HIV article
 
Back 
 
 
Enfuvirtide Antiviral Activity despite Rebound Viremia and Resistance Mutations: Fitness Tampering or a Case of Persistent Braking on Entering? Editorial
 
 
  The Journal of Infectious Diseases Feb 1, 2007;195:318-321
 
Caryn Morse1 and Frank Maldarelli2
 
1Critical Care Medicine Department, Warren Grant Magnuson Clinical Center, and 2HIV Drug Resistance Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
 
(See the article
by Deeks et al.)
 
"....These data are provocative in suggesting that enfuvirtide may have partial antiviral activity in the presence of strong drug resistance mutations. The study poses 2 broad questions that converge on essential drug resistance issues...."
 
Combination antiretroviral therapy has revolutionized the care of HIV-1-infected individuals, producing profound improvements in morbidity and mortality. Therapy does not, however, cure HIV infection, and the goal of combination therapy remains the suppression of HIV-1 replication below the limit of detection. Despite a growing array of antiretroviral therapy options, virologic suppression is difficult for many patients to achieve, and treatment of drug-resistant virus is a critical challenge confronting HIV therapeutics.
 
Resistance emerges as a result of both virologic and clinical factors. HIV-1 replication is rapid and error prone, yielding a large, genetically diverse virus population. As a consequence, many single amino-acid changes conferring drug resistance may preexist as low frequency polymorphisms before the initiation of antiviral therapy [1, 2]. Individuals who received their diagnosis of HIV infection before the widespread use of combination antiretroviral regimens often received serial therapy with 1 or 2 agents, resulting in the accumulation of additional drug-resistance mutations to each new approved agent. Poor adherence to therapy has been associated with higher rates of virologic failure and the emergence of drug resistance, and sequential regimens for drug-resistant virus fail in patients [3-6] more often than initial regimens in drug-naive individuals [7-9].
 
Current therapeutic regimens to treat highly drug-resistant HIV-1 remain inadequate. The recommendation that regimens to treat drug-resistant HIV-1 should include at least 2 new drugs to which patients have not been exposed [10] is often impossible to fulfill if patients have extensive prior therapy experience. Recently, several studies have suggested the presence of residual virologic activity for some antivirals even in the presence of mutations [11-14]. In this issue of the Journal of Infectious Diseases, Deeks et al. [15] report the presence of virologic benefit of enfuvirtide (T-20) in the presence of rebound viremia and enfuvirtide resistance mutations.
 
Enfuvirtide is the first of a new class of antiretrovirals targeting membrane fusion, an essential energy-requiring step in HIV infection. CD4 receptor and coreceptor binding by glycoprotein (gp) 120 triggers conformational changes in gp41. The rearrangement of specific gp41 heptad-repeat regions 1 and 2 (HR-1 and -2, respectively) into a 6-helix bundle functions as a spring loaded mechanism to provide the energy necessary for membrane fusion. Enfuvirtide is a synthetic 36-aa peptide that binds to HR-1, disrupting interactions with HR-2 and interrupting the fusion reaction [16].
 
The clinical efficacy of enfuvirtide was demonstrated in 2 phase III studies, which demonstrated its superiority with an optimized antiretroviral background over optimized background alone (the T-20 vs. Optimized Regimen Only [TORO] studies) in highly treatment-experienced patients. Groups receiving regimens that contained enfuvirtide had significantly greater proportion of patients with a 1-log10 decrease in HIV-1 RNA levels, <400 copies HIV-1 RNA/mL plasma, and <50 copies HIV-1 RNA/mL plasma. CD4+ T cell increases were significantly greater in patients receiving regimens that contained enfuvirtide, compared with patients receiving optimized regimens only [17, 18]. Additional studies have documented that week-12 data predicted durable viral suppression at 24, 48, and 96 weeks of therapy [19, 20]. Enfuvirtide activity extends to HIV-1 group M (A, B, C, H, A/G, B/H, CRF01-AE, CRF02-AG, CRF05-DF, and CRF11-cpx) [21] and group O [22, 23].
 
Enfuvirtide is an injectable antiretroviral with a number of drawbacks: drug reconstitution can require up to 45 min-1 h, and twice-daily subcutaneous administration may be difficult for lipodystrophic individuals with limited available subcutaneous fat. Enfuvirtide is associated with significant side effects, notably injection-site reactions, and it is expensive. Because enfuvirtide has not been studied extensively in antiretroviral drug-naive patients, it received accelerated Food and Drug Administration approval in March of 2003 for use in treatment-experienced patients only.
 
Like all antiretrovirals, resistance emerges on therapy with enfuvirtide. HR regions have structural constraints that are genetically well conserved. Despite the relatively constant nature of HR-1 and HR-2, baseline susceptibility to enfuvirtide varies considerably; unlike pro-pol HIV-1 drug targets, which have a relatively narrow (<10-fold) variation in drug susceptibility in HIV-1, baseline susceptibility to enfuvirtide varies by >1000-fold in IC50 among untreated HIV-1 isolates [24, 25]. During therapy, a number of mutations (singly or in combination) in a region of HR-1 at 36-45 aa result in rebound viremia, including G36V, N42D or T, N43D, L44M, and L45M [21, 25-30]. The time frame of the emergence of drug resistance in monotherapy studies (2-3 weeks) suggests that such a number of single mutations preexisted in the virus population and were simply selected by therapy [2, 29]. Although the bulk of enfuvirtide resistance may be attributable to the region at 36-45 aa [28], sensitivity may be affected by regions outside heptad repeats [26, 28, 30, 31] and by receptor density and fusion rates [32]. It is not clear whether coreceptor sensitivity affects enfuvirtide resistance; the results of in vitro studies have suggested R5 viruses are intrinsically less sensitive to enfuvirtide; Melby et al. [33] reported a shift from dual-tropic to R5-tropic viruses in treated patients in the TORO-1 and -2 studies, which suggests that enfuvirtide may have greater activity against R4-tropic viruses.
 
Post hoc analyses of patients receiving enfuvirtide in the TORO trials and in other studies have demonstrated sustained CD4+ T cell responses even in patients who did not achieve a 1-log reduction in viral RNA levels, which suggests an immunologic benefit of continued therapy despite persistent viremia [20, 34]. Deeks et al. hypothesize that, if persistent virologic effect is present, discontinuing enfuvirtide would lead to an increase in HIV-1 viremia. Similar short-term ("shortstop") or long-term discontinuation strategies have been used to study the efficacy of the nucleoside reverse-transcriptase inhibitor, nonnucleoside reverse-transcriptase inhibitor, and protease inhibitor components of combination antiretroviral therapy [11-14, 35]. In the study by Deeks et al., patients receiving enfuvirtide for at least 24 weeks with persistent viremia >400 HIV copies/mL plasma were invited to enroll and discontinue enfuvirtide for 12 weeks. Those with >0.5-log increases in viral RNA levels or those with CD4+ T cell decreases of >50% were encouraged to restart enfuvirtide either in the context of a new or an existing drug background. Outcome measures included viral RNA levels, sequential genotypic and phenotypic analyses, and CD4+ T cell counts in peripheral blood.
 
Overall, patients who discontinued the enfuvirtide portion of their regimen had a significant increase in viral RNA levels; the greatest increases were detected within 4 weeks after discontinuation but were sustained over the course of 24 weeks (increase of 0.19 log10 HIV RNA copies/mL, repeated-measures regression model). In the majority of patients, virologic rebound occurred before phenotypic assays detected a shift in IC50, which suggests that enfuvirtide had suppressive activity against resistant virus. Patients were viremic during therapy and, overall, had decreasing CD4+ T cell counts. Slopes after the discontinuation of enfuvirtide were also negative but were not significantly different from the negative slope before therapy. In this pilot study, therefore, it is not known whether the modest but significant virologic effect had clinical or immunologic consequences.
 
These data are provocative in suggesting that enfuvirtide may have partial antiviral activity in the presence of strong drug resistance mutations. The study poses 2 broad questions that converge on essential drug resistance issues.
 
What is the virologic and molecular basis for continued drug efficacy in the presence of resistance mutations? In vitro assays presented by Deeks et al. and by other researchers have suggested that enfuvirtide-resistant envelopes exhibit delayed viral entry kinetics or reduced replicative capacity [36]. Other possibilities, including coreceptor shift [33] and replication or fitness differences [37], may help explain the observation, although studies of some enfuvirtide-resistant env suggest high replication capacity [38].
 
What is contribution of enfuvirtide drug levels to persistent antiviral effects? In phase II studies, trough levels varied by 6-fold and exceeded the IC50s of wild-type virus by 8-10 fold [39-41]. In the TORO studies, resistant virus emerged with IC50 values in range of 4 to >200-fold [25]. It is possible that enfuvirtide levels in vivo approximate the IC50s of some resistant isolates, which accounts for partial antiviral activity.
 
The clinical benefit of continuing enfuvirtide in the presence of rebound viremia remains uncertain. Persistence pays off, but for whom? Does the virologic improvement demonstrated in this trial translate into an immunologic or clinical benefit, or is it simply an opportunity for continued drug pressure to select progressively more resistant mutants? At present, these data and those presented elsewhere are not sufficiently compelling to expand indications for the use of enfuvirtide or to amend guidelines regarding its use. The temptation to regard this virologic effect as "intermediate" activity is not supported with the same robust clinical trial data that established enfuvirtide's activity, and it should be avoided for the purposes of reporting standard genotyping and phenotyping.
 
As Deeks et al. point out, larger trials are necessary to evaluate clinical and immunologic responses, the duration of the potential benefit, and the profile of adverse effects of continued enfuvirtide therapy in the setting of rebound viremia. Clinical end-point studies may be feasible in patients with advanced HIV-1 disease who are considering the use of enfuvirtide. Balanced against the cost, inconvenience, and potential adverse effects of continuing enfuvirtide in the setting of resistance, randomized trials, appropriately designed with a focus on sample size, inclusion criteria, and outcome measures, are warranted.
 
References
1. Bonhoeffer S, Nowak MA. Pre-existence and emergence of drug resistance in HIV-1 infection. Proc Biol Sci 1997; 264:631-7. First citation in article
| PubMed | CrossRef

2. Coffin JM. HIV population dynamics in vivo: implications for genetic variation, pathogenesis, and therapy. Science 1995; 267:483-9. First citation in article
| PubMed | CrossRef
3. Deeks SG, Hecht FM, Swanson M, et al. HIV RNA and CD4 cell count response to protease inhibitor therapy in an urban AIDS clinic: response to both initial and salvage therapy. AIDS 1999; 13:F35-43. First citation in article
| PubMed | CrossRef
4. Fatkenheuer G, Theisen A, Rockstroh J, et al. Virological treatment failure of protease inhibitor therapy in an unselected cohort of HIV-infected patients. AIDS 1997; 11:F113-6. First citation in article
| PubMed | CrossRef
5. Hammer SM, Vaida F, Bennett KK, et al. Dual vs single protease inhibitor therapy following antiretroviral treatment failure: a randomized trial. JAMA 2002; 288:169-80. First citation in article
| PubMed | CrossRef
6. Manfredi R, Chiodo F. Limits of deep salvage antiretroviral therapy with nelfinavir plus either efavirenz or nevirapine, in highly pre-treated patients with HIV disease. Int J Antimicrob Agents 2001; 17:511-6. First citation in article
| PubMed | CrossRef
7. Gulick RM, Ribaudo HJ, Shikuma CM, et al. Three- vs four-drug antiretroviral regimens for the initial treatment of HIV-1 infection: a randomized controlled trial. JAMA 2006; 296:769-81. First citation in article
| PubMed | CrossRef
8. Gulick RM, Ribaudo HJ, Shikuma CM, et al. Triple-nucleoside regimens versus efavirenz-containing regimens for the initial treatment of HIV-1 infection. N Engl J Med 2004; 350:1850-61. First citation in article
| PubMed | CrossRef
9. Walmsley S, Bernstein B, King M, et al. Lopinavir-ritonavir versus nelfinavir for the initial treatment of HIV infection. N Engl J Med 2002; 346:2039-46. First citation in article
| PubMed | CrossRef
10. US Department of Health and Human Services Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-infected adults and adolescents\October 10, 2006. Available at: http://aidsinfo.nih.gov/Guidelines/GuidelineDetail.aspx?MenuItem=Guidelines&Search=Off&GuidelineID=7&ClassID=1. Accessed 20 December 2006. First citation in article
11. Campbell TB, Shulman NS, Johnson SC, et al. Antiviral activity of lamivudine in salvage therapy for multidrug-resistant HIV-1 infection. Clin Infect Dis 2005; 41:236-42. First citation in article
| Full Text | PubMed 12. Deeks SG, Hoh R, Neilands TB, et al. Interruption of treatment with individual therapeutic drug classes in adults with multidrug-resistant HIV-1 infection. J Infect Dis 2005; 192:1537-44. First citation in article
| Full Text | PubMed 13. Maggiolo F, Callegaro A, Ripamonti D, et al. In vivo determination of stavudine activity in the presence of TAM [abstract 706]. In: Programs and abstracts of the 12th Conference on Retroviruses and Opportunistic Infections (Boston). Alexandria, VA: Foundation for Retrovirology and Human Health, 2005:323. First citation in article
14. Maldarelli FP, Palmer S, Kearney S, et al. Short duration, single drug discontinuation to assess the activity of individual drugs in patients failing antiretroviral therapy. Antivir Ther 2003; 8:S149. First citation in article
15. Deeks SG, Hoh R, Neilands TB, et al. Interruption of enfuvirtide in HIV-1-infected adults with incomplete viral suppression on an enfuvirtide-based regimen. J Infect Dis 2007; 195:387-91 (in this issue). First citation in article
16. Chen RY, Kilby JM, Saag MS. Enfuvirtide. Expert Opin Investig Drugs 2002; 11:1837-43. First citation in article
| PubMed | CrossRef
17. Lalezari JP, Henry K, O'Hearn M, et al. Enfuvirtide, an HIV-1 fusion inhibitor, for drug-resistant HIV infection in North and South America. N Engl J Med 2003; 348:2175-85. First citation in article
| PubMed | CrossRef
18. Lazzarin A, Clotet B, Cooper D, et al. Efficacy of enfuvirtide in patients infected with drug-resistant HIV-1 in Europe and Australia. N Engl J Med 2003; 348:2186-95. First citation in article
| PubMed | CrossRef
19. Nelson M, Arasteh K, Clotet B, et al. Durable efficacy of enfuvirtide over 48 weeks in heavily treatment-experienced HIV-1-infected patients in the T-20 versus optimized background regimen only 1 and 2 clinical trials. J Acquir Immune Defic Syndr 2005; 40:404-12. First citation in article
| PubMed | CrossRef
20. Raffi F, Katlama C, Saag M, et al. Week-12 response to therapy as a predictor of week 24, 48, and 96 outcome in patients receiving the HIV fusion inhibitor enfuvirtide in the T-20 versus Optimized Regimen Only (TORO) trials. Clin Infect Dis 2006; 42:870-7. First citation in article
| Full Text | PubMed 21. Van Laethem K, Schrooten Y, et al. A genotypic resistance assay for the detection of drug resistance in the human immunodeficiency virus type 1 envelope gene. J Virol Methods 2005; 123:25-34. First citation in article
| PubMed | CrossRef
22. Chinnadurai R, Munch J, Dittmar MT, Kirchhoff F. Inhibition of HIV-1 group M and O isolates by fusion inhibitors. AIDS 2005; 19:1919-22. First citation in article
| PubMed 23. Poveda E, Barreiro P, Rodes B, Soriano V. Enfuvirtide is active against HIV type 1 group O. AIDS Res Hum Retroviruses 2005; 21:583-5. First citation in article
| PubMed | CrossRef
24. Labrosse B, Labernardiere JL, Dam E, et al. Baseline susceptibility of primary human immunodeficiency virus type 1 to entry inhibitors. J Virol 2003; 77:1610-3. First citation in article
| PubMed | CrossRef
25. Melby T, Sista P, DeMasi R, Kirkland T, et al. Characterization of envelope glycoprotein gp41 genotype and phenotypic susceptibility to enfuvirtide at baseline and on treatment in the phase III clinical trials TORO-1 and TORO-2. AIDS Res Hum Retroviruses 2006; 22:375-85. First citation in article
| PubMed | CrossRef
26. Perez-Alvarez L, Carmona R, Ocampo A, et al. Long-term monitoring of genotypic and phenotypic resistance to T20 in treated patients infected with HIV-1. J Med Virol 2006; 78:141-7. First citation in article
| PubMed | CrossRef
27. Sista PR, Melby T, Davison D, et al. Characterization of determinants of genotypic and phenotypic resistance to enfuvirtide in baseline and on-treatment HIV-1 isolates. AIDS 2004; 18:1787-94. First citation in article
| PubMed | CrossRef
28. Su C, Melby T, DeMasi R, Ravindran P, Heilek-Snyder G. Genotypic changes in human immunodeficiency virus type 1 envelope glycoproteins on treatment with the fusion inhibitor enfuvirtide and their influence on changes in drug susceptibility in vitro. J Clin Virol 2006; 36:249-57. First citation in article
| PubMed | CrossRef
29. Wei X, Decker JM, Liu H, et al. Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion inhibitor (T-20) monotherapy. Antimicrob Agents Chemother 2002; 46:1896-905. First citation in article
| PubMed | CrossRef
30. Xu L, Pozniak A, Wildfire A, et al. Emergence and evolution of enfuvirtide resistance following long-term therapy involves heptad repeat 2 mutations within gp41. Antimicrob Agents Chemother 2005; 49:1113-9. First citation in article
| PubMed | CrossRef
31. Aquaro S, D'Arrigo R, Svicher V, et al. Specific mutations in HIV-1 gp41 are associated with immunological success in HIV-1-infected patients receiving enfuvirtide treatment. J Antimicrob Chemother 2006; 58:714-22. First citation in article
| PubMed | CrossRef
32. Kilby JM, Hopkins S, Venetta TM, et al. Potent suppression of HIV-1 replication in humans by T-20, a peptide inhibitor of gp41-mediated virus entry. Nat Med 1998; 4:1302-7. First citation in article
| PubMed | CrossRef
33. Melby T, Despirito M, Demasi R, Heilek-Snyder G, Greenberg ML, Graham N. HIV-1 coreceptor use in triple-class treatment-experienced patients: baseline prevalence, correlates, and relationship to enfuvirtide response. J Infect Dis 2006; 194:238-46. First citation in article
| Full Text | PubMed 34. Poveda E, Rodes B, Lebel-Binay S, Faudon JL, Jimenez V, Soriano V. Dynamics of enfuvirtide resistance in HIV-infected patients during and after long-term enfuvirtide salvage therapy. J Clin Virol 2005; 34:295-301. First citation in article
| PubMed | CrossRef
35. Maggiolo F, Callegaro A, Gregis G, et al. In vivo determination of tenofovir antiviral effect in the presence of TAM [abstract 707]. In: Programs and abstracts of the 12th Conference on Retroviruses and Opportunistic Infections (Boston). Alexandria, VA: Foundation for Retrovirology and Human Health, 2005:323. First citation in article
36. Reeves JD, Lee FH, Miamidian JL, Jabara CB, Juntilla MM, Doms RW. Enfuvirtide resistance mutations: impact on human immunodeficiency virus envelope function, entry inhibitor sensitivity, and virus neutralization. J Virol 2005; 79:4991-9. First citation in article
| PubMed | CrossRef
37. Lu J, Sista P, Giguel F, Greenberg M, Kuritzkes DR. Relative replicative fitness of human immunodeficiency virus type 1 mutants resistant to enfuvirtide (T-20). J Virol 2004; 78:4628-37. First citation in article
| PubMed | CrossRef
38. Labrosse B, Morand-Joubert L, Goubard A, et al. Role of the envelope genetic context in the development of enfuvirtide resistance in human immunodeficiency virus type 1-infected patients. J Virol 2006; 80:8807-19. First citation in article
| PubMed | CrossRef
39. Lalezari JP, DeJesus E, Northfelt DW, et al. A controlled phase II trial assessing three doses of enfuvirtide (T-20) in combination with abacavir, amprenavir, ritonavir and efavirenz in non-nucleoside reverse transcriptase inhibitor-naive HIV-infected adults. Antivir Ther 2003; 8:279-87. First citation in article
| PubMed 40. Lalezari JP, Patel IH, Zhang X, et al. Influence of subcutaneous injection site on the steady-state pharmacokinetics of enfuvirtide (T-20) in HIV-1-infected patients. J Clin Virol 2003; 28:217-22. First citation in article
| PubMed | CrossRef
41. Wheeler DA, Lalezari JP, Kilby JM, et al. Safety, tolerability, and plasma pharmacokinetics of high-strength formulations of enfuvirtide (T-20) in treatment-experienced HIV-1-infected patients. J Clin Virol 2004; 30:183-90. First citation in article
| PubMed | CrossRef
 
 
 
 
  icon paper stack View Older Articles Back to Top   www.natap.org