Abstract
Purpose of review
Current antiretroviral treatment regimens represent significant improvements in the management of HIV-1 infection; however, these regimens have not achieved a functional or sterilizing cure. One barrier to achieving a cure may be suboptimal antiretroviral concentrations in sanctuary sites throughout the body, including the central nervous system, gut-associated lymphoid tissue, lymph nodes, and tissue macrophages. This review will focus on the problems associated with achieving effective concentrations in these restricted sanctuary sites, and potential strategies to overcome these barriers.
Recent findings
Sufficient data exist to conclude that antiretroviral drug distribution is not uniform throughout the body. Low tissue/reservoir concentrations may be associated with viral replication. Multiple means to increase drug concentrations in sanctuary sites are being investigated, including modification of currently utilized drugs, blockade of transporters and enzymes that affect drug metabolism and pharmacokinetics, and local drug administration. Accumulating data suggest these methods increase antiretroviral concentrations in reservoirs of viral replication. No method has yet resulted in the complete clearance of HIV.
Summary
New strategies for increasing antiretroviral concentrations in predominant sites of viral replication may provide more effective means for elimination of viral sanctuaries. Additional research is necessary to optimize antiretroviral tissue distribution in order to inhibit virus replication fully, and avoid resistance and replenishment of viral reservoirs that may persist in the face of antiretroviral therapy.
Keywords: HIV-1, resistance, sanctuary, transporter
INTRODUCTION
The use of combination highly active antiretroviral therapy (ART) can achieve levels of HIV-1-RNA in plasma (viral load) that are undetectable by conventional assays in the vast majority of HIV-1-infected individuals [1–3]. In spite of this factor, there is growing evidence for residual viral replication in patients on suppressive ART [4▪,5]. There is also evidence for low-level viremia in ART-suppressed patients, although the origin of this low-level viremia and whether it is a result of residual viral replication is unclear [6]. Neither a sterilizing cure, defined as a complete elimination of HIV-1 from the body, nor a functional cure, in which there is no viral replication, but all traces of the virus have not been eliminated, has been achieved through ART [7]. Upon discontinuation of ART, there is a rapid recrudescence of viremia from long-lived reservoirs, leading to a decline in clinical parameters and the development of resistance [8,9,10▪▪]. There may be multiple obstacles to achieving complete clearance of the viral reservoirs, but one hypothesis, drawn from data demonstrating pharmacologic sanctuaries for antiretroviral (ARV) drugs in animals, is that concentrations of all agents in the ART regimens are not sufficient to fully suppress viral replication in all reservoir compartments. In this review we will discuss the locations and properties of these reservoirs, as well as potential strategies to achieve clinically effective concentrations in these reservoirs.
PHARMACOLOGIC SANCTUARIES
In general, drugs do not distribute evenly throughout the whole body [11▪,12,13]. The reasons for this are complex and multifactorial. Absorption and distribution vary by drug and by location depending on the physicochemical properties of the drug, the size and rate of perfusion of the target site, and protein-binding of the drug. Additional factors include if a drug is a substrate of influx or efflux transporters, metabolic enzymes, and patient-specific polymorphisms for transporters and metabolic enzymes. These complex factors can lead to significant differences in drug concentrations in different compartments, as well as different drug concentrations in these compartments between patients. Whereas measurements of drug concentrations in plasma are easy to obtain, the concentrations in restricted compartments and the sites of action for a given drug are considerably more complex to obtain and to quantify. One illustrative study utilized [18F]FPMPA, a radiolabeled tenofovir analog, to determine distribution in rats. [18F]FPMPA concentrations in spleens and mesenteric lymph nodes were two-fold lower than plasma concentrations, mesenteric lymph nodes and testes were four-fold lower, and brain concentrations were 25-fold lower than plasma concentrations [14]. Differences in drug concentrations in peripheral compartments may limit the inability of ART to fully suppress HIV-1 replication. In HIV-1-infected patients, Solas et al. [15] assessed concentrations of indinavir, nelfinavir, and lopinavir in plasma and a number of sanctuaries including cerebrospinal fluid (CSF), lymphoid tissue, and semen, and found considerable variation in drug concentrations between these sites, and between the various medications.
Gut- and rectal-associated lymphoid tissue
Lymphoid tissue in the gut (gut-associated lymphoid tissue, or GALT), as well as in the rectum (rectal-associated lymphoid tissue, or RALT) is the major site of viral replication in nonvirologically suppressed patients and may also serve as a sanctuary for HIV-1. Even in the presence of long-term ART, there are reports of HIV-1 residing in this tissue [16]. Chun et al. [16] have suggested that latently infected T cells in the gut comprise a key reservoir in HIV-1-infected patients. They observed significantly higher levels of HIV-1 DNA normalized to CD4+ T-cell count in GALT tissue than in either resting or activated CD4+ T cells in the blood [16]. They found high degrees of sequence similarity between virus in the GALT and virus in peripheral blood mononuclear cells (PBMCs), suggesting the gut provides a site for further infection of circulating PBMCs. Others have shown similarity between HIV-1 sequences in the gut and plasma in HIV-1-positive individuals, suggesting that even with undetectable HIV-1 RNA in plasma, these patients still have ongoing replication in the gut, and that there is equilibrium between these two compartments [17▪,18]. Additional studies suggest viral concentrations are higher along the entire gastrointestinal tract as compared with the blood of HIV-1+ patients treated with ART [19]. Ratios of unspliced RNA to DNA were also lower in the colon and rectum of these patients, compared with PBMCs, suggesting a low rate of HIV-1 transcription [19]. Latent virus in this reservoir can be released into the general circulation [4▪].
The causes for these differences in viral burden are likely multifactorial. One pharmacologic explanation is the expression of drug-metabolizing enzymes and transporters is not uniform along the gastrointestinal tract; this would be expected to give rise to regional differences in antiretroviral concentrations. For example, considerable differences in expression for transporters including BCRP, multi-drug resistant protein (MRP) 2, and P-glycoprotein (P-gp) exist among patients and different sections of the gut [20,21▪]. Small differences in P-gp, BCRP, OCTN2, and MCT1 expression occur along the length of the small intestine [20]. The colon has higher expression of MCT1, OCTN2, and MRP3 than the small intestine [20]. Human studies have shown considerable variation in cytochrome P450 (CYP) enzymes, including CYP3A expression along the length of the gastrointestinal tract, with the majority of CYP expression occurring in the proximal section of the gut [22,23]. These differences may contribute to the presence of sanctuary sites that afford the virus some degree of protection from antiretroviral suppression.
Lymph node
Mesenteric lymph nodes have been identified as a sanctuary site. Horiike et al. [10▪▪] found effective suppression of plasma viral RNA in simian immunodeficiency virus (SIV)-infected macaques treated with ART. However, when they examined tissue from these animals, they found detectable virus levels in the spleens and lymph nodes [10▪▪]. Detectable vDNA and vRNA in lymphoid tissue collected at necropsy has been shown in aviremic ART-treated macaques infected with a chimera of SIV containing HIV-1 reverse transcriptase [24]. These animals were treated with efavirenz, emtricitabine, and tenofovir for 26 weeks. Detectable virus was found in a variety of lymph nodes, including mesenteric, axial, inguinal, iliac, and cervical lymph nodes [24]. Bourry et al. [25] observed that in SIV-infected macaques treated with zidovudine/lamivudine/indinavir (ZDV/3TC/IDV), there were low concentrations of 3TC in lymphoid tissue. 3TC concentrations in these compartments were inversely correlated with virus persistence in these compartments [25]. Solas et al. [15] reported IDV concentrations in lymph nodes approximately two-fold greater than in plasma, whereas nelfinavir displayed a ratio of 0.58, lopinavir 0.21, and ritonavir 0.64 of drug concentration in the lymph node to drug concentration in plasma. The inability of the body to clear virus from this compartment in the presence of ART supports a hypothesis of suboptimal drug distribution and underscores one challenge to achieve complete antiretroviral suppression and eradication of the virus.
Central nervous system and genital tract
The central nervous system (CNS) and both the male and female reproductive tract are sanctuaries for HIV-1 [26,27,28▪,29–33]. ARV distribution to these compartments has been extensively discussed and this review will not focus on these sites.
Macrophages
Macrophages, in general, and intestinal macrophages, in particular, are thought to be a reservoir for HIV-1. Zalar et al. [18] assessed the presence of virus in duodenal tissue. Similar to the findings of others, they found detectable virus despite undetectable plasma viremia. They performed flow cytometry and PCR on duodenal macrophages, and found that 0.06% of the lamina propria mononuclear cells were HIV-1-infected [18]. Whereas this percentage appeared low, given the large number of cells in this location, this actually represented a significant number of infected cells. Infected macrophages may function as a ‘Trojan horse’ for the spread of virus throughout the body, most notably to the CNS [34]. Monocytes and macrophages express efflux transporters, including P-gp and MRP, which may result in decreased intracellular concentrations of antiretrovirals medications that are substrates [35–37]. Jorajuria et al. [37] investigated the antiretroviral properties of ZDV and IDV in human monocyte-derived macrophages. They observed greater inhibition of HIV-1 replication by ZDV and IDV in cells also treated with P-gp or MRP1 inhibitors [37]. This indicates that efflux transporters expressed on macrophages maintain sub-therapeutic concentrations in these cells, and explains a potential mechanism by which macrophages may function as a sanctuary for HIV-1. It is worth noting that differences exist in the intracellular environment between macrophages and T lymphocytes, which may result in some antiretrovirals working either more or less effectively in these cells [38].
OVERCOMING PHARMACOLOGIC SANCTUARIES
In patients on suppressive ART, the persistence of virus in sites such as the GALT, RALT, and lymph node may contribute to ongoing immune activation, incomplete immune reconstitution, abnormal vaccine responses, and reduced life expectancy compared with age-matched controls. This, coupled with evidence for poorer ARV distribution to these tissue sites, illustrates barriers to achieving either a functional or sterilizing cure for HIV-1 infection. Overcoming these pharmacologic sanctuaries with new drugs or new drug delivery approaches that achieve drug concentrations in lymphoid tissues that fully suppress virus replication should prevent maintenance of persistent virus reservoirs, and thereby improve prospects for full immune reconstitution and a functional or sterilizing cure of HIV-1 infection.
Compartment-targeted therapy
Whereas few treatments for HIV-1 have used the strategy of targeting specific compartments of ARVs for eliminating virus in these sanctuaries, this approach is being utilized for other diseases, and may hold promise for eliminating HIV-1 from restricted sanctuaries. The treatment of individuals with childhood acute lymphoblastic leukemia commonly includes intrathecal chemotherapy, with minimal long-term side effects [39]. Similarly, patients with cytomegalovirus retinitis have been treated with intravitreal ganciclovir [40]. In the treatment of HIV-1, local administration is primarily being investigated for microbicides, but may theoretically be of use for residual virus replication in sanctuary compartments. Studies assessing microbicides in primates treated with either intra-vaginal or intrarectal administration of tenofovir have displayed detectable concentrations for 24 h postadministration of the drug product [41]. Similar studies utilizing a 1% tenofovir gel vaginally in monkeys found concentrations in vaginal lymphocytes greater than the IC95 for the virus at 4 and 24 h postadministration [42].
Transporter and metabolism blockage
Strategies to increase concentrations in sanctuary sites include use of drugs to block influx and efflux transporters or the metabolism of ARVs. Ritonavir, and now cobicistat, have established roles in HIV pharmacotherapy as CYP3A enzyme inhibitors [43▪▪,44]. In-vitro research suggests that ritonavir and cobicistat also enhance intestinal absorption. Studies with Caco-2 monolayers (a cell line derived from human epithelial colorectal adenocarcinoma cells, commonly used in in-vitro absorption studies) show increased transport of ARVs including atazanavir, darunavir, and tenofovir alafenamide fumarate (GS-7340) across the monolayers [45▪▪]. This result is accomplished in part by inhibition of P-gp and BCRP. Similar findings have been observed for ritonavir [46▪▪]. This inhibition of efflux transporters in the gut may lead to increased concentrations in plasma, but it is not known if this increase will have a direct effect on drug concentrations in compartments. Namanja et al. [47▪] have recently reported an abacavir prodrug dimer that can inhibit P-gp efflux. The prodrug was designed to increase abacavir CNS penetration. They utilized in-vitro assays, as well as a brain capillary model, and found this molecule was able to inhibit P-gp in the cell culture system, and is converted back to abacavir inside cells [47▪]. They suggest that this approach may be utilized for other antiretrovirals as a means to increase penetration past the blood–brain barrier. P-gp blockers have been studied as adjunctive treatment in cancer, but no therapeutic benefit has been established [48].
Nanomedicine
Nano-scale antiretroviral therapies (nanoART), usually defined as ARVs formulated into particles with a size of less than 100 nm, have been proposed as an approach to increase concentrations of ARVs in sanctuary sites [49▪,50,51]. Human monocyte-derived macrophages (MDMs) exposed to nanoART consisting of atazanavir, efavirenz, and ritonavir show rapid uptake of the nanoART, and slow release over an extended time period (15–20 days) [52]. These nanoART products can be utilized to establish drug depots for a sustained response, allowing weekly, and even less frequent administration [49▪]. Significant concentrations of nanoART products are found in a variety of tissues, including the liver, spleen, kidneys, and lungs of animals for a number of days postadministration [49▪]. As circulating monocytes and macrophages distribute throughout the body, including to the brain and secondary lymphoid tissue, this factor has been proposed as a means to overcome barriers preventing access of antiretrovirals [52,53]. Nanoformulated ART engulfed by mononuclear phagocytes cocultured with human brain microvascular endothelial cells (HBMECs) can effectively transfer engulfed ART to these cells [54▪▪]. When the nanoART was coated with folate they found even higher uptake in the HBMECs; folate-coated nanoART given to mice achieved three-to four-fold greater concentrations in the brain than noncoated[54▪▪]. Kinman et al. [55] developed a lipid-IDV nanoparticle that resulted in a six-fold increase in IDV lymph node concentrations in macaques treated with the nanoparticle product as compared with a soluble formulation of IDV. Endsley and Ho [56▪▪] have recently described peptide-coated lipid nanoparticles of IDV as a means to target the nanoparticles to CD4+ cells, and observed significantly higher antiviral activity. The development of nanoproducts may play an important role in overcoming barriers to achieving adequate concentrations in sanctuary compartments and further study is warranted [57].
Modification of drugs and prodrugs and new drug development
The process of modifying the chemical structure of existing ARVs and developing new ARVs with improved drug distribution properties offers another approach to improve pharmacokinetic/pharmacodynamic properties in sanctuary sites. Tenofovir alafenamide fumarate (GS-7340) is a prodrug of tenofovir with improved penetration into lymphoid tissue in clinical development. [58]. The drug displays variable absorption, with larger doses resulting in a greater percentage of the dose being absorbed than with lower doses, and is very stable in intestinal tissue [59▪▪]. This situation is believed to be due to the ability of the drug to saturate intestinal transporters, including P-gp [59▪▪]. After oral administration, GS-7340 achieves concentrations in lymph nodes 5–15-fold higher than after oral administration of tenofovir disoproxil fumarate (TDF) [60]. GS-7340 produces intracellular concentrations in PBMCs approximately 150-fold higher than in plasma. This high degree of intracellular penetration results in GS-7340 having a selectivity index (a measure of the cytotoxic concentration to the effective concentration) approximately 10-fold higher than that of TDF [60]. Another example of drug formulation technology to enhance compartment penetration is a conjugate of zidovudine and ursodeoxycholic acid [61▪]. Ursodeoxycholic acid is capable of penetrating the blood brain barrier, and it is proposed this conjugate may allow increased concentration in the CNS. In-vitro studies utilizing human retinal pigment epithelium layers show higher permeation from the apical to basolateral compartments of the cells, as compared with the opposite, showing increased permeability of this drug product [61▪].
CONCLUSION
Accumulating data support the presence of pharmacologic sanctuaries and the possibility that they are an impediment to achieving a functional or sterilizing cure for HIV-1 infection with current ART. In sanctuaries such as the lymphoid tissue, antiretroviral concentrations appear to be insufficient to achieve full suppression of virus replication, and these sanctuaries appear capable of providing a site for development of resistance. Based on the fact that lymphoid tissues are the primary site of HIV-1 replication, the presence of these pharmacologic sanctuaries point to a fundamental gap in our understanding of ARV pharmacokinetic and pharmacodynamic characteristics necessary to achieve complete suppression of HIV-1 replication at its source. A number of strategies are available to improve drug concentration in these sanctuaries, either by increasing plasma concentrations, increasing drug penetration, or enhancing drug delivery. Additional research and development, and the translation of promising approaches to overcome pharmacologic sanctuaries into clinical investigations, are fundamental steps to a cure for HIV infection.
KEY POINTS.
Current antiretroviral therapies are unable to achieve complete suppression of viral replication throughout the body.
Residual viral replication and the development of resistance may arise in part because the concentrations of current antiretrovirals (ARVs) in sanctuary sites, such as lymphoid tissue, are suboptimal.
Potential strategies to improve antiretroviral penetration into sanctuaries include targeting sanctuary sites specifically, blockade of drug metabolism or influx/efflux transporters, modification of drug products to increase absorption and distribution, and nanomedicine.
Footnotes
Conflicts of interest
Supported in part by grants from the National Institute of Allergy and Infectious Diseases PO1 AI074340 (TWS, MS and CVF) and UO1 AI068636 (CVF).
REFERENCES AND RECOMMENDED READING
Papers of particular interest, published within the annual period of review, have been highlighted as:
▪ of special interest
▪▪ of outstanding interest Additional references related to this topic can also be found in the Current World Literature section in this issue (p. 251).
- 1.Choi JY, Song YG, Kim YH, et al. Factors associated with HIV-1 proviral DNA loads in patients with undetectable plasma RNA load. J Kor Med Sci. 2009;24:152–154. doi: 10.3346/jkms.2009.24.1.152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Natarajan V, Bosche M, Metcalf JA, et al. HIV-1 replication in patients with undetectable plasma virus receiving HAART. Lancet. 1999;353:119–120. doi: 10.1016/s0140-6736(05)76156-0. [DOI] [PubMed] [Google Scholar]
- 3.Verhofstede C, Van Wanzeele F, Reynaerts J, et al. Viral load assay sensitivity and low level viremia in HAART treated HIV patients. J Clin Virol. 2010;47:335–339. doi: 10.1016/j.jcv.2010.01.008. [DOI] [PubMed] [Google Scholar]
- 4▪.Buzón MJ, Codoñer FM, Frost SDW, et al. Deep molecular characterization of HIV-1 dynamics under suppressive HAART. PLoS Pathog. 2011;7:e1002314. doi: 10.1371/journal.ppat.1002314. The variance of HIV DNA sequences in patients was characterized, and suggests that virus reservoirs exist in these patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sharkey ME, Teo I, Greenough T, et al. Persistence of episomal HIV-1 infection intermediates in patients on highly active antiretroviral therapy. Nature Med. 2000;6:76–81. doi: 10.1038/71569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Palmer S, Maldarelli F, Wiegand A, 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. doi: 10.1073/pnas.0800050105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lewin SR, Rouzioux C. HIV cure and eradication: how will we get from the laboratory to effective clinical trials? Aids. 2011;25:885–897. doi: 10.1097/QAD.0b013e3283467041. [DOI] [PubMed] [Google Scholar]
- 8.Angelis DSA, Tateno AF, Diaz RS, et al. HIV-1 drug resistance genotypic profiles in children with undetectable plasma viremia during antiretroviral therapy. Braz J Infect Dis. 2011;15:60–65. doi: 10.1016/s1413-8670(11)70141-6. [DOI] [PubMed] [Google Scholar]
- 9.Ching N, Nielsen-Saines KA, Deville JG, et al. Autologous neutralizing antibody to human immunodeficiency virus-1 and replication-competent virus recovered from CD4+ T-cell reservoirs in pediatric HIV-1-infected patients on HAART. AIDS Res Human Retrovirus. 2010;26:585–591. doi: 10.1089/aid.2008.0274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10▪▪.Horiike M, Iwami S, Kodama M, et al. Lymph nodes harbor viral reservoirs that cause rebound of plasma viremia in SIV-infected macaques upon cessation of combined antiretroviral therapy. Virology. 2012;423:107–118. doi: 10.1016/j.virol.2011.11.024. This study examined reservoirs in macaques infected with SIV. They observed considerable amounts of vRNA in the lymph nodes, less in the lungs and intestines, and lower amounts in other tissues. When the animals had ART discontinued, virus rebound occurred, suggesting that the lymph node provides a sanctuary for virus replication in the presence of ART. [DOI] [PubMed] [Google Scholar]
- 11▪.Clavel C, Peytavin G, Tubiana R, et al. Etravirine concentrations in the cervicovaginal compartment in HIV-1-infected women receiving etravirine-containing antiretroviral therapy: DIVA 02 study. Antimicrob Agents Chemother. 2012;56:4018–4020. doi: 10.1128/AAC.06474-11. Cervicovaginal fluid from women receiving etravirine as part of their ART was assessed for drug levels and antiretroviral levels. They observed good penetration of etravirine into this compartment, as well as undetectable HIV RNA. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ivanovic J, Bellagamba R, Nicastri E, et al. Use of darunavir/ritonavir once daily in treatment-naive pregnant woman: pharmacokinetics, compartmental exposure, efficacy and safety. AIDS. 2010;24:1083–1084. doi: 10.1097/QAD.0b013e32833653b2. [DOI] [PubMed] [Google Scholar]
- 13.Kwara A, DeLong A, Rezk N, et al. Antiretroviral drug concentrations and HIV RNA in the genital tract of HIV-infected women receiving long-term highly active antiretroviral therapy. Clin Infect Dis. 2008;46:719–725. doi: 10.1086/527387. [DOI] [PubMed] [Google Scholar]
- 14.Di Mascio M, Srinivasula S, Bhattacharjee A, et al. Antiretroviral tissue kinetics: in vivo imaging using positron emission tomography. Antimicrob Agents Chemother. 2009;53:4086–4095. doi: 10.1128/AAC.00419-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Solas C, Lafeuillade A, Halfon P, et al. Discrepancies between protease inhibitor concentrations and viral load in reservoirs and sanctuary sites in human immunodeficiency virus-infected patients. Antimicrob Agents Chemother. 2003;47:238–243. doi: 10.1128/AAC.47.1.238-243.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Chun TW, Nickle DC, Justement JS, et al. Persistence of HIV in gut-associated lymphoid tissue despite long-term antiretroviral therapy. J Infect Dis. 2008;197:714–720. doi: 10.1086/527324. [DOI] [PubMed] [Google Scholar]
- 17▪.Imamichi H, DeGray G, Dewar RL, et al. Lack of compartmentalization of HIV-1 quasispecies between the gut and peripheral blood compartments. J Infect Dis. 2011;204:309–314. doi: 10.1093/infdis/jir259. The investigators here collected peripheral blood, as well as colon and ileum biopsy samples from patients treated with ART. They observed transcriptionally active HIV sequences in the gut of the patients, and similar transcripts in the blood of the patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zalar A, Figueroa MI, Ruibal-Ares B, et al. Macrophage HIV-1 infection in duodenal tissue of patients on long term HAART. Antiviral Res. 2010;87:269–271. doi: 10.1016/j.antiviral.2010.05.005. [DOI] [PubMed] [Google Scholar]
- 19.Yukl SA, Gianella S, Sinclair E, 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. doi: 10.1086/656722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Englund G, Rorsman F, Rönnblom A, et al. Regional levels of drug transporters along the human intestinal tract: co-expression of ABC and SLC transporters and comparison with Caco-2 cells. Eur J Pharm Sci. 2006;29:269–277. doi: 10.1016/j.ejps.2006.04.010. [DOI] [PubMed] [Google Scholar]
- 21▪.Tucker TGHA, Milne AM, Fournel-Gigleux S, et al. Absolute immunoquantification of the expression of ABC transporters P-glycoprotein, breast cancer resistance protein and multidrug resistance-associated protein 2 in human liver and duodenum. Biochem Pharmacol. 2012;83:279–285. doi: 10.1016/j.bcp.2011.10.017. The authors collected duodenal and liver samples from patients, and quantified expression of BCRP, MRP2, and P-gp. They found considerable variation between patients on expression of these transporters. [DOI] [PubMed] [Google Scholar]
- 22.Paine MF, Hart HL, Ludington SS, et al. The human intestinal cytochrome P450 ‘pie’. Drug Metab Dispos. 2006;34:880–886. doi: 10.1124/dmd.105.008672. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Zhang QY, Dunbar D, Ostrowska A, et al. Characterization of human small intestinal cytochromes P-450. Drug Metab Dispos. 1999;27:804–809. [PubMed] [Google Scholar]
- 24.North TW, Higgins J, Deere JD, et al. Viral sanctuaries during highly active antiretroviral therapy in a nonhuman primate model for AIDS. J Virol. 2010;84:2913–2922. doi: 10.1128/JVI.02356-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bourry O, Mannioui A, Sellier P, et al. Effect of a short-term HAART on SIV load in macaque tissues is dependent on time of initiation and antiviral diffusion. Retrovirology. 2010;7:78. doi: 10.1186/1742-4690-7-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Cu-Uvin S, Caliendo AM, Reinert S, et al. Effect of highly active antiretroviral therapy on cervicovaginal HIV-1 RNA. AIDS. 2000;14:415–421. doi: 10.1097/00002030-200003100-00015. [DOI] [PubMed] [Google Scholar]
- 27.Halfon P, Giorgetti C, Khiri H, et al. Semen may harbor HIV despite effective HAART: another piece in the puzzle. PloS One. 2010;5:e10569. doi: 10.1371/journal.pone.0010569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28▪.Launay O, Tod M, Tschöpe I, et al. Residual HIV-1 RNA and HIV-1 DNA production in the genital tract reservoir of women treated with HAART: the prospective ANRS EP24 GYNODYN study. Antiviral Ther. 2011;16:843–852. doi: 10.3851/IMP1856. Cervicovaginal samples from HIV+ women were assessed after the initiation of ART. Whereas many patients displayed undetectable HIV-1 RNA, many still had transient periods of viral shedding. [DOI] [PubMed] [Google Scholar]
- 29.Letendre S, Marquie-Beck J, Capparelli E, et al. Validation of the CNS penetration-effectiveness rank for quantifying antiretroviral penetration into the central nervous system. Arch Neurol. 2008;65:65–70. doi: 10.1001/archneurol.2007.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lorello G, La Porte C, Pilon R, et al. Discordance in HIV-1 viral loads and antiretroviral drug concentrations comparing semen and blood plasma. HIV Med. 2009;10:548–554. doi: 10.1111/j.1468-1293.2009.00725.x. [DOI] [PubMed] [Google Scholar]
- 31.Marcelin AG, Tubiana R, Lambert-Niclot S, et al. Detection of HIV-1 RNA in seminal plasma samples from treated patients with undetectable HIV-1 RNA in blood plasma. AIDS. 2008;22:1677–1679. doi: 10.1097/QAD.0b013e32830abdc8. [DOI] [PubMed] [Google Scholar]
- 32.Neely MN, Benning L, Xu J, et al. Cervical shedding of HIV-1 RNA among women with low levels of viremia while receiving highly active antiretroviral therapy. J Acquir Immune Defic Syndr. 2007;44:38–42. doi: 10.1097/01.qai.0000248352.18007.1f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zink MC, Brice AK, Kelly KM, et al. Simian immunodeficiency virus-infected macaques treated with highly active antiretroviral therapy have reduced central nervous system viral replication and inflammation but persistence of viral DNA. J Infect Dis. 2010;202:161–170. doi: 10.1086/653213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Meltzer MS, Skillman DR, Gomatos PJ, et al. Phagocytes in the pathogenesis of human immunodeficiency virus infection. Ann Rev Immunol. 1990;8:169–194. doi: 10.1146/annurev.iy.08.040190.001125. [DOI] [PubMed] [Google Scholar]
- 35.Dallas S, Schlichter L, Bendayan R. Multidrug resistance protein (MRP) 4-and MRP 5-mediated efflux of 9-(2-phosphonylmethoxyethyl) adenine by microglia. J Pharmacol Exp Ther. 2004;309:1221–1229. doi: 10.1124/jpet.103.063966. [DOI] [PubMed] [Google Scholar]
- 36.Dallas S, Zhu X, Baruchel S, et al. Functional expression of the multidrug resistance protein 1 in microglia. J Pharmacol Exp Ther. 2003;307:282–290. doi: 10.1124/jpet.103.054304. [DOI] [PubMed] [Google Scholar]
- 37.Jorajuria S, Dereuddre-Bosquet N, Becher F, et al. ATP binding cassette multidrug transporters limit the anti-HIV activity of zidovudine and indinavir in infected human macrophages. Antiviral Ther. 2004;9:519–528. [PubMed] [Google Scholar]
- 38.Gavegnano C, Schinazi RF. Antiretroviral therapy in macrophages: implication for HIV eradication. Antiviral Chem Chemother. 2009;20:63–78. doi: 10.3851/IMP1374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kadan-Lottick NS, Brouwers P, Breiger D, et al. Comparison of neurocognitive functioning in children previously randomly assigned to intrathecal methotrexate compared with triple intrathecal therapy for the treatment of childhood acute lymphoblastic leukemia. J Clin Oncol. 2009;27:5986–5992. doi: 10.1200/JCO.2009.23.5408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bakshi N, Fahle G, Sereti I, et al. Cytomegalovirus retinitis successfully treated with ganciclovir implant in a patient with blood ganciclovir resistance and ocular ganciclovir sensitivity. Eye. 2012;26:759–760. doi: 10.1038/eye.2012.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Nuttall J, Kashuba A, Wang R, et al. Pharmacokinetics of tenofovir following intravaginal and intrarectal administration of tenofovir gel to rhesus macaques. Antimicrob Agents Chemother. 2012;56:103–109. doi: 10.1128/AAC.00597-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Dobard C, Sharma S, Martin A, et al. Durable protection from vaginal simian-human immunodeficiency virus infection in macaques by tenofovir gel and its relationship to drug levels in tissue. J Virol. 2012;86:718–725. doi: 10.1128/JVI.05842-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43▪▪.Elion R, Cohen C, Gathe J, et al. Phase 2 study of cobicistat versus ritonavir each with once-daily atazanavir and fixed-dose emtricitabine/tenofovir df in the initial treatment of HIV infection. AIDS. 2011;25:1881–1886. doi: 10.1097/QAD.0b013e32834b4d48. This phase 2 trial compared cobicistat and ritonavir as pharmacoenhancers for two different antiretroviral regimens. Both regimens enhanced with either cobicistat or ritonavir displayed similar extents of virus replication inhibition. Similar findings were observed for adverse effects between cobicistat and ritonavir. [DOI] [PubMed] [Google Scholar]
- 44.Xu L, Liu H, Murray BP, et al. Cobicistat (GS-9350): a potent and selective inhibitor of human CYP3A as a novel pharmacoenhancer. ACS Medicinal Chem Lett. 2010;1:209–213. doi: 10.1021/ml1000257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45▪▪.Lepist EI, Phan TK, Roy A, et al. Cobicistat boosts the intestinal absorption of transport substrates, including HIV protease inhibitors and GS-7340, in vitro. Antimicrob Agents Chemother. 2012;56:5409–5413. doi: 10.1128/AAC.01089-12. The authors assessed the effects of cobicistat on transporter molecules, including P-gp and BCRP. They found that in in-vitro experiments, cobicistat increased flux of a number of antiretrovirals, including atazanavir, darunavir, lopinavir, and GS-7340. This suggests that in addition to CYP3A inhibitory effects, cobicistat may also have effects on antiretroviral absorption. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46▪▪.Holmstock N, Annaert P, Augustijns P. Boosting of HIV protease inhibitors by ritonavir in the intestine: the relative role of cytochrome P450 and P-glycoprotein inhibition based on Caco-2 monolayers versus in situ intestinal perfusion in mice. Drug Metab Dispos. 2012;40:1473–1477. doi: 10.1124/dmd.112.044677. This study examines the effect of ritonavir on P-gp function in vitro and in an in-situ intestinal perfusion model. They observed significant increases in saquinavir, indinavir, lopinavir, and darunavir in these models. The authors suggest that ritonavir may block P-gp function in addition to its traditionally viewed role as an inhibitor of CYP3A4. [DOI] [PubMed] [Google Scholar]
- 47▪.Namanja HA, Emmert D, Davis DA, et al. Toward eradicating HIV reservoirs in the brain: inhibiting P-glycoprotein at the blood-brain barrier with prodrug abacavir dimers. J Am Chem Soc. 2011;134:2976–2980. doi: 10.1021/ja206867t. The authors describe an abacavir dimer that they developed as a means to inhibit P-gp function at the blood–brain barrier. They observed that these dimers are able to inhibit P-gp efflux effectively in vitro. They additionally found that within cells the dimers dissociate into monomeric forms, allowing them to function. They suggest that this approach may be of use for other antiretrovirals. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Coley HM. Overcoming multidrug resistance in cancer: clinical studies of p-glycoprotein inhibitors. Methods Mol Biol. 2010;596:341–358. doi: 10.1007/978-1-60761-416-6_15. [DOI] [PubMed] [Google Scholar]
- 49▪.Roy U, McMillan JE, Alnouti Y, et al. Pharmacodynamic and antiretroviral activities of combination nanoformulated antiretrovirals in HIV-1-infected human peripheral blood lymphocyte: reconstituted mice. J Infect Dis. 2012;206:1577–1588. doi: 10.1093/infdis/jis395. The authors developed a nanoART system capable of a sustained response with weekly administration. When administered to mice with human peripheral blood lymphocytes, they observed that treatment decreased virus replication more potently than orally administered drugs. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Spreen W, Ford SL, Chen S, et al. Pharmacokinetics, safety and tolerability of the HIV integrase inhibitor S/GSK1265744 long acting parenteral nanosuspension following single dose administration to healthy adults; XIX International Aids conference; Washington DC, USA. 2012. [Google Scholar]
- 51.van et Klooster G, Hoeben E, Borghys H, et al. Pharmacokinetics and disposition of rilpivirine (TMC278) nanosuspension as a long-acting injectable antiretroviral formulation. Antimicrob Agents Chemother. 2010;54:2042–2050. doi: 10.1128/AAC.01529-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Nowacek AS, McMillan JE, Miller R, et al. Nanoformulated antiretroviral drug combinations extend drug release and antiretroviral responses in HIV-1-infected macrophages: implications for neuroAIDS therapeutics. J Neuroimmune Pharmacol. 2010;5:592–601. doi: 10.1007/s11481-010-9198-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Wong HL, Chattopadhyay N, Wu XY, et al. Nanotechnology applications for improved delivery of antiretroviral drugs to the brain. Adv Drug Deliv Rev. 2010;62:503–517. doi: 10.1016/j.addr.2009.11.020. [DOI] [PubMed] [Google Scholar]
- 54▪▪.Kanmogne GD, Singh S, Roy U, et al. Mononuclear phagocyte intercellular crosstalk facilitates transmission of cell-targeted nanoformulated antiretroviral drugs to human brain endothelial cells. Int J Nanomed. 2012;7:2373–2388. doi: 10.2147/IJN.S29454. The authors describe a nanoformulated ART product which can be internalized by macrophages. These macrophages can then transfer the nanoART past the blood–brain barrier. When the nanoART particles were coated with folate, they observed concentrations in the brain of mice 3–4 times higher than animals that received noncoated particles. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kinman L, Bui T, Larsen K, et al. Optimization of lipid-indinavir complexes for localization in lymphoid tissues of HIV-infected macaques. J Acquir Immune Defic Syndr. 2006;42:155–161. doi: 10.1097/01.qai.0000214822.33905.87. [DOI] [PubMed] [Google Scholar]
- 56▪▪.Endsley AN, Ho RJY. Enhanced anti-HIV efficacy of Indinavir after inclusion in CD4 targeted lipid nanoparticles. J Acquir Immune Defic Syndr. 2012;61:417–424. doi: 10.1097/QAI.0b013e3182653c1f. The authors report on peptide coated indinavir nanoparticles targeted to the CD4 receptor of T cells. These coated nanoparticles displayed increased antiviral activities as compared to soluble drug. The nanoparticles were shown to be specific for CD4+ cells by the utilization of a CD4 blocking antibody. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Schroeder A, Heller DA, Winslow MM, et al. Treating metastatic cancer with nanotechnology. Nature Rev Cancer. 2011;12:39–50. doi: 10.1038/nrc3180. [DOI] [PubMed] [Google Scholar]
- 58.Markowitz M, Zolopa A, Ruane P, et al., editors. GS-7340 Demonstrates greater declines in HIV-1 RNA than tenofovir disoproxil fumarate during 14 days of monotherapy in HIV-1 infected subjects; 18th Conference on Retroviruses and Opportunistic Infections (CROI 2011); Boston: 2011. [Google Scholar]
- 59▪▪.Babusis D, Phan TK, Lee WA, et al. Mechanism for effective lymphoid cell and tissue loading following oral administration of nucleotide prodrug GS-7340. Mol Pharm. 2012;10:459–466. doi: 10.1021/mp3002045. This study describes the distribution of GS-7340 and its active metabolite in plasma and in PBMCs. They describe that GS-7340 is capable of saturating intestinal efflux transporters as part of its absorption. They further describe the stability of the drug in intestinal and hepatic tissue. They finally describe efficient loading into PBMCs, and high levels of TFV-DP in those cells. [DOI] [PubMed] [Google Scholar]
- 60.Lee WA, He GX, Eisenberg E, et al. Selective intracellular activation of a novel prodrug of the human immunodeficiency virus reverse transcriptase inhibitor tenofovir leads to preferential distribution and accumulation in lymphatic tissue. Antimicrob Agents Chemother. 2005;49:1898–1906. doi: 10.1128/AAC.49.5.1898-1906.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61▪.Dalpiaz A, Paganetto G, Pavan B, et al. Zidovudine and ursodeoxycholic acid conjugation: design of a new prodrug potentially able to bypass the active efflux transport systems of the central nervous system. Molec Pharmaceutic. 2012;9:957–968. doi: 10.1021/mp200565g. The authors here describe a novel prodrug consisting of zidovudine and ursodeoxycholic acid as a means to increase drug penetration beyond the blood–brain barrier. They found increased permeability in in-vitro experiments, suggesting that this product can cross barriers, and can hydrolyze once it bypasses these barriers. [DOI] [PubMed] [Google Scholar]