HIV reservoirs - Project Inform


[PDF]HIV reservoirs - Project Informb1ca250e5ed661ccf2f1-da4c182123f5956a3d22aa43eb816232.r10.cf1.rackcdn.com...

2 downloads 131 Views 790KB Size

SPECIAL REVIEW

HIV reservoirs: pathogenesis and obstacles to viral eradication and cure Tae-Wook Chun and Anthony S. Fauci Plasma HIV viremia can be suppressed and maintained below the limits of detection for prolonged periods of time in the vast majority of HIV-infected individuals who receive antiretroviral therapy (ART). Thus, the clinical outcome for HIV-infected individuals who have access to these drugs is dramatically improved. However, ART alone cannot eradicate HIV in infected individuals and this impediment is likely in part due to the persistence of viral reservoirs in the peripheral blood and lymphoid tissues of infected individuals despite the suppression of plasma viremia. In recent years, major research efforts have been dedicated to a better understanding of the pathogenesis of persistent HIV infection and to the development of therapeutic strategies aimed at eradicating virus in infected individuals receiving ART. In this review, we discuss the pathophysiology of CD4þ T-cell HIV reservoirs, including recent advances in our understanding of the mechanisms of persistent viral infection and perspectives for eradication of HIV in ß 2012 Wolters Kluwer Health | Lippincott Williams & Wilkins infected individuals.

AIDS 2012, 26:1261–1268 Keywords: antiretorviral therapy, cure, eradication, HIV, viral reservoirs

Introduction Antiretroviral therapy (ART) dramatically suppresses HIV replication in the vast majority of infected individuals, resulting in a substantial decline in morbidity and mortality in both developed and developing nations where drugs are available [1–7]. Shortly after the wide availability of combination ART involving at least three drugs in 1996 and the demonstration that plasma viremia could be rapidly and profoundly suppressed to below the level of detection, optimism grew in the field that HIV could potentially be eradicated in infected individuals receiving ART for a few years [8]. However, the prospect for eradication of HIV diminished considerably in 1997 following the publication of three independent studies in which the persistence of a small but detectable pool of latently infected, resting CD4þ T cells carrying replication-competent virus was documented in virtually all study patients who had received clinically effective ART

for up to 3 years [9–11]. Moreover, a number of studies demonstrated rapid viral rebound shortly after discontinuation of therapy in infected individuals in whom profound and sustained suppression of plasma viremia had been achieved for prolonged periods [12–17]. This finding casts doubt on the feasibility of eradication of HIV by ART alone. These sobering observations subsequently led to the intensified pursuit of new avenues of research with a focus on the cellular and molecular characterization of the latent HIV reservoir and the development of strategies designed to eliminate persistently infected CD4þ T cells in individuals receiving ART. Following the initial observation over a decade ago that latently infected, resting CD4þ T cells persist in infected individuals receiving clinically effective ART [9–11, 18,19], considerable progress has been made regarding our understanding of the pathogenesis of HIV reservoirs in the era of successful ART (Fig. 1). However, the

Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA. Correspondence to Tae-Wook Chun, PhD, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10, Room 6A32, 9000 Rockville Pike, Bethesda, MD 20892, USA. Tel: +1 301 496 0890; fax: +1 301 402 5920; e-mail: [email protected] Received: 7 February 2012; revised: 6 March 2012; accepted: 21 March 2012. DOI:10.1097/QAD.0b013e328353f3f1

ISSN 0269-9370 Q 2012 Wolters Kluwer Health | Lippincott Williams & Wilkins

1261

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

1262

AIDS

2012, Vol 26 No 10 Presence of latent viral reservoir in infected individuals receiving ART9,10,11

Characterization of pre-integration latency (in vitro)26

Identification of resting CD4+ T cells carrying integrated HIV DNA (ex vivo)20

Description of HIV latency in cell lines23

1986

1987

Role of transcription factors in HIV latency25

1990

1991

1995

Identification of resting CD4+ T cells carrying preintegration complex (ex vivo)27 Quantitation of latent tissue viral reservoir and total HIV burden in infected individuals28

Development of single copy assay72

Establishment of latent viral reservoir during acute infection29 Rebound of plasma viremia despite treatment with IL-2 and/or anti-CD3 antibody30,85

Stability of latent viral reservoir18

1997

1998

Attempt to purge latent viral reservoir and/or antiCD3 antibody83,84,85

1999

2000

2001

2002

Role of latent viral reservoir in plasma viral rebound after discontinuation of ART16,17 Evidence for decay of latent viral reservoir in acute treated individuals32

Persistent plasma viremia originating from unknown viral reservoir74

Inability to eliminate latent viral reservoir using HDACi88,89,90

Evidence for on-going HIV replication in infected individuals receiving ART57

2003

2004

2005

Case report on eradication of HIV95

2006

2007

2008

Attempt to purge latent viral reservoir using HDACi97 Accelerated decay of latent viral reservoir upon intensification of ART52 Higher HIV burden in GALT vs blood of infected individuals receiving ART66,67.68.69

Gene therapy to achieve eradication of HIV93,94

Evidence for ongoing viral replication upon intensification of ART80,81

2009

2010

2011

Inability of intensified antiretroviral therapy to lower residual plasma viremia76,77,78,79 Homeostatic proliferation of latent viral reservoir as a mechanism of HIV persistence42

Fig. 1. Timeline. Highlights of research on basic and clinical aspects of HIV reservoirs conducted over the past two decades.

The latent HIV reservoir in resting CD4R T cells: a major impediment to the eradication of virus

integrated form of viral DNA in the genome of the host cells [20]. This study was followed by a comprehensive analysis of the frequency of resting CD4þ T cells carrying integrated HIV DNA that were capable of producing replication-competent virus upon cellular activation in both the blood and lymphoid tissues of untreated infected individuals [28]. This study estimated that the total body burden of the latent viral reservoir was very small (fewer than 10 million cells per infected person) and suggested at least two reasons why such infected cells could persist for extended periods in vivo. First, the latently infected, resting CD4þ T cells exhibited primarily a memory phenotype (CD45ROþ), implying that such infected cells could be long-lived. Second, resting CD4þ T cells carrying infectious HIV could do so without expressing viral antigens on their cell surface, thereby enabling them to escape recognition by the host immune system and to resist virus-induced cytopathic effects. Indeed, several follow-up studies demonstrated that the latent viral reservoir carrying replication-competent HIV persists in virtually all infected individuals receiving clinically effective ART. This observation casts serious doubt on the feasibility of eradicating virus by therapy alone [9–11,18,19].

The presence of latently infected, resting CD4þ T cells carrying integrated HIV DNA in infected individuals was first demonstrated in 1995 [20], 2 years prior to the report of effective ART in the clinical setting [21,22]. However, the concept of HIV latency had been previously introduced on the basis of the studies involving the establishment of HIV-infected cell lines expressing low levels of virus [23] and the use of various cytokines [24] and transcription factors [25] to reverse this state of viral latency. Subsequently, primary resting CD4þ T cells carrying unintegrated HIV DNA (preintegration latency) were shown to generate infectious virus upon cellular activation in vitro [26] and ex vivo [27]. Then, the concept of postintegration latency was introduced in a report in which a very small fraction of resting CD4þ T cells in viremic HIV-infected individuals were shown to carry the

Having established that the latent viral reservoir persists in infected individuals receiving ART despite profound and sustained suppression of plasma viremia, a series of studies was launched in the late 1990s and early 2000s with a goal of better understanding the pathophysiologic mechanisms involved in the establishment and maintenance of HIV latency. In particular, in anticipation of the possibility that early initiation of ART may prevent the generation of viral latency, studies were undertaken to determine the approximate length of time required for establishing a pool of latently infected, resting CD4þ T cells. It became evident that the latent HIV reservoir establishes itself very early during the course of infection. This conclusion was based on the observation that initiation of ARTas early as 10 days following the onset of clinical symptoms associated with primary HIV infection

prospects for eradicating HIV in a sizable proportion of infected individuals receiving ART remain unclear and will likely remain so until the precise mechanisms by which persistent HIV reservoirs are established and maintained are fully understood. In addition, the sources of persistent infection and rebounding plasma viremia upon discontinuation of therapy need to be identified as well as the mode of viral transmission/spread of infection within lymphoid tissues. Finally, the development of clinically relevant, novel therapeutic agents targeting persistently infected cells must be assessed in infected individuals receiving ART. This article will review the state of our knowledge of CD4þ T-cell-associated HIV reservoirs and discuss recent therapeutic developments aimed at eradicating the virus in infected individuals.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

HIV reservoirs: pathogenesis and viral eradication Chun and Fauci

did not prevent the establishment of a pool of latently infected, resting CD4þ T cells carrying replicationcompetent virus [29]. In addition, the latent viral reservoir was shown to expand rapidly following the re-emergence of plasma viremia after discontinuation of ART in HIV-infected individuals with undetectable plasma viremia [30]. There are a number of challenges associated with both identifying individuals during primary HIV infection [31] and initiating ART prior to the appearance of high levels of plasma viremia [29]. Nonetheless, several studies have attempted to determine the half-life of latently infected, resting CD4þ T cells and the duration of ART required to eliminate these infected cells in early-treated individuals. Two longitudinal studies demonstrated that the half-life of the latent viral reservoir was approximately 4–6 months in HIV-infected individuals who initiated ART during the acute/early phase of infection [32,33]. These studies projected that it would require at least 7–8 years of continuous therapy to eradicate HIV using the assumptions that ARTwas 100% effective at suppressing viral replication and that no other viral reservoirs existed in vivo [32,33]. These predictions were in sharp contrast to other longitudinal studies that primarily focused on infected individuals who initiated ART during the chronic phase of infection and in which the half-life of the latent viral reservoir exceeded 44 months. This observation suggested that even 60 years of ART may not be sufficient to completely eradicate HIV [18,19].

to date and is widely recognized as one of the major obstacles to achieving eradication of virus [43,44], low levels of HIV replication may persist in infected individuals despite their treatment with clinically effective ART [32,45–55]. In recent years, the lack of consensus on the life span of the latent viral reservoir [19,32,33] has sparked an intense debate regarding the possibility that low levels of HIV replication in subsets of CD4þ T cells in the lymphoid tissue may contribute to replenishment of a pool of infected resting CD4þ T cells. By this mechanism, the overall half-life of the viral reservoir could be indefinitely extended. In this regard, previous mathematical models have suggested that productively infected CD4þ T cells have a relatively short in-vivo half-life (<1 day) following initiation of ART [8,56]. The data indicate that such cells should no longer be present in infected individuals who had been receiving clinically effective ART for extended periods. However, activated CD4þ T cells, enriched at high purity from the blood of a viremic individual receiving ART, carry HIV proviral DNA and are capable of spontaneously producing virions in culture [57]. Furthermore, in this study, phylogenetic analyses of HIV env provided evidence for virologic cross-talk between infected resting and activated CD4þ T cells in the absence of detectable plasma viremia [57]. These findings raise the possibility that ongoing HIV replication may persist in nonresting CD4þ T-cell reservoirs in aviremic individuals.

Assuming that the latent viral reservoir persists in virtually all HIV-infected individuals receiving ART and that this reservoir is considered to be one of the major impediments to eradicating HIV [9–11,18,19], major efforts have been directed towards elucidating the mechanism(s) by which these infected cells persist in vivo. A number of cellular and molecular mechanisms that could explain HIV persistence have been proposed over the years. These include a paucity of cellular factors required for robust HIV expression in infected cells [34,35], modification of chromatin structure that prevents viral replication [36,37], methylation of the 50 long terminal repeat region of HIV proviral DNA [38,39], interference of HIV expression by the proximal host gene promoters [40,41], homeostatic proliferation regenerating latently infected cells in the absence of cell death [42], and reversion of productively infected, activated CD4þ T cells to the resting memory state without any specific mechanisms [28]. Most likely, more than one of these, or other yet to be identified mechanisms, may contribute to the maintenance of the latent viral reservoir in vivo.

Without treatment, HIV replicates primarily in the lymphoid tissues [58,59] with extremely high levels of viral replication [60–62] and extensive destruction of CD4þ T cells [63–65] in the gut-associated lymphoid tissue (GALT) of infected animals and humans. Recent studies have therefore shifted focus to the GALT as a major viral reservoir in infected individuals receiving ART. Such studies have demonstrated that the overall HIV burden is substantially higher in the GALT than in the blood of infected individuals receiving ART for long time periods [66–69].

Persistence of HIV in infected individuals receiving antiretroviral therapy: beyond the latent viral reservoir Although the pool of latently infected, resting CD4þ T cells has been the most extensively studied HIV reservoir

1263

Collectively, these findings suggest that low levels of ongoing viral replication, along with the persistence of HIV in the latent viral reservoir in the blood, may occur in the absence of detectable plasma viremia (<50 copies of HIV RNA). Therefore, strategies targeting productively infected cells in the tissue compartment must be developed to further decrease the number of infected cells. The recent introduction of newer classes of antiretroviral drugs [70,71] ([72], this issue) combined with the development of highly sensitive assays that are capable of detecting plasma-associated HIV RNA at the single copy level (below 50 copies of HIV RNA per ml of plasma) [73–76] should play an important role in studying and addressing the issue of persistent virus reservoirs. In this regard, intensification of conventional ART with newer classes of drugs was considered a possible approach to further reduce the residual pool of virus if low levels of

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

1264

AIDS

2012, Vol 26 No 10

ongoing viral replication were indeed occurring in treated infected individuals. Unfortunately, this line of investigation has led to further disagreement. On the one hand, several recent studies have demonstrated that intensification of ART does not further reduce residual plasma viremia [77–80]. Therefore, ongoing HIV replication did not appear to occur and reactivation of the latent viral reservoir seemed to be the main mechanism by which residual plasma viremia is maintained in infected individuals receiving ART. On the other hand, two studies taking a different approach have demonstrated an accumulation of circularized HIV DNA and a significant reduction in the level of cell-associated RNA in the blood [81] and GALT [82], respectively, upon intensification of ART in aviremic individuals. Furthermore, evidence for cell-to-cell transmission of HIV in the presence of antiretroviral drugs has been recently demonstrated [83]. This finding implies that low levels of ongoing HIV replication in the tissue compartment may not be detectable as plasma viremia. Given the conflicting data regarding the virologic outcome of intensification of ART, clinical studies involving comprehensive immunologic and virologic analyses of a large number of study participants will be necessary to better delineate the role of residual HIV replication in infected individuals receiving ART. In addition, the precise origin and clinical importance of residual plasma viremia need to be further elucidated to better understand the dynamics of HIV infection in the setting of effective ART.

Therapeutic strategies for the eradication of persistent viral reservoirs in HIV-infected individuals receiving antiretroviral therapy: bench to bedside A major thrust of HIV therapeutic research over the past few years has been the development of clinical strategies aimed at eradicating the virus in infected individuals who receive ART. The pursuit of a cure for HIV disease began in earnest over a decade ago with strategies that directly or indirectly stimulated latently infected, resting CD4þ T cells in vivo. These studies were based on the assumption that activation of the latent viral reservoir would result in rapid cell death due to HIV-induced cytopathic effects and that the infectious virus released by these activated cells would be contained by the administration of ART. In this regard, it has been shown that a combination of cytokines, namely IL-2, IL-6, and TNF-a, can induce HIV replication from the latent viral reservoir ex vivo [35]. Indeed, several clinical studies have attempted to purge the latent HIV reservoir in infected individuals receiving ART using immune-activating agents, such as IL-2 [84] and anti-CD3 antibody [85,86]. Among these approaches, one nonrandomized study demonstrated a marked diminution of the size of the pool of latently infected, resting CD4þ T cells in infected individuals

receiving ART with repeated cycles of intermittent IL-2 compared with those who received ART alone [84]. Of note, no resting CD4þ T cells carrying replicationcompetent HIV could be detected in the blood and lymphoid tissue of these patients [84]. Nonetheless, rapid rebound of plasma viremia was observed following empiric interruption of ART in these patients [30]; similar rebounds of viremia following interruption of ART were seen in patients treated with anti-CD3 antibody with ART [86]. Taken together, these results strongly suggest that strategies involving activation of the latent HIV reservoir with potent T-cell stimulators may not be sufficient to purge the virus from infected, resting CD4þ T cells and thus achieve total eradication of HIV infection. In recent years, molecular studies on the persistence of HIV in latently infected cells have suggested that repression of chromatin structure may play a role in the inhibition of HIV transcription and that inhibitors of histone deacetylases (HDACs) may allow expression of viral RNA from the latent viral reservoir [87,88] (Table 1). One of these HADC inhibitors, valproic acid (VPA), has been tested extensively in vitro and ex vivo as a potential candidate for purging the latent HIV reservoir. One study demonstrated a diminution of the size of the latent viral reservoir in a small number of infected individuals receiving ART [98]. However, several subsequent studies have demonstrated no measurable effect of VPA on the frequency of resting CD4þ T cells carrying latent HIV in vivo [89–91]. These conflicting results may be due in part to the fact that HDAC inhibitors are not potent stimulators of HIV expression in latently infected, resting CD4þ T cells and do not promote T-cell activation. Currently, different HADC inhibitors are being tested in clinical trials in order to investigate whether more potent agents, such as suberoylanilide hydroxamic acid [99], are better capable of purging the latent virus. In addition to the aforementioned therapeutic agents that target the latent HIV reservoir, the modification of host genetics for the purpose of generating HIV-resistant CD4þ T cells is under active investigation (Table 1). Such an approach was inspired by a recent case report from Germany describing an HIV-infected patient with acute myeloid leukemia who underwent multiple rounds of chemotherapy and stem cell transplants from a CCR5D32 homozygous donor. The virus appears to be completely eliminated in this individual [96,97]. Remarkably, the plasma viremia of this patient did not rebound following discontinuation of ART and his viral load has remained fully suppressed for several years in the absence of ART [96,97]. On the basis of the initial success in a mouse model [93,100], a number of clinical trials have been recently launched in which autologous CD4þ T cells are treated with a zinc-finger nuclease that is designed to specifically eliminate or reduce the expression of HIV coreceptors CCR5 or CXCR4 ex vivo [94,95]. These genetically modified, HIV-resistant CD4þ T cells were

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

[92]

[93–95]

Ineffectiveness on the latent viral reservoir Limited availability of human-tested therapeutic vaccines Low efficacy in infected individuals with compromised immune systems

Unknown fate of viral reservoir after transfusion with genetically modified cells

Cytolytic CD8þ T-cell-mediated killing upon recognition of viral antigen

Generation of HIV-resistant CD4þ T cells

Generation of HIV-resistant CD4þ T cells

Latently and/or productively infected CD4þ T cells

CD4þ T cells

CD4þ T cells

Conclusion

HDACi, histone deacetylase inhibitor; SAHA, suberoylanilide hydroxamic acid; VPA, valproic acid.

Bone marrow transplantation from CCR5D32 homozygous donor Stem cell transplantation

Difficulties in identifying HLA-matched donors Need for chemotherapy and/or immunosuppressive therapy prior to transplantation

Zinc finger nuclease-mediated elimination of HIV co-receptors Gene therapy

Need for chemotherapy prior to transfusion

Therapeutic vaccine Enhancement of anti-HIV immunity

1265

then transfused back into the autologous donors. Preliminary findings have demonstrated a repopulation and steady increase in modified CD4þ T cells in the blood and lymphoid tissues in some study participants [94,95]. However, the fate of preexisting HIV reservoirs in such patients remains to be determined as does the question of whether chemotherapy will be necessary to eliminate the wild-type CCR5þ/CXCR4þ CD4þ T cells that may harbor infectious virus prior to transfusion of genetically modified cells. Future clinical trials involving long-term follow-up and discontinuation of ART will shed light on the feasibility of HIV eradication using these novel, more targeted genetic approaches.

[96,97]

[89–91] Induction of HIV may not lead to cell death Minimal impact on productively and recently infected CD4þ T cells Induction of HIV expression HDACi (VPA, SAHA) Elimination of latent HIV reservoir

Latently infected resting CD4þ T cells

References Challenges Mode of action Target cells Agent/drug Approach

Table 1. Current therapeutic strategies aimed at eradicating HIV in infected individuals receiving antiretroviral therapy.

HIV reservoirs: pathogenesis and viral eradication Chun and Fauci

Despite more than a decade of intense research and recent therapeutic advances, achieving a true eradication of HIV remains a daunting challenge for the HIV/AIDS scientific community. The following key issues must be addressed and implemented by the scientists in the field if the eradication of HIV in infected individuals is to someday become a realistic goal (Fig. 2). First, it is necessary to gain a better understanding of the immunologic and virologic factors/mechanisms that contribute to the persistence of HIV in infected individuals receiving ART. In particular, the following need to be fully delineated: the precise mechanism by which the latent viral reservoir is established and maintained; the role of tissue-derived productively infected cells in the persistence of virus; the origin of rebounding plasma viremia upon discontinuation of therapy; and the effect of immune activation on viral burden in infected individuals receiving ART. Second, the impact of the use of aggressive ARTregimens administered as early as possible in the course of HIV infection on the establishment and size of the HIV reservoir and on our ability to ultimately eradicate this reservoir needs to be addressed. Third, clinically relevant and reproducible high-throughput virologic assays need to be developed in order to measure the efficacy of agents that are designed to target persistent HIV reservoirs. Fourth, strategies aimed at targeting both latent (purging agents) and nonlatent viral reservoirs (HIV-specific killing agents such as immunotoxins) may need to be pursued simultaneously in order to eradicate HIV. Given that single cells carrying infectious HIVare potentially capable of re-igniting viral replication in the absence of antiretroviral drugs, a variety of cell types including cells of monocyte/macrophage lineage in tissue compartments, such as the lymph nodes and other organs, need to be examined and fully characterized as a potential sanctuary for the virus. Finally, eradication strategies may need to include immune-based therapies, such as therapeutic vaccination [92] that are capable of boosting the host immune responses against HIV and thus contribute to the eradication process.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

1266

AIDS

2012, Vol 26 No 10 Use of aggressive drug regimens

Use of virus purging agents

Early initiation of antiretroviral therapy

Enhancement of anti-HIV immunity

Use of HIVspecific killing agents

Modification of host genetics Better understanding of HIV basic science

Fig. 2. Potential strategies for eradicating HIV in infected individuals receiving antiretroviral therapy.

Considering that a true sterilizing cure will be very difficult to achieve in a large proportion of infected individuals even in the best of circumstances involving early access to therapy, the possibility of a ‘functional’ cure should be pursued. In this case, the virus would not be entirely eradicated, but HIV-specific immune responses are boosted to prevent the rebound of plasma viremia upon discontinuation of ART. The early initiation of ART following acute infection would enhance the possibility of such an immune-based functional cure by minimizing the size of the viral reservoir. The maintenance of this functional cure may likely require multiple rounds of immune enhancement over several years to ensure that the small amount of residual HIV is not re-ignited in the absence of ART.

Acknowledgements Conflicts of interest There are no conflicts of interest.

References 1. Palella FJ Jr, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med 1998; 338:853–860. 2. Mocroft A, Ledergerber B, Katlama C, Kirk O, Reiss P, d’Arminio Monforte A, et al. Decline in the AIDS and death rates in the EuroSIDA study: an observational study. Lancet 2003; 362:22–29. 3. Walensky RP, Paltiel AD, Losina E, Mercincavage LM, Schackman BR, Sax PE, et al. The survival benefits of AIDS treatment in the United States. J Infect Dis 2006; 194:11–19. 4. The Antiretroviral Therapy Cohort Collaboration. Life expectancy of individuals on combination antiretroviral therapy in high-income countries: a collaborative analysis of 14 cohort studies. Lancet 2008; 372:293–299. 5. van Sighem AI, Gras LA, Reiss P, Brinkman K, de Wolf F. Life expectancy of recently diagnosed asymptomatic HIV-infected patients approaches that of uninfected individuals. AIDS 2010; 24:1527–1535.

6. Donnell D, Baeten JM, Kiarie J, Thomas KK, Stevens W, Cohen CR, et al. Heterosexual HIV-1 transmission after initiation of antiretroviral therapy: a prospective cohort analysis. Lancet 2010; 375:2092–2098. 7. Koenig SP, Rodriguez LA, Bartholomew C, Edwards A, Carmichael TE, Barrow G, et al. Long-term antiretroviral treatment outcomes in seven countries in the Caribbean. J Acquir Immune Defic Syndr 2012; 59:e60–e71. 8. Perelson AS, Essunger P, Cao Y, Vesanen M, Hurley A, Saksela K, et al. Decay characteristics of HIV-1-infected compartments during combination therapy. Nature 1997; 387:188–191. 9. Chun TW, Stuyver L, Mizell SB, Ehler LA, Mican JA, Baseler M, et al. Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc Natl Acad Sci U S A 1997; 94:13193–13197. 10. Wong JK, Hezareh M, Gunthard HF, Havlir DV, Ignacio CC, Spina CA, et al. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 1997; 278:1291–1295. 11. Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 1997; 278:1295–1300. 12. Davey RT Jr, Bhat N, Yoder C, Chun TW, Metcalf JA, Dewar R, et al. HIV-1 and T cell dynamics after interruption of highly active antiretroviral therapy (HAART) in patients with a history of sustained viral suppression. Proc Natl Acad Sci U S A 1999; 96:15109–15114. 13. Neumann AU, Tubiana R, Calvez V, Robert C, Li TS, Agut H, et al. HIV-1 rebound during interruption of highly active antiretroviral therapy has no deleterious effect on reinitiated treatment. Comet Study Group. AIDS 1999; 13:677–683. 14. Harrigan PR, Whaley M, Montaner JS. Rate of HIV-1 RNA rebound upon stopping antiretroviral therapy. AIDS 1999; 13:F59–F62. 15. Garcia F, Plana M, Vidal C, Cruceta A, O’Brien WA, Pantaleo G, et al. Dynamics of viral load rebound and immunological changes after stopping effective antiretroviral therapy. AIDS 1999; 13:F79–F86. 16. Chun TW, Davey RT Jr, Ostrowski M, Shawn Justement J, Engel D, Mullins JI, et al. Relationship between preexisting viral reservoirs and the re-emergence of plasma viremia after discontinuation of highly active antiretroviral therapy. Nat Med 2000; 6:757–761. 17. Zhang L, Chung C, Hu BS, He T, Guo Y, Kim AJ, et al. Genetic characterization of rebounding HIV-1 after cessation of highly active antiretroviral therapy. J Clin Invest 2000; 106: 839–845. 18. Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, Pierson T, et al. Latent infection of CD4R T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat Med 1999; 5:512–517. 19. Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, et al. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4R T cells. Nat Med 2003; 9:727–728. 20. Chun TW, Finzi D, Margolick J, Chadwick K, Schwartz D, Siliciano RF. In vivo fate of HIV-1-infected T cells: quantitative analysis of the transition to stable latency. Nat Med 1995; 1:1284–1290. 21. Gulick RM, Mellors JW, Havlir D, Eron JJ, Gonzalez C, McMahon D, et al. Treatment with indinavir, zidovudine, and lamivudine in adults with human immunodeficiency virus infection and prior antiretroviral therapy. N Engl J Med 1997; 337:734–739. 22. Hammer SM, Squires KE, Hughes MD, Grimes JM, Demeter LM, Currier JS, et al. A controlled trial of two nucleoside analogues plus indinavir in persons with human immunodeficiency virus infection and CD4 cell counts of 200 per cubic millimeter or less. AIDS Clinical Trials Group 320 Study Team. N Engl J Med 1997; 337:725–733. 23. Folks T, Powell DM, Lightfoote MM, Benn S, Martin MA, Fauci AS. Induction of HTLV-III/LAV from a nonvirus-producing T-cell line: implications for latency. Science 1986; 231:600– 602. 24. Poli G, Kinter A, Justement JS, Kehrl JH, Bressler P, Stanley S, et al. Tumor necrosis factor alpha functions in an autocrine manner in the induction of human immunodeficiency virus expression. Proc Natl Acad Sci U S A 1990; 87:782–785.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

HIV reservoirs: pathogenesis and viral eradication Chun and Fauci 25. Nabel G, Baltimore D. An inducible transcription factor activates expression of human immunodeficiency virus in T cells. Nature 1987; 326:711–713. 26. Zack JA, Arrigo SJ, Weitsman SR, Go AS, Haislip A, Chen IS. HIV-1 entry into quiescent primary lymphocytes: molecular analysis reveals a labile, latent viral structure. Cell 1990; 61:213–222. 27. Bukrinsky MI, Stanwick TL, Dempsey MP, Stevenson M. Quiescent T lymphocytes as an inducible virus reservoir in HIV-1 infection. Science 1991; 254:423–427. 28. Chun TW, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H, et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 1997; 387:183– 188. 29. Chun TW, Engel D, Berrey MM, Shea T, Corey L, Fauci AS. Early establishment of a pool of latently infected, resting CD4(R) T cells during primary HIV-1 infection. Proc Natl Acad Sci U S A 1998; 95:8869–8873. 30. Chun TW, Davey RT Jr, Engel D, Lane HC, Fauci AS. Re-emergence of HIV after stopping therapy. Nature 1999; 401:874–875. 31. Branson BM, Stekler JD. Detection of acute HIV infection: we can’t close the window. J Infect Dis 2012; 205:521-524. 32. Zhang L, Ramratnam B, Tenner-Racz K, He Y, Vesanen M, Lewin S, et al. Quantifying residual HIV-1 replication in patients receiving combination antiretroviral therapy. N Engl J Med 1999; 340:1605–1613. 33. Chun TW, Justement JS, Moir S, Hallahan CW, Maenza J, Mullins JI, et al. Decay of the HIV reservoir in patients receiving antiretroviral therapy for extended periods: implications for eradication of virus. J Infect Dis 2007; 195:1762– 1764. 34. Williams SA, Greene WC. Regulation of HIV-1 latency by T-cell activation. Cytokine 2007; 39:63–74. 35. Chun TW, Engel D, Mizell SB, Ehler LA, Fauci AS. Induction of HIV-1 replication in latently infected CD4R T cells using a combination of cytokines. J Exp Med 1998; 188:83–91. 36. Coull JJ, Romerio F, Sun JM, Volker JL, Galvin KM, Davie JR, et al. The human factors YY1 and LSF repress the human immunodeficiency virus type 1 long terminal repeat via recruitment of histone deacetylase 1. J Virol 2000; 74:6790–6799. 37. Ylisastigui L, Coull JJ, Rucker VC, Melander C, Bosch RJ, Brodie SJ, et al. Polyamides reveal a role for repression in latency within resting T cells of HIV-infected donors. J Infect Dis 2004; 190:1429–1437. 38. Blazkova J, Trejbalova K, Gondois-Rey F, Halfon P, Philibert P, Guiguen A, et al. CpG methylation controls reactivation of HIV from latency. PLoS Pathogens 2009; 5:e1000554. 39. Kauder SE, Bosque A, Lindqvist A, Planelles V, Verdin E. Epigenetic regulation of HIV-1 latency by cytosine methylation. PLoS Pathogens 2009; 5:e1000495. 40. Han Y, Lin YB, An W, Xu J, Yang HC, O’Connell K, et al. Orientation-dependent regulation of integrated HIV-1 expression by host gene transcriptional readthrough. Cell Host Microbe 2008; 4:134–146. 41. Lenasi T, Contreras X, Peterlin BM. Transcriptional interference antagonizes proviral gene expression to promote HIV latency. Cell Host Microbe 2008; 4:123–133. 42. Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med 2009; 15:893–900. 43. Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, Pomerantz RJ. The challenge of finding a cure for HIV infection. Science 2009; 323:1304–1307. 44. Trono D, Van Lint C, Rouzioux C, Verdin E, Barre-Sinoussi F, Chun TW, et al. HIV persistence and the prospect of long-term drug-free remissions for HIV-infected individuals. Science 2010; 329:174–180. 45. Furtado MR, Callaway DS, Phair JP, Kunstman KJ, Stanton JL, Macken CA, et al. Persistence of HIV-1 transcription in peripheral-blood mononuclear cells in patients receiving potent antiretroviral therapy. N Engl J Med 1999; 340:1614–1622. 46. Gunthard HF, Havlir DV, Fiscus S, Zhang ZQ, Eron J, Mellors J, et al. Residual human immunodeficiency virus (HIV) type 1 RNA and DNA in lymph nodes and HIV RNA in genital secretions and in cerebrospinal fluid after suppression of viremia for 2 years. J Infect Dis 2001; 183:1318–1327.

1267

47. Zhu T, Muthui D, Holte S, Nickle D, Feng F, Brodie S, et al. Evidence for human immunodeficiency virus type 1 replication in vivo in CD14(R) monocytes and its potential role as a source of virus in patients on highly active antiretroviral therapy. J Virol 2002; 76:707–716. 48. Izopet J, Cazabat M, Pasquier C, Sandres-Saune K, Bonnet E, Marchou B, et al. Evolution of total and integrated HIV-1 DNA and change in DNA sequences in patients with sustained plasma virus suppression. Virology 2002; 302:393–404. 49. Sharkey ME, Teo I, Greenough T, Sharova N, Luzuriaga K, Sullivan JL, et al. Persistence of episomal HIV-1 infection intermediates in patients on highly active antiretroviral therapy. Nat Med 2000; 6:76–81. 50. Strain MC, Gunthard HF, Havlir DV, Ignacio CC, Smith DM, Leigh-Brown AJ, et al. Heterogeneous clearance rates of longlived lymphocytes infected with HIV: intrinsic stability predicts lifelong persistence. Proc Natl Acad Sci U S A 2003; 100:4819–4824. 51. Natarajan V, Bosche M, Metcalf JA, Ward DJ, Lane HC, Kovacs JA. HIV-1 replication in patients with undetectable plasma virus receiving HAART. Highly active antiretroviral therapy. Lancet 1999; 353:119–120. 52. Ramratnam B, Ribeiro R, He T, Chung C, Simon V, Vanderhoeven J, et al. Intensification of antiretroviral therapy accelerates the decay of the HIV-1 latent reservoir and decreases, but does not eliminate, ongoing virus replication. J Acquir Immune Defic Syndr 2004; 35:33–37. 53. Patterson BK, McCallister S, Schutz M, Siegel JN, Shults K, Flener Z, et al. Persistence of intracellular HIV-1 mRNA correlates with HIV-1-specific immune responses in infected subjects on stable HAART. AIDS 2001; 15:1635–1641. 54. Delobel P, Sandres-Saune K, Cazabat M, Pasquier C, Marchou B, Massip P, et al. R5 to X4 switch of the predominant HIV-1 population in cellular reservoirs during effective highly active antiretroviral therapy. J Acquir Immune Defic Syndr 2005; 38:382–392. 55. Wightman F, Solomon A, Khoury G, Green JA, Gray L, Gorry PR, et al. Both CD31(R) and CD31 naive CD4(R) T cells are persistent HIV type 1-infected reservoirs in individuals receiving antiretroviral therapy. J Infect Dis 2010; 202:1738– 1748. 56. Perelson AS, Neumann AU, Markowitz M, Leonard JM, Ho DD. HIV-1 dynamics in vivo: virion clearance rate, infected cell life-span, and viral generation time. Science 1996; 271:1582–1586. 57. Chun TW, Nickle DC, Justement JS, Large D, Semerjian A, Curlin ME, et al. HIV-infected individuals receiving effective antiviral therapy for extended periods of time continually replenish their viral reservoir. J Clin Invest 2005; 115:3250– 3255. 58. Pantaleo G, Graziosi C, Demarest JF, Butini L, Montroni M, Fox CH, et al. HIV infection is active and progressive in lymphoid tissue during the clinically latent stage of disease. Nature 1993; 362:355–358. 59. Embretson J, Zupancic M, Ribas JL, Burke A, Racz P, TennerRacz K, et al. Massive covert infection of helper T lymphocytes and macrophages by HIV during the incubation period of AIDS. Nature 1993; 362:359–362. 60. Veazey RS, DeMaria M, Chalifoux LV, Shvetz DE, Pauley DR, Knight HL, et al. Gastrointestinal tract as a major site of CD4R T cell depletion and viral replication in SIV infection. Science 1998; 280:427–431. 61. Mattapallil JJ, Douek DC, Hill B, Nishimura Y, Martin M, Roederer M. Massive infection and loss of memory CD4R T cells in multiple tissues during acute SIV infection. Nature 2005; 434:1093–1097. 62. Li Q, Duan L, Estes JD, Ma ZM, Rourke T, Wang Y, et al. Peak SIV replication in resting memory CD4R T cells depletes gut lamina propria CD4R T cells. Nature 2005; 434:1148–1152. 63. Guadalupe M, Reay E, Sankaran S, Prindiville T, Flamm J, McNeil A, et al. Severe CD4R T-cell depletion in gut lymphoid tissue during primary human immunodeficiency virus type 1 infection and substantial delay in restoration following highly active antiretroviral therapy. J Virol 2003; 77:11708–11717. 64. Brenchley JM, Schacker TW, Ruff LE, Price DA, Taylor JH, Beilman GJ, et al. CD4R T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med 2004; 200:749–759.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

1268

AIDS

2012, Vol 26 No 10

65. Mehandru S, Poles MA, Tenner-Racz K, Horowitz A, Hurley A, Hogan C, et al. Primary HIV-1 infection is associated with preferential depletion of CD4R T lymphocytes from effector sites in the gastrointestinal tract. J Exp Med 2004; 200:761–770. 66. Yukl SA, Gianella S, Sinclair E, Epling L, Li Q, Duan L, et al. Differences in HIV burden and immune activation within the gut of HIV-positive patients receiving suppressive antiretroviral therapy. J Infect Dis 2010; 202:1553–1561. 67. Chun TW, Nickle DC, Justement JS, Meyers JH, Roby G, Hallahan CW, et al. Persistence of HIV in gut-associated lymphoid tissue despite long-term antiretroviral therapy. J Infect Dis 2008; 197:714–720. 68. Macal M, Sankaran S, Chun TW, Reay E, Flamm J, Prindiville TJ, et al. Effective CD4R T-cell restoration in gut-associated lymphoid tissue of HIV-infected patients is associated with enhanced Th17 cells and polyfunctional HIV-specific T-cell responses. Mucosal Immunol 2008; 1:475–488. 69. Sheth PM, Chege D, Shin LY, Huibner S, Yue FY, Loutfy M, et al. Immune reconstitution in the sigmoid colon after longterm HIV therapy. Mucosal Immunol 2008; 1:382–388. 70. Havlir DV. HIV integrase inhibitors: out of the pipeline and into the clinic. N Engl J Med 2008; 359:416–418. 71. Fatkenheuer G, Pozniak AL, Johnson MA, Plettenberg A, Staszewski S, Hoepelman AI, et al. Efficacy of short-term monotherapy with maraviroc, a new CCR5 antagonist, in patients infected with HIV-1. Nat Med 2005; 11:1170–1172. 72. Vella S, Schwartla¨nder B, Sow SP, Eholie SP, Murphy RL. The history of antiretroviral therapy and of its implementation in resource-limited areas of the world. AIDS 2012; 26:1231– 1241. 73. Palmer S, Wiegand AP, Maldarelli F, Bazmi H, Mican JM, Polis M, et al. New real-time reverse transcriptase-initiated PCR assay with single-copy sensitivity for human immunodeficiency virus type 1 RNA in plasma. J Clin Microbiol 2003; 41:4531–4536. 74. Palmer S, Maldarelli F, Wiegand A, Bernstein B, Hanna GJ, Brun SC, et al. Low-level viremia persists for at least 7 years in patients on suppressive antiretroviral therapy. Proc Natl Acad Sci U S A 2008; 105:3879–3884. 75. Bailey JR, Sedaghat AR, Kieffer T, Brennan T, Lee PK, WindRotolo M, et al. Residual human immunodeficiency virus type 1 viremia in some patients on antiretroviral therapy is dominated by a small number of invariant clones rarely found in circulating CD4R T cells. J Virol 2006; 80:6441–6457. 76. Chun TW, Murray D, Justement JS, Hallahan CW, Moir S, Kovacs C, et al. Relationship between residual plasma viremia and the size of HIV proviral DNA reservoirs in infected individuals receiving effective antiretroviral therapy. J Infect Dis 2011; 204:135–138. 77. Dinoso JB, Kim SY, Wiegand AM, Palmer SE, Gange SJ, Cranmer L, et al. Treatment intensification does not reduce residual HIV-1 viremia in patients on highly active antiretroviral therapy. Proc Natl Acad Sci U S A 2009; 106:9403–9408. 78. Gandhi RT, Zheng L, Bosch RJ, Chan ES, Margolis DM, Read S, et al. The effect of raltegravir intensification on low-level residual viremia in HIV-infected patients on antiretroviral therapy: a randomized controlled trial. PLoS Med 2010; 7:e1000321. 79. Hatano H, Hayes TL, Dahl V, Sinclair E, Lee TH, Hoh R, et al. A randomized, controlled trial of raltegravir intensification in antiretroviral-treated, HIV-infected patients with a suboptimal CD4R T cell response. J Infect Dis 2011; 203:960–968. 80. McMahon D, Jones J, Wiegand A, Gange SJ, Kearney M, Palmer S, et al. Short-course raltegravir intensification does not reduce persistent low-level viremia in patients with HIV-1 suppression during receipt of combination antiretroviral therapy. Clin Infect Dis 2010; 50:912–919. 81. Buzon MJ, Massanella M, Llibre JM, Esteve A, Dahl V, Puertas MC, et al. HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med 2010; 16:460–465. 82. Yukl SA, Shergill AK, McQuaid K, Gianella S, Lampiris H, Hare CB, et al. Effect of raltegravir-containing intensification on HIV burden and T-cell activation in multiple gut sites of HIVpositive adults on suppressive antiretroviral therapy. AIDS 2010; 24:2451–2460.

83. Sigal A, Kim JT, Balazs AB, Dekel E, Mayo A, Milo R, et al. Cellto-cell spread of HIV permits ongoing replication despite antiretroviral therapy. Nature 2011; 477:95–98. 84. Chun TW, Engel D, Mizell SB, Hallahan CW, Fischette M, Park S, et al. Effect of interleukin-2 on the pool of latently infected, resting CD4R T cells in HIV-1-infected patients receiving highly active antiretroviral therapy. Nat Med 1999; 5:651– 655. 85. Prins JM, Jurriaans S, van Praag RM, Blaak H, van Rij R, Schellekens PT, et al. Immuno-activation with anti-CD3 and recombinant human IL-2 in HIV-1-infected patients on potent antiretroviral therapy. AIDS 1999; 13:2405–2410. 86. Kulkosky J, Nunnari G, Otero M, Calarota S, Dornadula G, Zhang H, et al. Intensification and stimulation therapy for human immunodeficiency virus type 1 reservoirs in infected persons receiving virally suppressive highly active antiretroviral therapy. J Infect Dis 2002; 186:1403–1411. 87. He G, Margolis DM. Counterregulation of chromatin deacetylation and histone deacetylase occupancy at the integrated promoter of human immunodeficiency virus type 1 (HIV-1) by the HIV-1 repressor YY1 and HIV-1 activator Tat. Mol Cell Biol 2002; 22:2965–2973. 88. Williams SA, Chen LF, Kwon H, Ruiz-Jarabo CM, Verdin E, Greene WC. NF-kappaB p50 promotes HIV latency through HDAC recruitment and repression of transcriptional initiation. EMBO J 2006; 25:139–149. 89. Siliciano JD, Lai J, Callender M, Pitt E, Zhang H, Margolick JB, et al. Stability of the latent reservoir for HIV-1 in patients receiving valproic acid. J Infect Dis 2007; 195:833–836. 90. Sagot-Lerolle N, Lamine A, Chaix ML, Boufassa F, Aboulker JP, Costagliola D, et al. Prolonged valproic acid treatment does not reduce the size of latent HIV reservoir. AIDS 2008; 22:1125–1129. 91. Archin NM, Eron JJ, Palmer S, Hartmann-Duff A, Martinson JA, Wiegand A, et al. Valproic acid without intensified antiviral therapy has limited impact on persistent HIV infection of resting CD4R T cells. AIDS 2008; 22:1131–1135. 92. Rosenberg ES, Graham BS, Chan ES, Bosch RJ, Stocker V, Maenza J, et al. Safety and immunogenicity of therapeutic DNA vaccination in individuals treated with antiretroviral therapy during acute/early HIV-1 infection. PLoS One 2010; 5:e10555. 93. Holt N, Wang J, Kim K, Friedman G, Wang X, Taupin V, et al. Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo. Nat Biotechnol 2010; 28:839–847. 94. Lalezari J, Mitsuyasu R, Deeks S, Wang S, Lee G, Holmes M, et al. Successful and persistent engraftment of ZFN-M-R5-D autologous CD4 T cells (SB-728-T) in aviremic HIV-infected subjects on HAART. In: Proceedings of the 18th Conference on Retroviruses and Opportunistic Infections; 2011; Boston, MA. 95. Tebas P, Levine B, Binder G, Hoxie J, Collman R, Gregory P, et al. Disruption of CCR5 in zinc finger nuclease-treated CD4 T cells: phase I trials. In: Proceedings of the 18th Conference on Retroviruses and Opportunistic Infections; 2011; Boston, MA. 96. Hutter G, Nowak D, Mossner M, Ganepola S, Mussig A, Allers K, et al. Long-term control of HIV by CCR5Delta32/Delta32 stem-cell transplantation. N Engl J Med 2009; 360:692– 698. 97. Allers K, Hutter G, Hofmann J, Loddenkemper C, Rieger K, Thiel E, et al. Evidence for the cure of HIV infection by CCR5Delta32/Delta32 stem cell transplantation. Blood 2011; 117:2791–2799. 98. Lehrman G, Hogue IB, Palmer S, Jennings C, Spina CA, Wiegand A, et al. Depletion of latent HIV-1 infection in vivo: a proof-of-concept study. Lancet 2005; 366:549–555. 99. Archin NM, Espeseth A, Parker D, Cheema M, Hazuda D, Margolis DM. Expression of latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid. AIDS Res Human Retroviruses 2009; 25:207–212. 100. Perez EE, Wang J, Miller JC, Jouvenot Y, Kim KA, Liu O, et al. Establishment of HIV-1 resistance in CD4R T cells by genome editing using zinc-finger nucleases. Nat Biotechnol 2008; 26:808–816.

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.