 2: Summary of Week 24 Efficacy Results Includes all patients who received at least one dose of study medication P<0.001* Difference: +49(95% CI: 31, 67) P<0.001*Difference: +51(95% CI: 33, 69) P<0.0001* P<0.0001* Patients (%) Mean change from baselinein CD4 count (cells/mm3) HIV-1 RNA <50 copies/mL† Mean Change from Baseline in CD4 Count‡ * versus OBT alone† HIV-1 RNA value imputed as baseline if missing or if patient discontinued before 24 weeks‡Last observation carried forward Elna van der Ryst et al. ICAAC 2007; abstract H-715. Reprinted with permission.

  30. R5* X4† NR/NP D/M*† Dual/mixed tropic virus population Only CCR5-tropic virus detected Only CXCR4-tropic virus detected Non-reportable/ non-phenotypable Characterization of Maraviroc Resistance in MOTIVATE 1 and 2: Study Overview OBJECTIVE: To study changes in HIV-1 tropism in patients who experienced treatment failure in the MOTIVATE 1 and 2 studies MOTIVATE 1 and MOTIVATE 2 Phase 3 studies in treatment-experienced patients (N=1,075) Tropism determined for all patients at screening, baseline, and all visits where VL>500 c/mL (Trofile™ assay, Monogram Biosciences) Assessment of CD4 count at failure, time of failure, and occurrence of Category C events by tropism result * CCR5-using virus; †CXCR4-using virus Elna van der Ryst et al. ICAAC 2007; abstract H-715. Reprinted with permission.

  31. D D D R X X R R D D X R X D R D X D R D D R X X R D R X R D X X R D R X D D D X R X R R R X X X R D R D D D X X R R R D D X R R R R R X R X X R X X R X X R X X X D R R X R R X D R R X X X X R X X X X R X X X R X R X R R X R D R X X R R X X D X X R R X R MOTIVATE 1 and 2: Viral Populations That May Exist Within a Patient A) Pure R5 X4 B) Mixed Dual/mixed (D/M) tropism Elna van der Ryst et al. ICAAC 2007; abstract H-715. Reprinted with permission.

  32. 500 400 300 200 100 0 -100 0 100 200 300 MOTIVATE 1 and 2: CXCR4-UsingClones Were Detected at Low Frequency in the Baseline Sample Patient T6 R5 R5 DM DM DM DM DM DM R5 R5 6 5 4 • CXCR4-using clones detected at baseline (7%) • No CCR5-tropic clones on treatment HIV-1 RNA (log10 copies/mL) 3 CD4 Count (cells/mm3) 2 1 Time Since First Administration (Day) Lewis M et al. XVI International HIV Drug Resistance Workshop, June 2007, Abstract 56 Elna van der Ryst et al. ICAAC 2007; abstract H-715. Reprinted with permission.

  33. R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R D D D D R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R D D D D D D D X R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R R D X D D MOTIVATE 1 and 2: Selective Inhibition of R5 Viruses Can Lead to a Change in Tropism Result to D/M or X4 • Trofile™ (like all resistance tests) measures relative proportions (not absolute amounts) of different viruses (Panel A) • Selective inhibition of a majority virus type, increases the sensitivity to detect the minor variant (Panel B) A B MVC D/M R5 Elna van der Ryst et al. ICAAC 2007; abstract H-715. Reprinted with permission.

  34. MOTIVATE 1 and 2: Selective Inhibition of R5 Viruses Can Lead to a Change in Tropism Result to D/M or X4 • Maraviroc selectively inhibits R5 virus • If maraviroc is administered as part of a sub-optimal regimen, pre-existing low (undetected) levels of D/M or X4 virus will emerge as the dominant viral population • Since the D/M or X4 virus is pre-existing, time to failure is shorter than with R5 virus (where maraviroc resistance must be selected de novo) • Similar to the rapid outgrowth of pre-existing (archived) drug-resistant virus when failed ARV therapy is reinitiated after treatment interruption • After withdrawal of maraviroc, selective pressure on R5 virus is removed, allowing R5 virus to re-emerge as the dominant population • Reversion to R5 takes approximately 16 weeks, consistent with loss of 3TC1 or enfuvirtide2 resistance after withdrawal of these ARVs 1. Deeks S, et al. J Infect Dis2005; 192:1537-44. 2. Deeks et al. J Infect Dis 2007;195:387-91. Elna van der Ryst et al. ICAAC 2007; abstract H-715. Reprinted with permission.

  35. MOTIVATE 1: Trial Design Randomization 1:2:2 N=601 OBT* + placebo OBT* + maraviroc (150 mg† QD) OBT* + maraviroc (150 mg† BID) Screening(6 weeks) 48w 0 • Stable pre-study ARV regimen, or no ARVs for ≥ 4 weeks • Resistance to and/or ≥ 6 months’ experience with ≥ one ARVfrom three classes (≥ two for PIs) 24w • Patient eligibility criteria: • R5HIV-1 infection • HIV-1RNA ≥5,000 copies/mL • Patients stratified by: • Enfuvirtide use in OBT • HIV-1RNA < and ≥100,000 copies/mL at screening * OBT = optimized background therapy of 3–6 ARVs (PK boosting doses of RTV not counted as an ARV) † Patients receiving a PI (except TPV) and/or delavirdine in their OBT received 150 mg dose of MVC, all other patients received 300 mg dose of MVC Jacob P. Lalezari et al. ICAAC 2007; abstract H-718a. Reprinted with permission.

  36. OBT alone (N=118) MVC QD + OBT (N=232) MVC BID + OBT (N=235) MOTIVATE 1 – Week 48:Mean Change From Baseline in HIV-1 RNA Includes all patients who received at least one dose of study medication Study week 24 48 Mean change in HIV-1 RNA from baseline (log10 copies/mL) Difference: -0.85* (97.5% CI: -1.22, -0.49) Difference: -0.79* (97.5% CI: -1.14, -0.44) Difference: -1.02* (97.5% CI: -1.39, -0.66) Difference: -0.92* (97.5% CI: -1.28, -0.57) HIV-1 RNA value imputed as baseline if patient discontinued before 48 weeks*Treatment difference vs OBT alone Jacob P. Lalezari et al. ICAAC 2007; abstract H-718a. Reprinted with permission.

  37. MOTIVATE 1 – Week 48:Percentage of Patients With Undetectable HIV-1 RNA Jacob P. Lalezari et al. ICAAC 2007; abstract H-718a. Reprinted with permission.

  38. OBT alone Maraviroc QD + OBT Maraviroc BID + OBT 59 59 91 91 108 108 30 30 75 75 72 72 ENF first use ENF first use ENF experienced/resistance ENF experienced/resistance MOTIVATE 1 and 2: Proportion of Patients Receiving ENF With Undetectable HIV-1 RNA at Week 48 According to ENF First Use <400 copies/mL <50 copies/mL Patients (%) N= Last observation carried forward Jacob P. Lalezari et al. ICAAC 2007; abstract H-718a. Reprinted with permission.

  39. MOTIVATE 1 and 2 – Week 48: Change in CD4+ Cell Count From Baseline by Tropism Result at Time of Failure * Includes patients with non-reportable/non-phenotypable tropism result at baseline and patients with non-reportable/non-phenotypable/missing tropism result at time of failure Jacob P. Lalezari et al. ICAAC 2007; abstract H-718a. Reprinted with permission.

  40. HIV Drug Resistance and theComplications of HIV/HAART

  41. TITAN: Development of Primary Protease Inhibitor Mutations and NRTI Resistance-Associated Mutations Upon Virologic Failure Simon F. De Meyer et al. ICAAC 2007; abstract H-1020. Reprinted with permission.

  42. TITAN: Loss of Susceptibility to Antiretrovirals in Virologic Failures (VFs) upon VF Simon F. De Meyer et al. ICAAC 2007; abstract H-1020. Reprinted with permission.

  43. Genotypic Results and Virological Response After Interruption of NNRTI-Based Treatment Adapted from Somnuek Sungkanuparph et al. ICAAC 2007; abstract H-368.

  44. Change in cGFR: Abacavir (ABC) vs Tenofovir (TDF) Christopher Polk et al. ICAAC 2007; abstract H-383. Reprinted with permission.

  45. Changes in Renal Function Among 10 Patients Categorized as Having “Current Renal Dysfunction” With Both Baseline and 12-Month Values, HIV Outpatient Study, November 2001 – September 2005 Benjamin Young et al. ICAAC 2007; abstract H-382. Reprinted with permission.

  46. ACTG 5102: Lipid Metabolism Pablo Tebas et al. ICAAC 2007; abstract H-378. Reprinted with permission.

  47. ACTG 5102: Changes in Immune Activation Pablo Tebas et al. ICAAC 2007; abstract H-378. Reprinted with permission.

  48. CPCRA 060: Time to CD4 < 350 cells/µL, Therapy Initiation or Death Matthew B. Goetz et al. ICAAC 2007; abstract H-1027. Reprinted with permission.

  49. Comparison of Luciferase Activity (RLUs) Between Standard and Enhanced Trofile Assays Jacqueline D. Reeves et al. ICAAC 2007; abstract H-1026. Reprinted with permission.

  50. Sensitivity to Detect Minor CXCR4-Using Subpopulations Jacqueline D. Reeves et al. ICAAC 2007; abstract H-1026. Reprinted with permission.

More Related