Abstract
Nucleoside antiretroviral agents are chiral small molecules that have distinct advantages compared to other classes including long intracellular half-lives, low protein binding, sustained antiviral response when a dose is missed, and ease of chemical manufacture. They mimic natural nucleosides and target a unique but complex viral polymerase that is essential for viral replication. They remain the cornerstone of highly active antiretroviral therapy (HAART) and are usually combined with non-nucleoside reverese transcriptase and protease inhibitors to provide powerful antiviral responses to prevent or delay the emergence of drug-resistant human immunodeficiency virus (HIV). The pharmacological and virological properties of a selected group of nucleoside analogs are described. Some of the newer nucleoside analogs have a high genetic barrier to resistance development. The lessons learned are that each nucleoside analog should be treated as a unique molecule since any structural modification, including a change in the enantiomeric form, can affect metabolism, pharmacokinetics, efficacy, toxicity and resistance profile.
Keywords: Antiviral agents, Pharmacology, Nucleoside analogs, HIV
Nucleoside analogs as a class have a well-established regulatory history, with 11 currently approved by the US Food and Drug Administration (US FDA) for the treatment of human immunodeficiency virus (HIV), hepatitis B virus (HBV) or hepatitis C virus (HCV). The challenge in developing antiviral therapies is to inhibit viral replication without injuring the host cell. In HIV, a major key target for drug development is HIV reverse transcriptase (HIV-RT), a unique viral polymerase. This enzyme is active early in the viral replication cycle and converts the virus’ genetic information, which is stored as RNA into DNA by reverse transcription, a process necessary for continued viral replication. Nucleoside reverse transcriptase inhibitors (NRTI) mimic natural nucleosides. In the triphosphate form, each NRTI competes with one of the four naturally occurring 2′-deoxynucleoside 5′-triphosphates (dNTP), namely, dCTP, TTP, dATP or dGTP, for binding and DNA chain elongation near the active site of HIV-1 RT.
By 2006, eight HIV NRTI were approved by the US FDA including: Zidovudine® (ZDV, AZT), Videx® (ddI, didanosine), Hivid® (ddC, zalcitabine), Zerit® (d4T, stavudine), Ziagen® (abacavir sulfate, ABC), Epivir® (3TC, lamivudine), Emtriva® (FTC, emtricitabine) and Viread® (tenofovir disoproxil fumarate, TDF). These NRTI are the backbone of modern highly active antiretroviral therapies (HAART) (Schinazi, 1991; Yeni et al., 2004). Current first line HAART regimens combine two NRTI together with either a protease inhibitor (PI) or non-nucleoside reverse transcriptase inhibitor (NNRTI). These drug cocktails have markedly decreased mortality and morbidity from HIV-1 infections in the developed world (Carpenter et al., 2000; Erickson-Viitanen et al., 2003). Existing therapies cannot eradicate HIV infection because of the compartmentalization of the virus and its latent properties (Siliciano and Siliciano, 2004; Siliciano, 2005). In fact new data indicate that drug holidays in subjects that have undetected HIV in their serum are generally contraindicated and that a backbone of NRTI is essential to suppress viral recrudescence (Deeks et al., 2005). Therefore, chronic therapy remains the standard of care for the foreseeable future. Many regimens eventually fail, due primarily to lack of adherence to strict regimens, delayed toxicities and/or the emergence of drug-resistant HIV strains (Pereira and Paridaen, 2004). This review discusses ZDV, 3TC, FTC, TDF and ABC, five of the most commonly used potent antiretroviral drugs approved by the US FDA (Fig. 1). We will also discuss newer potent NRTI undergoing clinical evaluation, including Racivir [RCV, (±)-β−2′,3′-dideoxy-5-fluoro-3′-thiacytidine], Dexelvucitabine (β-d-2′,3′-didehydro-2′,3′-dideoxy-5-fluorocytidine, reverset, RVT, d-D4FC, DFC), Amdoxovir [AMDX, (−)-β-d-2,6-diaminopurine dioxolane, DAPD], AVX754 [SPD-754, (−)dOTC, (−)-2′-deoxy-3′-oxa-4′-thiocytidine], and two NRTI in preclinical development, 9-(β-d-1,3-dioxolan-4-yl)-2-aminopurine (APD) and (−)-(2R,4R)-1-(2-hydroxymethyl-1,3-dioxolan-4-yl)thymine (DOT) (Fig. 1).
Fig. 1.

Chemical structures of nucleoside approved by US FDA and under development.
1. NRTI efficacy
NRTI require phosphorylation by intracellular enzymes to their active fraudulent NRTI triphosphate (NRTI-TP) analog form, which compete with the natural dNTP for incorporation by HIV-1 RT into the elongating proviral DNA. Since all current NRTI lack a 3′-hydroxyl group, incorporation results in chain termination (Lipsky, 1996). The fact that a specific NRTI-TP competes with its natural nucleotide each time it appears in the active site near the reverse-transcribing viral RNA chain, produces multiple opportunities for NRTI blockade during a single round of reverse transcription. The long intracellular half-life of many NRTI-TP can result in a significant post-antibiotic effect, which may sometimes be exploited for designing more convenient dose regimens that are more forgiving when it comes to missed doses compared to PI and NNRTI (Sommadossi, 1982). Therefore, unlike NNRTI and PI, in which the antiviral pharmacodynamics is directly predicted by the between dose trough plasma concentrations (Cmin) (Acosta et al., 1999; Fletcher, 1999; Fletcher et al., 2000; Lorenzi et al., 1997; Stein et al., 1996), the efficacy of NRTI may be more related to maximal plasma concentrations (Cmax), which would drive cellular accumulation and phosphorylation of the NRTI to the long-lived triphosphate, unless phosphorylation is rate limiting (e.g., see ZDV below).
Factors that influence NRTI efficacy and dose frequency include: the drug concentration versus time profile in plasma (pharmacokinetics), the efficiency of cellular uptake of the NRTI levels of phosphorylation to NRTI-TP, and its inhibition constant (Ki) with the HIV-1 RT. For example, (−)-FTC has an 8–10h half-life (t1/2) in the plasma (Table 1). Therefore, based on an expected 24h trough concentration in plasma, a once a day dosing regimen may seem inadequate (~4×plasma t1/2 value=12% of the Cmax value). However, cellular levels of FTC-TP reach levels of ~4pmol/106 cells (approximately 20μM, assuming a volume of 0.1μl/106 cells), which is much greater than the Ki value (1.4μM) versus HIV-1 RT (Feng et al., 1999). The t1/2 of the active FTC-TP is 29–56h (Wang et al., 2004). Therefore, (−)-FTC can be dosed once daily. Likewise, 3TC accumulates 17.5±6.4pmol of 3TC-TP per 106 primary human peripheral blood mononuclear (PBM) cells (approximately 87.5μM) and the Ki value is 5M (Feng et al., 1999), and has an intracellular t1/2 of 15–16h. Therefore, 3TC may be administered once daily (300 mg) or twice daily (150 mg) (Solas et al., 1998). The active metabolite of TDF, TFV-diphosphate (TFV-DP), has an intracellular t1/2 of 60 to greater than 175h, which strongly supports once-daily dosing (Back et al., 2005; Hawkins et al., 2005; Moore et al., 1999b; Yuen et al., 2004). Plasma pharmacokinetics, recommended oral doses, maximal cellular concentrations, half-lives of the various NTP, Ki values, and expected maximal viral load reductions, when used in monotherapy, and common resistance mutations of the various NRTI in humans are summarized in Table 1.
Table 1.
Pharmacological properties of selected nucleoside reverse transcriptase inhibitors (NRTI)
| NRTI | |||||||||
|---|---|---|---|---|---|---|---|---|---|
| FDA approved | Clinical trial | ||||||||
| ZDVa | 3TCb | (—)-FTCc | TDFd | ABC/CBVe | RCVf | RVTg | DAPD/DXGh | AVX-754i | |
| Sponsor | GSK | GSK/Shire | Gilead sciences | Gilead sciences | GSK | Pharmasset Inc. | Pharmasset Inc. | RFS Pharma LLC | Avexa Ltd. |
| Competing natural nucleosides | dThd | dCyd | dCyd | dAdo | dGuo | dCyd | dCyd | dGuo | dCyd |
| Mean single dose AUC (μg/ml × h) | 2.01 | 16.6 | 11.8 ± 2.9 | 2.29–3.02 | 5.31–5.48 | 10.7 ± 1.9 | 11.73 ± 2.08 | N/Aj | N/A |
| Cmax (μg/ml) | 1.64 | 3.5 | 2.2 ± 0.6 | 3.26 | 0.64–4.38 | 2.6 ± 0.4 | 1.75 ± 0.36 | N/A | N/A |
| t1/2 in plasma (h) | 0.5–3 | 5–7 | 8–10 | 12–17 | 1.54 ± 0.63 | 6.67 ± 1.48 | 5.2 | 4–7 | N/A |
| EC50 vs. HIV-1 in stimulated PBM cells (μM) | 0.01–0.09 | 0.003–0.15 | 0.001–0.69 | 0.01–0.02 | 0.91–0.98 | 0.007–0.087 | 0.6 ± 0.3 | DAPD 4; DXG 0.11–0.25 | N/A |
| Intracellular t1/2 of NTP (in HIV infected humans, h) | 3–4 | 15–16 | 29–56 | 60 to >175 | 12–21 | N/A | 13–17 (in vitro) | 9.4 | 6–7 |
| Ki of NTP vs. wild type HIV-1-RT enzyme (μM) | 0.035 | 5.0 ± 0.8 | 1.4 | 0.21 ± 0.04 | 0.11 ± 0.03 | N/A | 0.1 | 0.05 | N/A |
| Ki-DNA pol γ (μM) | 187 ± 54 | 9.2 ± 0.9 | 62.9 ± 8.4 | 40.3 ± 5.7 | 13.0±1.5 | N/A | 0.82 ± 0.2 | 4.3 ± 0.4 | N/A |
| Toxicity indexk (in vitro) | 1.05 | 1.2 | N/A | 2.1 | 1.0 | N/A | 662 | N/A | N/A |
| NTP-levels in PBM cells (pmol/106 cells) measured infected humans | 0.07–0.024 | 17.5 ± 6.4 | 1.9–4 | 0.085–0.11 | 0.09–0.20 | N/A | N/A | 0.7 | 1.65–6.10 |
| Recommended oral dose for HIV | 300 mg bid | 300 mg qd and 150 mg bid | 200 mg qd | 300 mg qd | 600 mg qd | 600 mg qd | 200 mg qd | 500 mg bid | 200–800 mg bid 800–1200 mg qd |
| Maximal log10—copies/ml reduction in viral load (monotherapy) | 0.85 (14 days) | 2.25 (14 days) | 1.9 (14 days) | 1.5 (14 days) | 1.1–1.8 (4 weeks) | 2.0–2.4 (day 14) | 1.77 (10 days) | 1.3–1.5(14 days) | 1.65 (10 days) |
| Signature resistance mutations selected in vitro | TAMs | M184V | M184V | K65R | M184V, L74V, K65R, Y115F | M184V | K65R | K65R or L74V | K65R, V75I, M184V |
| Activity vs. HBV in culture | No | Yes | Yes | Yes | No | Yes | No | Yes | N/A |
| Phase of development | Approved 1986, generic 2005 | Approved 1995 | Approved 2003 | Approved 2001 | Approved 1998 | 2b | 2b | 2b | 2b |
Abbreviations: Zidovudine® (ZDV, AZT), Epivir® (3TC, lamivudine), Emtriva® (FTC, emtricitabine), Viread® (tenofovir disoproxil fumarate, TDF), Ziagen® (abacavir sulfate, ABC), Carbovir® (CBV), Racivir [RCV, (±)-β−2′,3′-dideoxy-5-fluoro-3′-thiacytidine], Dexelvucitabine® (β-d-2′,3′-didehydro-2′,3′-dideoxy-5-fluorocytidine, reverset, RVT, d-D4FC, DFC), Amdoxovir® [AMDX, (−)-β-d-2,6-diaminopurine dioxolane, DAPD], 9-(beta-d-1,3-Dioxolan-4-yl)guanine (DXG), AVX754 [SPD-754, (−)dOTC, (−)-2′-deoxy-3′-oxa-4′-thiocytidine].
[Combivir package insert. GlaxoSmithkline]; (Anderson et al., 2004; Back et al., 2005; Barreiro et al., 2004; Cannard et al., 1998; Johnson et al., 2001; Murakami et al., 2004; Piliero, 2004; Reardon and Miller, 1990; Sharma et al., 2004; Wang et al., 1999).
[Combivir package insert. GlaxoSmithkline]; (Anderson et al., 2004; Back et al., 2005; Barreiro et al., 2004; Hawkins et al., 2005; Herzmann et al., 2005; Johnson et al., 1999, 2001; Moore et al., 1999b; Murakami et al., 2004; Piliero, 2004; Sharma et al., 2004; Solas et al., 1998; Wang et al., 1999; Yuen et al., 2004).
[Emtriva package insert. Gilead Sciences]; (Anderson et al., 2004; Back et al., 2005; Bang and Scott, 2003; Barreiro et al., 2004; Dando and Wagstaff, 2004; Feng et al., 2004a,b; Molina et al., 2004; Sharma et al., 2004; Wang et al., 2004).
(Anderson et al., 2004; Back et al., 2005; Barditch-Crovo et al., 2001; Barreiro et al., 2004; Hawkins et al., 2005; Johnson et al., 2001; Louie et al., 2003; Murakami et al., 2004; Piliero, 2004; Robbins et al., 1998; Sharma et al., 2004; Suo and Johnson, 1998; White et al., 2002, 2005).
[Ziagen package insert. GlaxoSmithkline]; (Anderson et al., 2004; Back et al., 2005; Hawkins et al., 2005; Hervey and Perry, 2000; Johnson et al., 2001; Murakami et al., 2004; Piliero, 2004; Sharma et al., 2004; Wang et al., 1999; White et al., 2005).
http://www.natap.org/2004/pdf/posRep04.pdf; (Murakami et al., 2004; Otto, 2004; Schinazi et al., 2002; Stuyver et al., 2004; Youle and NATAP.org, 2004).
(Chen et al., 1999; Furman et al., 2001; Gulick, 2003; Jeffrey et al., 2003; Kewn et al., 2003; Otto, 2004; Thompson et al., 2005).
http://www.natap.org/x2004/pdf/posRep04.pdf; (Adams et al., 2004; Gu et al., 2006; Otto, 2004; Youle and NATAP.org, 2004).
N/A: not available.
Toxicity index is defined as the relative increase in the time required to replicate the mitochondrial genome based on the rates of incorporation and removal of chain terminators and is calculated using a formula. Please refer to text.
In vitro studies suggest that it is preferable to combine NRTI that employ different initial kinases, to avoid competitive inhibition of their respective phosphorylation enzymes (Bethell et al., 2003, 2004a,b, 2005; Havlir et al., 2000). For example, 3TC and FTC interfere with the phosphorylation of the 2′-deoxycytidine analog AVX754, resulting in a significant reduction in its active NTP (AVX754-TP), in primary human PBM cells in vitro and in PBM cells isolated from treated subjects (Bethell et al., 2004a,b, 2005). Similarly, the combination of the thymidine analogs ZDV and D4T, is contraindicated in the clinic, since both use cytosolic thymidine kinase (TK1) for their initial phosphorylation (Havlir et al., 2000). However, this dogma may not necessarily hold true for all nucleosides. Co-incubation of RVT with 3TC at clinically relevant concentrations, does not decrease the accumulation of 3TC-TP in T-cell lymphoma (CEM) or primary human PBM cells. This may be related to the similarities in Km values towards 2′-deoxycytidine kinase (dCK), since neither compound was able to displace the other efficiently. Further, intracellular levels of RVT-TPwereonlydecreasedat3TCconcentrationsgreaterthan those observed in the clinic in CEM cells (Hernandez-Santiago et al., 2004). Nevertheless, caution is warranted when using nucleoside analogs that share the same initial kinase. Possible strategies for overcoming competitive phosphorylation may include staggering the times of administration, thereby decreasing the total substrate load (sum of Cmax values in plasma) on the enzymes, or increasing the dose of the agent less able to compete (RVT in this case) (Hernandez-Santiago et al., 2004). However, such strategies remain to be tested in carefully designed clinical trials since the intracellular t1/2 of RVT and 3TC is high.
2. NRTI mitochondrial toxicity
HIV infection may be associated with reduction in mitochondrial DNA content and changes in mitochondrial morphology and function, which in some cases leads to clinical events such as myopathy or peripheral neuropathy (Moyle, 2005). Therefore, the toxicity of NRTI in vivo and in vitro are at least partially related to inhibition of mitochondrial DNA polymerase gamma, the sole enzyme responsible for the base excision repair of oxidative damage to mitochondrial DNA (Fromenty and Pessayre, 1997; Johnson et al., 2001; Kakuda, 2000; Lewis et al., 2006; Martin et al., 2003; Moyle, 2000; Zapor et al., 2004). This toxicity may be associated with a decrease in mitochondrial DNA and disrupted oxidative phosphorylation, evidenced by the toxic accumulation of non-esterified fatty acids, dicarboxylic acids and free radical damage (Kakuda, 2000). Published constants of inhibition of mitochondrial DNA polymerase gamma (Ki-DNA pol γ) derived using non-steady state kinetic measurement by various NRTI-TP are summarized in Table 1 (Feng et al., 2004a; Furman et al., 2001; Johnson et al., 2001; Murakami et al., 2004). Non-steady state values of Ki-DNA pol γ were quoted, since values determined using steady state methods may be unreliable due to the processive nature of DNA polymerases (Lee et al., 2003). Another more direct in vitro predictor of toxicity due to inhibition of Ki-DNA pol by NRTI is the invitro toxicity index (TI) (Johnson et al., 2001; Lee et al., 2003). TI is defined as the relative increase in the time required to replicating the mitochondrial genome based on the rates of incorporation and removal of chain terminators. TI is calculated using the formula:
where, D= ((kpol/Ki-DNA pol γ)dNTP)/(kpol/Ki-DNA pol γ)NRTI-TP.
In this equation, kcat represents the rate of incorporation of the natural dNTP; kexo represents the rate of excision of the chain terminator; [NRTI-TP]/[dNTP] is the ratio of the cellular concentrations of NRTI-TP and natural dNTP, respectively, where as D is the discrimination coefficient representing the ratio of specificities of the NRTI-TP and dNTP, respectively, and Kpol is the maximum rates of polymerization of the substrate into the mitochondrial DNA (Johnson et al., 2001; Lee et al., 2003). The values reported in Table 1 assume a [NRTI-TP]/[dNTP] ratio of 1, so that differences in mitochondrial uptake and phosphorylation are not reflected. A toxicity index of 1.0 defines zero toxicity, since the rate of replication of the genome is unaffected, while more toxic NRTI-TP have higher TI values (e.g., the TI for ddC triphosphate (ddC-TP) and d4T triphosphate (d4T-TP) are 160,000 and 3120, respectively). Although high values of TI are predictive of the in vivo toxicity for most NRTI, TI under-predicts the toxicity of ZDV (TI=1.05), suggesting that factors other than inhibition of DNA polymerase gamma may be responsible for ZDV toxicity (Johnson et al., 2001). ZDV and its reduced metabolite 3′-amino-3′-deoxythymidinedecreaselevels of globin mRNA in differentiating K-562 leukemia cells associated with decreased globin transcription in vitro (Weidner et al., 1992; Weidner and Sommadossi, 1990). This suggests that ZDV toxicity may also be related to disruption of the regulatory processes involved in the control of gene expression during development and differentiation of erythroid cells (Weidner et al., 1992; Weidner and Sommadossi, 1990), and may produce a decrease in the binding levels of major transcription factors involved in erythroid differentiation (Bridges et al., 1996). ZDV has also been shown to specifically inhibit β-globin gene in human erythroid progenitor cells at clinically relevant concentrations (Spiga et al., 1999). Our laboratory prefers determining mitochondrial toxicity using a cell-based assay over 14 days since in general nucleoside analogs (or NRTI-TP) require transport into the mitochondria and then phosphorylation to inhibit mtDNA synthesis (Bridges et al., 1999; Chen and Cheng, 1992; Stuyver et al., 2002).
3. NRTI for first line therapy
ZDV was the first antiretroviral drug tested in the clinic and was initially used as monotherapy. Clinical trials using combinations of two NRTI, e.g., ZDV with ddC, ddI or 3TC, demonstrated a more sustained antiviral and immunological response than ZDV alone (Delta Coordinating Committee, 2001; Hammer et al., 1996; Kuritzkes et al., 1996; Schooley et al., 1996; Staszewski et al., 1996). TK1 is the primarily enzyme responsible for ZDV phosphorylation to ZDV-monophosphate (ZDV-MP). However, mitochondrial thymidine kinase (TK2) has also been shown to activate ZDV in cultured monocytes/macrophages that do not express TK1, but to a much lesser degree (Arner et al., 1992; Eriksson et al., 1989, 1991; Furman et al., 1986). Since TK1 is cell cycle dependent, ZDV is phosphorylated to a much greater extent, and is more potent in activated than in resting cells. The TK1 activity in the bone marrow of rats followed a circadian pattern, and ZDV toxicity varied with time of administration (Zhang et al., 1993a,b). However, this has not been demonstrated in humans. Thymidylate kinase catalyzes the subsequent phosphorylation to ZDV-diphosphate (ZDV-DP) and is rate limiting with a Km of 12μM versus ZDV-MP (Furman et al., 1986; Lavie et al., 1997). ZDV treatment is limited by its toxic side effects in bone marrow cells which are manifested by anemia and neutropenia in about 30% of individuals and is partially dose dependent (Collier et al., 1990; Richman et al., 1987; Spiga et al., 1999). This toxicity may be partially related to inhibition of mitochondrial DNA polymerase gamma, the sole enzyme responsible for the base excision repair of oxidative damage to mitochondrial DNA. In addition, recent studies by McKee et al. demonstrated that ZDV is a potent inhibitor of thymidine phosphorylation in heart mitochondria (Johnson et al., 2001; Martin et al., 2003; McKee et al., 2004). Thus, the toxicity of AZT in some tissues may be mediated by disrupting the substrate supply of thymidine triphosphate (TTP) for mitochondrial DNA replication (McKee et al., 2004).
The current dose for ZDV for treating HIV infections is 300 mg bid. However, a study by Barry et al., demonstrated that when the ZDV dose was lowered from 300mg bid to 100 mg tid, the levels of active ZDV triphosphate (ZDV-TP) levels (related to antiviral efficacy), remained similar, while plasma concentrations of ZDV and intracellular ZDV-MP, associated with toxicity, varied linearly with dose (Barry et al., 1996). This result supports the possibility that the TK1 is super-saturated at ZDV doses used in the clinic (Km forTK1=12μM), and suggests that doses may be reduced without compromising efficacy (Furman et al., 1986). However, a recent study using ZDV at 600 mg once daily produced lower virus depletion than the 300 mg bid ZDV dose (Ruane et al., 2004), consistent with the short intracellular t1/2 (3–4h) of ZDV-TP in primary human PBM cells (Sharma et al., 2004), and the probable super-saturation of the TK1 kinase enzyme using this regimen. Super-saturation of TK1 would produce relatively lower ZDV-TP levels compared to the bid dose. The kinetics of phosphorylation for other NRTI discussed in this review are not saturated at clinically relevant doses. In contrast to the other NRTI mentioned, ZDV is eliminated primarily through the urinary tract in the inactive glucuronide (GAZT) form. GAZT is produced in the liver by glucuronidation by the enzyme human UDP-glucuronosyltransferase 2B7 (UGT2B7), and approximately 74% of the absorbed dose may be recovered in urine in that form (Barbier et al., 2000; Collins and Unadkat, 1989). Therefore, the clearance of ZDV may be reduced in the presence of other drugs that undergo significant glucuronidation (e.g., probenecid and sulfamethoxazole), resulting in increased plasma levels of ZDV (Morse et al., 1990). The first generic formulation for ZDV was approved in September 2005 by the US FDA. Pharmacokinetic and efficacy parameters for ZDV are summarized in Table 1 (Anderson et al., 2004; Barreiro et al., 2004; Cannard et al., 1998; Reardon and Miller, 1990; Wang et al., 1999). The population pharmacokinetics of ZDV have been reviewed elsewhere (Burger et al., 1994; Gitterman et al., 1990; Zhou et al., 1999).
The US FDA approved 3TC, marketed by GSK/Shire, for HIV-and HBV-infected persons. It is the first “l-nucleoside” and one of the most widely used nucleoside in HAART. 3TC is an important component of fixed nucleoside combinations including, Combivir® (containing 3TC and ZDV), Epzicom® (ABC and 3TC) and Trizivir® (ABC, 3TC and ZDV).Pertinent clinical pharmacokinetics of 3TC is summarized in Table 1 (Anderson et al., 2004; Barreiro et al., 2004; Johnson et al., 1999; Wang et al., 1999) and the population pharmacokinetics of 3TC have been described elsewhere (Moore et al., 1999a). 3TC has been approved for once- or twice-daily dose regimens (DeJesus et al., 2004; Sension et al., 2002). Unlike ZDV, the phosphorylation of 3TC to the 3TC triphosphate (3TC-TP) is not saturated at serum concentrations observed in the clinic and its phosphorylation is independent of position in the cell cycle. Furthermore, 3TC may produce higher ddNTP/dNTP ratios in resting cells, which may result in greater efficacy in non-stimulated lymphocytes (Gao et al., 1994). A major disadvantage of 3TC is the rapid development of resistance via a single point mutation (M184V), and resistance is likely to develop after only a few weeks of monotherapy (Eron et al., 1995). However, the M184V point mutation may be exploited in HAART combination therapy, since it may increase the susceptibility of certain resistant viruses to ZDV in some persons (Nijhuis et al., 1997), and may impair viral fitness (Miller et al., 2002). Therefore, 3TC is likely to continue to play an important role in many once-daily combinations. The anti-HBV activity of 3TC is also limited by a relatively rapid development of resistance which increases with the duration of treatment (Dienstag et al., 1999a,b; Dore et al., 1999; Stuyver et al., 2006).
FTC may exist as either the (+) of (−)-enantiomer. FTC in the (−)-enantiomer form is currently marketed by Gilead Sciences for treating HIV infections either alone (Emtricitabine®) or in combination with TDF in a fixed dose combination (Truvada®). Pharmacological parameters for FTC are summarized in Table 1 (Anderson et al., 2004; Bang and Scott, 2003; Barreiro et al., 2004; Molina et al., 2004). Mechanistic studies indicated that the triphosphate of (−)-FTC (FTC-TP) is a more potent inhibitor of HIV-1 RT than 3TC-TP (Feng et al., 1999; Ray et al., 2002, 2003). Testing of the resolved enantiomers has revealed (+)-FTC to be nontoxic and approximately 10- and 40-fold less potent inhibitor of HIV and HBV replication, respectively, in cell culture, than (−)-FTC (Doong et al., 1991; Furman et al., 1992; Schinazi et al., 1992). The Ki values of the (−)- and (+)-enantiomers of FTC-TP are 2.1 and 8.6μM, respectively, versus purified HIV-1 RT (Feng et al., 1999; Schinazi et al., 1992). (+)-FTC, but not the (−)-enantiomer may be deaminated by cytidine deaminase to the nontoxic inactive metabolite (+)-β−2′,3′-dideoxy-5-fluoro-3-thiauridine [(+)-FTU]. The extent of deamination to (+)-FTU varies between species according to enzyme levels and substrate specificity of this enzyme (Hurwitz et al., 2005). (−)-FTC was stable to deamination, regardless of the mammalian species tested, although some deamination has been observed by bacterial cytidine deaminase (Furman et al., 1992). The 50:50 racemic mixture of the (−)- and (+)-enantiomers of FTC (Racivir) is being developed by Pharmasset Inc., and is currently in Phase 2b clinical trials (Darque et al., 1999; Furman et al., 1992; Korba et al., 2000). Neither rats, rabbits, dogs nor rhesus monkeys was able to predict the pharmacokinetics of RCV reliably in humans. However, interspecies scaling was possible using a combination of animals (Cui et al., 1996; Frick et al., 1993, 1994; Hurwitz et al., 2005; Moore et al., 1997; Schinazi et al., 1992). A small phase Ib/IIa study was performed in which RCV was administered once daily orally to HIV-1-infected treatment-naïve male volunteers for 14 days at doses between 200 and 600 mg, in combination with stavudine and efavirenz. RCV was well tolerated, and the pharmacokinetics remained linear in that dose range (Herzmann et al., 2005). Viral loads dropped in all dosage groups, with mean maximal reductions of 2.1–2.6 log10 below baseline on day 28, and remained suppressed for more than 2 weeks following the last dose. Virus loads remained more than 1 log10 below baseline levels 21 days after the last dose (Herzmann et al., 2005). Studies performed in vitro suggest that RCV like (−)-FTC and 3TC selects for M184V, but at a slower rate. Furthermore, RCV inhibited the development of the T215Y resistance mutation that is associated with resistance to antiretroviral agents such as ZDV and D4T (Schinazi et al, 1997). Therefore, treatment regimens with RCV may offer some advantages over (−)-FTC in the clinical setting and could prove efficacious against viral mutants containing the M184V/I mutations. Results from a clinical study testing this hypothesis are currently underway.
TDF is an ester of the acyclic adenine nucleotide phosphonate, and was developed to improve the poor oral absorption of TFV(9-[2-(phosphonomethoxy)propyl]adenine), also known as PMPA (Lyseng-Williamson et al., 2005). Oral dosing of TDF results in approximately 25–30% bioavailability measured as circulating TFV concentrations. The pharmacokinetics of TFV is dose-proportional and similar in healthy volunteers and HIV-infected individuals. The oral bioavailability of tenofovir is enhanced by administration with a high-fat meal, but is similar at steady state when administered with or without a typical meal (Barditch-Crovo et al., 2001; Kearney et al., 2004). The population pharmacokinetics of TDF have been described elsewhere (Jullien et al., 2005b). Pharmacokinetic and efficacy parameters for TDF are summarized in Table 1 (Louie et al., 2003; Robbins et al., 1998; Suo and Johnson, 1998; White et al., 2002). Since TDF already contains a phosphate group it bypasses the need for an initial potentially rate limiting monophosphorylation and is readily metabolized to the active metabolite TFV-diphosphate (TFV-DP) in cells. Due to the longevity of the intracellular TFV-DP, TDF is administered once daily. TDF is also marketed as a once-daily co-formulation pill called Truvada® that contains 300 mg of TFV (equivalent to 245 mg of TFV) and 200 mg of FTC (Dando and Wagstaff, 2004).
ABC is a structural analog of 2′-deoxyguanosine. ABC has a more complex phosphorylation pathway than other NRTI. The initial phosphorylation step involves conversion to ABC monophosphate (ABC-MP) by adenosine phosphotransferase, followed by enzymatic deamination to carbovir monophosphate (CBV-MP), which then undergoes two additional enzymatic phosphorylation steps to form the active carbovir triphosphate (CBV-TP) by cellular kinases (Daluge et al., 1997; Faletto et al., 1997). Phosphorylation is not saturated at clinical doses of ABC. CBV-TP competes with 2′-deoxyguanosine triphosphate (dGTP) for viral RT and has a Ki of 21nM (Daluge et al., 1997; Faletto et al., 1997). Pertinent pharmacokinetic and efficacy parameters for ABC are summarized in Table 1 (Anderson et al., 2004; Hervey and Perry, 2000; Wang et al., 1999; White et al., 2005) and population pharmacokinetics are described elsewhere (Capparelli et al., 2005; Jullien et al., 2005a; Weller et al., 2000). Preliminary results reported from a pharmacokinetic study in 20 individuals taking 300 mg of ABC bid indicate that the cellular concentration of CBV-TP remained approximately two-fold higher than the Ki value for CBV-TP versus HIV-1 RT for 24 h post-dosing. Therefore, it was concluded that once-daily dosing of ABC is a viable option (Piliero, 2004). Preliminary results from other studies also suggest that ABC is effective at 300 mg bid and once-daily regimens when combined with 3TC (Goedken and Herman, 2005; Ruane and DeJesus, 2004).
4. Fixed dose co-formulation of NRTI
The development of fixed dose co-formulations of HAART drugs are desirable since they are convenient to take and simplify compliance and limit insurance co-payments. Co-formulations of NRTI include Truvada®, Combivir® and Epzicom®. Recent data indicate that Truvada in combination with Sustiva® (STV, Efavirenz) was superior to Combivir® with STV (Gallant et al., 2006). A triple drug co-formulation containing FTC, TDF and STV is also under development. However, studies of a fixed dose triple nucleoside combination regimen containing ABC, 3TC and ZDV (Trizivir®) demonstrated the onset of a more rapid virologic failure than regimens that include a PI or an NNRTI (Gulick et al., 2004).
5. NRTI developed for second line HAART
The emergence of resistant HIV strains during therapy has made it a major challenge to develop rgimens that delay, prevent or attenuate the onset of resistance. Since the dose of antiretroviral agents like 3TC are generally lower for HBV than HIV, it may be prudent to use anti-HBV agents that are not active against HIV in HBV positive individuals that are at risk for becoming infected with HIV, to avoid development of resistant HIV that may result from exposure of the virus to sub-therapeutic doses. Common resistance mutations, including TAMs, K65R and M184V, need to be a continued focus in HIV drug development. The emergence of mutations observed following exposure to the various NRTI discussed in this review are summarized in Table 1.
RVT, AVX-754 and AMDX are NRTI under development for use as second line therapies for individuals infected by viruses containing some of the common mutations. RVT, a 2′-deoxycytidine analog, is currently in Phase 2b clinical trials for the treatment of HIV infections by Pharmasset Inc. (Cohen et al., 2005). Preclinical studies indicate that RVT triphosphate (RVT-TP) had a long intracellular t1/2 in the range of 13–17h and inhibits replication of both wild type and mutant strains of HIV commonly observed during treatment with ZDV, 3TC and other NRTI (Schinazi et al., 2002). No mitochondrial toxicity or increase in the lactic acid was noted in human liver cells exposed for 14 days to the RVT. In 24 treatment-naïve patients, once-daily doses of 50, 100 and 200 mg as monotherapy induced a drop in viral load of approximately 1.77 logs after 10 days. Eighty-eight percent of subjects treated reached < 400 copies/ml. A recent Phase 2b clinical study demonstrated that RVT is a powerful drug against resistant viruses and it appears that no resistant viruses associated with RVT were selected (Erickson-Viitanen et al., 2006). Interestingly, RVT (200 mg dose, po) was highly effective in drug-experienced individuals that were not taking 3TC or FTC (mean reduction in viral load on week 16 was 1.4 and 1.5 log10) in optimized and non-optimized regimen, respectively, but was less effective in the presence of these oxathiolane cytosine analogs. Clinical parameters for RVT are summarized in Table 1 (Stuyver et al., 2004; Youle and NATAP.org, 2004). Recently, a phase 2b study was stopped due to elevated levels of lipase which could lead to pancreatitis in persons taking 200 mg RVT.
9-(Beta-dd-1,3-Dioxolan-4-yl)guanine (DXG) is a guanosine nucleoside with potent and selective activity against HIV-1, HIV-2 and HBV. The key pharmacological parameters of DXG are summarized in Table 1 (Gulick, 2003; Jeffrey et al., 2003; Kewn et al., 2003). DXG triphosphate (DXG-TP) is also a potent inhibitor of wild type and drug-resistant forms of HIV-1 RT including enzymes containing M184V/I, TAMs and the 69SS double insert, and of HBV in human cells (Chin et al., 2001; Lennerstrand et al., 2005; Seigneres et al., 2002; Ying et al., 2000). Resistance develops slowly in vitro, and is associated with mutations at K65R or L74V. However, DXG possesses limited aqueous solubility. AMDX (DAPD) is a 2,6-diaminopurine prodrug of DXG undergoing phase 2b clinical testing by RFS Pharma, LLC (Corbett and Rublein, 2001). AMDX is rapidly absorbed orally and is deaminated by adenosine deaminase (ADA) to DXG, before undergoing intracellular phosphorylation to DXG-TP (Chen et al., 1999; Cheng and Borroto-Esoda, 2004; Feng et al., 2004b). A study with 90 HIV-1 infected individuals receiving oral doses of AMDX between 25 and 500 mg bid and 600 mg qd (DAPD-101), demonstrated approximately linear pharmacokinetics with plasma DXG concentrations remaining higher than the EC50 value determined in vitro, for the entire dosing interval, for doses greater than 100 mg twice daily (Thompson et al., 2005). Peak plasma concentrations of AMDX and DXG were observed between 1 and 2h post-dosing and DXG plasma concentrations were much higher than those of AMDX (AUC ratios, 5–9). Mean terminal t1/2 values for AMDX and DXG were 1–2 and 4–7h, respectively. AMDX at 500 mg bid for 5 days produced a median reduction in plasma HIV-1 RNA of 1.3 log10 copies/ml in treatment naïve subjects, while HAART-experienced subjects had a median reduction in viral load was 0.7 log10 copies/ml (Thompson et al., 2005). HIV-1 isolated from subjects failing ZDV and 3TC therapies, remained sensitive to DXG in cell culture assays using isolated primary human PBM cells (Gu et al., 1999a,b; Mewshaw et al., 2002). A recent study indicated that AMDX was also effective in individuals that had failed in triple therapy that had a mean of six mutations in the pol region (Margolis et al., 2006).
AVX754 is another 2′-deoxycytidine analog with a similar structure to 3TC and FTC that is being developed by Avexa Ltd (Adams et al., 2004; Bethell et al., 2005; Taylor et al., 2000). AVX-754 has in vitro activity against a broad spectrum of thymidine associated mutations (TAMs) and is reported to be active against ZDV and 3TC-resistant virus in vitro, with a two-fold increase in the median effective concentration (EC50) when mutations at M184V and codon 215 were present. The susceptibility of HIV to the drug is also reduced two-fold by the presence of up to five TAMs (Bethell et al., 2003). Resistance is slower to develop in cell culture than 3TC, and like 3TC is associated with changes at codons K65R, V75I and M184V, making it potentially cross-resistant. Viruses containing these mutations may be up to four-fold less sensitive to AVX-754. A Phase 2a clinical trial in 63 HIV-infected individuals naïve to previous drug therapy demonstrated a statistically significant >1.4 log10 decrease in viral load. Furthermore, AVX754 was well tolerated and demonstrated good oral bioavailability and could be used in once-daily regimens (Gu et al., 2006; Otto, 2004).
6. Novel NRTI under pre-clinical development
APD is a very soluble prodrug of DXG in preclinical development. APD differs from DAPD since it requires oxidation by xanthine oxidase in the liver for activation. The oral pharmacokinetic of APD have been studied in mice and rhesus monkeys (Chen et al., 1996; Manouilov et al., 1997). APD demonstrated rapid oral absorption, and was rapidly converted to the active DXG metabolite. The terminal half-life of APD in plasma is two-fold to four-fold shorter than DXG (produced from APD) in both species (Chen et al., 1996; Manouilov et al., 1997). Furthermore, APD produced a high DXG AUC value in all treated animals. The bioavailability of APD was 41% in mice. High plasma concentrations of DXG were observed in monkeys despite low levels of the parent drug, suggesting a significant first-pass metabolism of APD. However, the oral bioavailability of APD was not calculated in monkeys, since no iv dose was administered. Further studies are warranted for this agent, which may be developed for HIV and HBV infections by RFS Pharma LLC.
DOT, a thymidine derivative is in the preclinical stage of development. This compound seems to have all the advantages of ZDV without the bone marrow toxicity and is also effective in the presence of important TAMs (Lennerstrand et al., 2006). Oral bioavailability in rats and monkeys is close to 100% (Schinazi et al., 2005). DOT has an EC50 of 0.43±0.07μM in primary human PBM cells and is significantly active against all of the commonly found NRTI-resistant HIV-1 mutants such as: D67N, K70R, T215Y, K219Q, K65R, and M184V (Chu et al., 2005).
7. Conclusions
Data contained in this review suggest that the field of nucleoside chemistry and biology has produced numerous highly effective agents for the treatment of HIV and HBV that have prolonged the lives of millions of infected persons. An “ideal” NRTI would demonstrate all or most of the following properties: have excellent oral bioavailability, and be a good substrate for cellular kinases for conversion to the active NRTI-TP while not competing for phosphorylation with other NRTI given concurrently. The NRTI-TP should have high affinity for the HIV RT enzyme (low Ki), and low affinity for human mitochondrial DNA polymerase, no mtDNA toxicity in cell systems on prolonged treatment, no human bone marrow toxicity, have a long intracellular half-life and an excellent pharmacological profile, including transport to various reservoirs where HIV is present. Furthermore, viral resistance to the NRTI should be rare and difficult to emerge. Although no “ideal” NRTI exists yet, data contained in this review suggest that the field of nucleoside chemistry and biology is expanding and that many of these goals have been reached. The continued fine-tuning of NRTI chemistry and biology should result in molecules that have the majority of the ideal characteristics described above, and these should provide additional treatment options for persons infected with HIV.
Acknowledgments
The preparation of this review was supported by the NIH grant 5R37-AI-41980, 2R37-AI-25899, Emory’s CFAR (5P30-AI-50409) and the Department of Veterans Affairs. RFS is entitled to future royalties derived from the sale of products related to the research described in this paper and is a founder of Pharmasset Inc and RFS Pharma LLC.
References
- Acosta E, Henry K, Baken L, Page L, Fletcher C, 1999. Indinavir concentrations and antiviral effect. Pharmacotherapy 19, 708–712. [DOI] [PubMed] [Google Scholar]
- Adams J, Sawyer J, Shiveley L, 2004. Intracellular SPD754 triphosphate pharmacokinetics following administration of SPD754 capsules. In: Proceedings of the 11th Conference on retroviruses and oppotunistic infections, Moscone West, San Francisco, CA. [Google Scholar]
- Anderson PL, Kakuda TN, Liechtenstein KA, 2004. The cellular pharmacology of nucleoside- and nucleotide-analogue reverse-transcriptase inhibitors and its relationship to clinical toxicities. Clin. Infect. Dis 38, 743–753. [DOI] [PubMed] [Google Scholar]
- Arner ES, Valentin A, Eriksson S, 1992. Thymidine and 3′-azido-3′-deoxythymidine metabolism in human peripheral blood lymphocytes and monocyte-derived macrophages. A study of both anabolic and catabolic pathways. J. Biol. Chem 267, 10968–10975. [PubMed] [Google Scholar]
- Back DJ, Burger DM, Flexner CW, Gerber JG, 2005. The pharmacology of antiretroviral nucleoside and nucleotide reverse transcriptase inhibitors: implications for once-daily dosing. J. Acquir. Immune Defic. Syndr 39 (1), S1–S23. [DOI] [PubMed] [Google Scholar]
- Bang LM, Scott LJ, 2003. Emtricitabine. An antiretroviral agent for HIV infection. Drugs 63, 2413–2424. [DOI] [PubMed] [Google Scholar]
- Barbier O, Turgeon D, Girard C, Green MD, Tephly TR, Hum DW, Belanger A, 2000. 3′-Azido-3′-deoxythymidine (AZT) is glucuronidated by human UDP-glucuronosyltransferase 2B7 (UGT2B7). Drug Metab. Dispos 28, 497–502. [PubMed] [Google Scholar]
- Barditch-Crovo P, Deeks SG, Collier A, Safrin S, Coakley DF, Miller M, Kearney BP, Coleman RL, Lamy PD, Kahn JO, McGowan I, Lietman PS, 2001. Phase I/II trial of the pharmacokinetics, safety, and antiretroviral activity of tenofovir disoproxil fumarate in human immunodeficiency virus-infected adults. Antimicrob. Agents Chemother 45, 2733–2739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barreiro P, Garcia-Benayas T, Rendon A, Rodriguez-Novoa S, Soriano V, 2004. Combinations of nucleoside/nucleotide analogues for HIV. AIDS Rev. 6, 234–243. [PubMed] [Google Scholar]
- Barry MG, Khoo SH, Veal GJ, Hoggard PG, Gibbons SE, Wilkins EG, Williams O, Breckenridge AM, Back DJ, 1996. The effect of zidovudine dose on the formation of intracellular phosphorylated metabolites. AIDS 10, 1361–1367. [DOI] [PubMed] [Google Scholar]
- Bethell R, Adams J, De Mys J, Lippens J, Richard A, Ren C, Collins P, Struthers-Semple C, Holdich T, Sawyer J, 2004a. Pharmacological evaluation of a dual deoxycytidine analogue combination: 3TC and SPD754. In: Proceedings of the 11th Conference on Retroviruses and Opportunistic Infections, San Francisco, CA, USA. [Google Scholar]
- Bethell R, Collins P, de Muys J, Lippens J, Hamelin B, Richard A, Sawyer J, 2004b. An in vitro evaluation of the intracellular anabolism of SPD754 and FTC alone and in combination. In: Proceedings of the 15th International AIDS Conference, Bangkok, Thailand. [Google Scholar]
- Bethell RC, Lie YS, Parkin NT, 2005. In vitro activity of SPD754, a new deoxycytidine nucleoside reverse transcriptase inhibitor (NRTI), against 215 HIV-1 isolates resistant to other NRTIs. Antivir. Chem. Chemother 16, 295–302. [DOI] [PubMed] [Google Scholar]
- Bethell RC, Parkin N, Lie Y, 2003. Antiviral activity of SPD-754 against clinical isolates of HIV-1 resistant to other nucleoside reverse transcriptase inhibitors In: Proceedings of the XII International HIV Drug Resistant workshop, Cabo del Sol, Los Cabos, Mexico. [Google Scholar]
- Bridges EG, Jiang Z, Cheng YC, 1999. Characterization of a dCTP transport activity reconstituted from human mitochondria. J. Biol. Chem 274, 4620–4625. [DOI] [PubMed] [Google Scholar]
- Bridges EG, Trentesaux C, Lahlil R, Spiga MG, Jeannesson P, Sommadossi JP, 1996. 3′-Azido-3′-deoxythymidine inhibits erythroid-specific transcription factors in human erythroid K562 leukemia cells. Eur. J. Haematol 56, 62–67. [DOI] [PubMed] [Google Scholar]
- Burger DM, Meenhorst PL, ten Napel CH, Mulder JW, Neef C, Koks CH, Bult A, Beijnen JH, 1994. Pharmacokinetics variability of zidovudine in HIV-infected individuals: subgroup analysis and drug interactions. AIDS 8, 1683–1689. [DOI] [PubMed] [Google Scholar]
- Cannard B, Sarfati SR, Richardson CC, 1998. Enhance binding of azidothymidine-resistant human immunodeficiency virus 1 reverse transcriptase to the 3′-azido-3′-deoxythymidine 5′-monophosphate-terminated primer. J. Biol. Chem 273, 14596–14604. [DOI] [PubMed] [Google Scholar]
- Capparelli EV, Letendre SL, Ellis RJ, Patel P, Holland D, McCutchan JA, 2005. Population pharmacokinetics of abacavir in plasma and cerebrospinal fluid. Antimicrob. Agents Chemother 49, 2504–2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carpenter CC, Cooper DA, Fischl MA, Gatell JM, Katzenteins, Montaner JS, Richman DD, Saag MS, Schechter M, Schooley RT, Thompson MA, Vella S, Yeni PG, Volberding PA, 2000. Antiretroviral therapy in adults: updated recommendations of the International Society-USA Panel. JAMA 19, 381–390. [DOI] [PubMed] [Google Scholar]
- Chen CH, Cheng YC, 1992. The role of cytoplasmic deoxycytidine kinase in the mitochondrial effects of the anti-human immunodeficiency virus compound, 2′,3′-dideoxycytidine. J. Biol. Chem 267, 2856–2859. [PubMed] [Google Scholar]
- Chen H, Boudinot FD, Chu CK, McClure HM, Schinazi RF, 1996. Pharmacokinetics of (−)-β-D-2-aminopurine dioxolane and (−)-β-D-2-amino-6-chloropurine dioxolane and their antiviral metabolite (−)-β-D-dioxolane guanine rhesus monkeys. Antimicrob. Agents Chemother 40, 2332–2336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen H, Schinazi RF, Rajagopalan P, Gao Z, Chu CK, McClure HM, Boudinot FD, 1999. Pharmacokinetics of (−)-beta-D-dioxolane guanine and prodrug (−)-beta-D-2,6-diaminopurine dioxolane in rats and monkeys. AIDS Res. Hum. Retroviruses 15, 1625–1630. [DOI] [PubMed] [Google Scholar]
- Cheng YC, Borroto-Esoda K, 2004. Anabolism of amdoxovir: phosphorylation of dioxolane guanosine and its 5-phosphates by mammalian phosphotransferases. Biochem. Pharmacol 68, 1879–1888. [DOI] [PubMed] [Google Scholar]
- Chin R, Shaw T, Torresi J, Sozzi V, Trautwein C, Bock T, Manns M, Isom H, Furman P, Locarnini S, 2001. In vitro susceptibilities of wild-type or drug-resistant hepatitis B virus to (−)-beta-D-2,6-diaminopurine dioxolane and 2′-fluoro-5-methyl-beta-L-arabinofuranosyluracil. Antimicrob. Agents Chemother 45, 2495–2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu CK, Yadav V, Chong YH, Schinazi RF, 2005. Anti-HIV activity of (−)-(2R,4R)-1-(2-hydroxymethyl-1,3-dioxolan-4-yl)thymine against drug resistant HIV-1 mutants and studies of its molecular mechanism. J. Med. Chem 48, 3949–3952. [DOI] [PubMed] [Google Scholar]
- Cohen C, Katlama C, Murphy R, Gathe J, Brinson C, Richmond G, Girad P-M, Fessel J, Liappis A, Puglia E, Rodwick B, Nadler J, O’Brien W, Arasteh K, Otto MJ, Erickson-Viitanen S, Levy R, 2005. Antiretroviral activity and tolerability of reverset (D-d4FC), a new fluorocytidine nucleoside analog when used in combination therapy in treatment-experienced patients: results of Phase IIb study RVT-203. In: Proceedings of the Third IAS Conference on HIV Pathogenesis and Treatment, Rio de Janeiro, Brazil. [Google Scholar]
- Collier AC, Bozzette S, Coombs RW, Causey DM, Schoenfeld DA, Spector SA, Pettinelli CB, Davies G, Richman DD, Leedom JM, Kidd P, Corey L, 1990. A pilot study of low-dose zidovudine in human immunodeficiency virus infection. N. Engl. J. Med 323, 1015–1021. [DOI] [PubMed] [Google Scholar]
- Collins JM, Unadkat JD, 1989. Clinical pharmacokinetics of zidovudine. An overview of current data. Clin. Pharmacokinet 17, 1–9. [DOI] [PubMed] [Google Scholar]
- Corbett AH, Rublein JC, 2001. DAPD (Emory University/Triangle Pharmaceuticals/Abbott Laboratories). Curr. Opin. Investig. Drugs 2, 348–353. [PubMed] [Google Scholar]
- Cui L, Schinazi RF, Gosselin G, Imbach J-L, Chu CK, Rando RF, Revankar GR, Sommadossi J-P, 1996. Effect of beta-enantiomeric and racemic nucleoside analogs on mitochondrial functions in HepG2 cells. Implications for predicting drug hepatotoxicity. Biochem. Pharmacol 52, 1577–1584. [DOI] [PubMed] [Google Scholar]
- Delta Coordinating Committee, 2001. Evidence for prolonged clinical benefit from initial combination antiretroviral therapy. HIV Med. 2, 181–188. [DOI] [PubMed] [Google Scholar]
- Daluge SM, Good SS, Faleto MB, Miller WH, Clair MH St., Boone LR, Tisdale M, Parry NR, Reardon JE, Dornsife RE, Averett DR, Krenitsky TA, 1997. 1592U89, a novel carbocyclic nucleoside analog with potent, selective anti-human immunodeficiency virus activity. Antimicrob. Agents Chemother 41, 1082–1093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dando TM, Wagstaff AJ, 2004. Emtricitabine/tenofovir disoproxil fumarate. Drugs 64, 2075–2082 (discussion 2090). [DOI] [PubMed] [Google Scholar]
- Darque A, Valette G, Rousseau F, Wang LH, Sommadossi J-P, Zhou X-J, 1999. Quantitation of intracellular triphosphate of emtricitabine in peripheral blood mononuclear cells from human immunodeficiency virus-infected patients. Antimicrob. Agents Chemother 43, 2245–2250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Deeks SG, Hoh R, Neilands TB, Liegler T, Aweeka F, Petropoulos CJ, Grant RM, Martin JN, 2005. Interruption of treatment with individual therapeutic drug classes in adults with multidrug-resistant HIV-1 infection. J. Infect. Dis 192, 1537–1544. [DOI] [PubMed] [Google Scholar]
- DeJesus E, McCarty D, Farthing CF, Shortino DD, Grinsztejn B, Thomas DA, Schrader SR, Castillo SA, Sension MG, Gough K, Madison SJ, 2004. Once-daily versus twice-daily lamivudine, in combination with zidovudine and efavirenz, for the treatment of antiretroviral-naive adults with HIV infection: a randomized equivalence trial. Clin. Infect. Dis 39, 411–418. [DOI] [PubMed] [Google Scholar]
- Dienstag JL, Schiff ER, Mitchell M, Casey DE Jr., Gitlin N, Lissoos T, Gelb LD, Condreay L, Crowther L, Rubin M, Brown N, 1999a. Extended lamivudine retreatment for chronic hepatitis B: maintenance of viral suppression after discontinuation of therapy. Hepatology 30, 1082–1087. [DOI] [PubMed] [Google Scholar]
- Dienstag JL, Schiff ER, Wright TL, Perrillo RP, Hann HW, Goodman Z, Crowther L, Condreay LD, Woessner M, Rubin M, Brown NA, 1999b. Lamivudine as initial treatment for chronic hepatitis B in the United States. N. Engl. J. Med 341, 1256–1263. [DOI] [PubMed] [Google Scholar]
- Doong SL, Tsai CH, Schinazi RF, Liotta DC, Cheng YC, 1991. Inhibition of the replication of hepatitis B virus in vitro by 2′,3′-dideoxy-3′-thiacytidine and related analogues. Proc. Natl. Acad. Sci. U.S.A 88, 8495–8499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dore GJ, Cooper DA, Barrett C, Goh LE, Thakrar B, Atkins M, 1999. Dual efficacy of lamivudine treatment in human immunodeficiency virus/hepatitis B virus-coinfected persons in a randomized, controlled study (CAESAR). The CAESAR Coordinating Committee. J. Infect. Dis 180, 607–613. [DOI] [PubMed] [Google Scholar]
- Erickson-Viitanen S, Hou K, Lloyd RJ, Mathis R, Burns D, Goyvaerts R, Levy R, 2006. Baseline genotype/phenotype, virological response, and lack of de novo resistance mutation generation during therapy with dexelvucitabine (formerly reverset) in study RVT-203. In: Proceedings of the 13th Conference on retroviruses and opportunistic infections, Denver, CO. [Google Scholar]
- Erickson-Viitanen S, Wu JT, Shi G, Unger S, King RW, Fish B, Klabe R, Gelenziunas R, Gallagher K, Otto MJ, Schinazi RF, 2003. Cellular pharmacology of D-d4FC, a nucleoside analog active against drug-resistant HIV. Antivir. Chem. Chemother 14, 39–47. [DOI] [PubMed] [Google Scholar]
- Eriksson BF, Chu CK, Schinazi RF, 1989. Phosphorylation of 3′-azido-2′,3′-dideoxyuridine and preferential inhibition of human and simian immunodeficiency virus reverse transcriptases by its 5′-triphosphate. Antimicrob. Agents Chemother 33, 1729–1734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eriksson S, Kierdaszuk B, Munch-Petersen B, Oberg B, Johansson NG, 1991. Comparison of the substrate specificities of human thymidine kinase 1 and 2 and deoxycytidine kinase toward antiviral and cytostatic nucleoside analogs. Biochem. Biophys. Res. Commun 176, 586–592. [DOI] [PubMed] [Google Scholar]
- Eron JJ, Benoit SL, Jemsek J, MacArthur RD, Santana J, Quinn JB, Kuritzkes DR, Fallon MA, Rubin M, 1995. Treatment with lamivudine, zidovudine, or both in HIV-positive patients with 200–500 CD4+ cells per cubic millimeter. North American HIV Working Party. N. Engl. J. Med 333, 1662–1669. [DOI] [PubMed] [Google Scholar]
- Faletto MB, Miller WH, Garvey EP, Clair MH St., Daluge SM, Good SS, 1997. Unique intracellular activation of the potent anti-human immunodeficiency virus agent 1592U89. Antimicrob. Agents Chemother 41, 1099–1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng JY, Murakami E, Zorca SM, Johnson AA, Johnson KA, Schinazi RF, Furman PA, Anderson KS, 2004a. Relationship between antiviral activity and host toxicity: comparison of the incorporation efficiencies of 2′,3′-dideoxy-5-fluoro-3′-thiacytidine-triphosphate analogs by human immunodeficiency virus type 1 reverse transcriptase and human mitochondrial DNA polymerase. Antimicrob. Agents Chemother 48, 1300–1306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng JY, Parker WB, Krajewski ML, Deville-Bonne D, Veron M, Krishnan P, Cheng YC, Borroto-Esoda K, 2004b. Anabolism of amdoxovir: phosphorylation of dioxolane guanosine and its 5-phosphates by mammalian phosphotransferases. Biochem. Pharmacol 68, 1879–1888. [DOI] [PubMed] [Google Scholar]
- Feng JY, Shi J, Schinazi RF, Anderson KS, 1999. Mechanistic studies show that (−)-FTC-TP is a better inhibitor of HIV-1 reverse transcriptase than 3TC-TP. FASEB J. 13, 1511–1517. [DOI] [PubMed] [Google Scholar]
- Fletcher CV, 1999. Pharmacologic considerations for therapeutic success with antiviral agents. Ann. Pharmacother 33, 989–995. [DOI] [PubMed] [Google Scholar]
- Fletcher CV, Brundage RC, Remmel RP, Page LM, Weller D, Calles NR, Simon C, Kline MW, 2000. Pharmacologic characteristics of indinavir, didanosine, and stavudine in human immunodeficiency virus-infected children receiving combination therapy. Antimicrob. Agents Chemother 44, 1029–1034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frick LW, Lambe CU, John L St., Taylor LC, Nelson DJ, 1994. Pharmacokinetic, oral bioavailability, and metabolism in mice and cynomolgus monkeys of (2′R,5′S)-cis-5-fluoro-1-[2-(hydroxymethyl)-1,3-oxathiolan-5-yl]cytosine, an agent active against human immunodeficiency virus and human hepatitis B virus. Antimicrob. Agents Chemother 38, 2722–2729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frick LW, John L St., Taylor LC, Painter GR, Furman PA, Liotta DC, Furfine ES, Nelson DJ, 1993. Pharmacokinetics, oral bioavailability, and metabolic disposition in rats of (−)-cis-5-fluoro-1-[2(hydroxymethyl)-1,3-oxathiolan-5-yl]cytosine, a nucleoside analog active against human immunodeficiency virus and hepatitis B virus. Antimicrob. Agents Chemother 37, 2285–2292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fromenty B, Pessayre D, 1997. Impaired mitochondrial function in microvesicular steatosis. Effects of drugs, ethanol, hormones and cytokines. J. Hepatol 26 (Suppl. 2), 43–53. [DOI] [PubMed] [Google Scholar]
- Furman PA, Davis M, Liotta DC, Paff MT, Frick LW, Nelson DJ, Dornsife RE, Wurster JA, Wilson LJ, Fyfe JA, Tuttle JV, Miller WH, Condreay L, Averett DR, Schinazi RF, Painter GR, 1992. The anti-hepatitis B activities, cytotoxicities, and anabolic profiles of the (−) and (+) enantiomers of cis-5-fluoro-1-[2-(hydroxymethyl)-1,3-oxathiolan-5-yl]cytosine (FTC). Antimicrob. Agents Chemother 36, 2686–2692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Furman PA, Fyfe JA, St Clair MH, Weinhold K, Rideout JL, Freeman GA, Lehrman SN, Bolognesi DP, Broder S, Mitsuya H, Barry DW, 1986. Phosphorylation of 3-azido-3-deoxythymidine and selective interaction of the 5-triphosphate with human immunodeficiency virus reverse transcriptase. Proc. Natl. Acad. Sci. U.S.A 83, 8333–8337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Furman PA, Jeffrey JL, Kiefer LL, Feng JY, Anderson KS, Borroto-Esoda K, Hill E, Copeland WC, Chu CK, Sommadossi J-P, Liberman I, Schinazi RF, Painter GR, 2001. Mechanism of action of 1-β-d-2,6-diaminopurine dioxolane, a prodrug of the human immunodeficiency virus type 1 inhibitor 1--d-dioxolane guanosine. Antimicrob. Agents Chemother 45, 158–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gallant JE, DeJesus E, Arribas JR, Pozniak AL, Gazzard B, Campo RE, Lu B, McColl D, Chuck S, Enejosa J, Toole JJ, Cheng AK, 2006. Tenofovir DF, emtricitabine, and efavirenz versus zidovudine, lamivudine, and efavirenz for HIV. N. Engl. J. Med 354, 251–260. [DOI] [PubMed] [Google Scholar]
- Gao W-Y, Agbaria R, Driscoll JS, Mitsuya H, 1994. Divergent anti-human immunodeficiency virus activity and anabolic phosphorylation of 2′,3′-dideoxynucleoside analogs in resting and activated human cells. J. Biol. Chem 269, 12633–12638. [PubMed] [Google Scholar]
- Gitterman SR, Drusano GL, Egorin MJ, Standiford HC, 1990. Population pharmacokinetics of zidovudine. Clin. Pharmacol. Ther 48, 161–167. [DOI] [PubMed] [Google Scholar]
- Goedken AM, Herman RA, 2005. Once-daily abacavir in place of twice daily administration. Ann. Pharmacother 39, 1302–1308. [DOI] [PubMed] [Google Scholar]
- Gu Z, Allard B, de Muys JM, Lippens J, Rando RF, Nguyen-Ba N, Ren C, McKenna P, Taylor DL, Bethell RC, 2006. In vitro antiretroviral activity and in vitro toxicity profile of SPD754, a new deoxycytidine nucleoside reverse transcriptase inhibitor for treatment of human immunodeficiency virus infection. Antimicrob. Agents Chemother 50, 625–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gu Z, Wainberg MA, Nguyen-Ba N, L’Heureux L, de Muys JM, Bowlin TL, Rando RF, 1999a. Mechanism of action and in vitro activity of 1′,3′-dioxolanylpurine nucleoside analogues against sensitive and drug-resistant human immunodeficiency virus type 1 variants. Antimicrob. Agents Chemother 43, 2376–2382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gu Z, Wainberg MA, Nguyen-Ba P, L’Heureux L, de Muys JM, Rando RF, 1999b. Anti-HIV-1 activities of 1,3-dioxolane guanine and 2,6-diaminopurine dioxolane. Nucleosides Nucleotides 18, 891–892. [DOI] [PubMed] [Google Scholar]
- Gulick RM, 2003. New antiretroviral drugs. Clin. Microbiol. Infect 9, 186–193. [DOI] [PubMed] [Google Scholar]
- Gulick RM, Ribaudo HJ, Shikuma CM, Lustgarten S, Squires KE, Meyer WA, Acosta EP, 2004. Triple-nucleoside regimens versus efavirenz-containing regimens for the initial treatment of HIV-1 infection. N. Engl. J. Med 350, 1850–1861. [DOI] [PubMed] [Google Scholar]
- Hammer SM, Katzenstein DA, Hughes MD, Gundacker H, Schooley RT, Haubrich RH, Henry WK, Lederman MM, Phair JP, Niu M, Hirsch MS, Merigan TC, 1996. A trial comparing nucleoside monotherapy with combination therapy in HIV-infected adults with CD4 cells counts from 200 to 500 per cubic millimeter. N. Engl. J. Med 335, 1081–1090. [DOI] [PubMed] [Google Scholar]
- Havlir DV, Tierney C, Friedland GH, Pollard RB, Smeaton L, Sommadossi J-P, Fox L, Kesseler H, Fife KH, Richman DD, 2000. In vivo antagonism with zidovudine plus stavudine combination therapy. J. Infect. Dis 182, 321–325. [DOI] [PubMed] [Google Scholar]
- Hawkins T, Veikley W, St Claire RL, Guyer B, Clark N, Kearney BP, 2005. Intracellular pharmacokinetics of tenofovir diphosphate, carbovir triphosphate, and lamivudine triphosphate in patients receiving triple-nucleoside regimens. J. Acquir. Immune Defic. Syndr 39, 406–411. [DOI] [PubMed] [Google Scholar]
- Hernandez-Santiago B, Rapp KL, Mathew J, Schinazi RF, 2004. Lack of antagonistic interactions for the two cytidine analogs Reverset™ and Lamivudine in vitro In: HIV DART 2004: Frontiers in Drug Development for Antiretroviral Therapies, Montego Bay, Jamaica. [Google Scholar]
- Hervey PS, Perry CM, 2000. Abacavir. A review of its clinical potential in patients with HIV infection. Drugs 60, 447–479. [DOI] [PubMed] [Google Scholar]
- Herzmann C, Arasteh K, Murphy RL, Schulbin H, Kreckel P, Drauz D, Schinazi RF, Beard A, Cartee L, Otto MJ, 2005. Safety, pharmacokinetics, and efficacy of (±)-β−2′,3′-dideoxy-5-fluoro-3′-thiacytidine with efavirenz and stavudine in antiretroviral-naive human immunodeficiency virus-infected patients. Antimicrob. Agents Chemother 49, 2828–2833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hurwitz SJ, Otto MJ, Schinazi RF, 2005. Comparative pharmacokinetics of racivir, (±)-beta-2′,3′-dideoxy-5-fluoro-3′-thiacytidine in rats, rabbits, dogs, monkeys and HIV-infected humans. Antivir. Chem. Chemother 16, 117–127. [DOI] [PubMed] [Google Scholar]
- Jeffrey JL, Feng JY, Qi CC, Anderson KS, Furman PA, 2003. Dioxolane guanosine 5′-triphosphate, an alternative substrate inhibitor of wild-type and mutant HIV-1 reverse transcriptase. Steady state and pre-steady state kinetic analyses. J. Biol. Chem 278, 18971–18979. [DOI] [PubMed] [Google Scholar]
- Johnson AA, Ray AS, Hanes J, Suo Z, Colacino JM, Anderson KS, Johnson KA, 2001. Toxicity of antiviral nucleoside analogs and the human mitochondrial DNA polymerase. J. Biol. Chem 276, 40847–40857. [DOI] [PubMed] [Google Scholar]
- Johnson MA, Moore KHP, Yuen GJ, Bye A, Pakes GE, 1999. Clinical pharmacokinetics of lamivudine. Clin. Pharmacokinet 36, 41–66. [DOI] [PubMed] [Google Scholar]
- Jullien V, Treluyer JM, Chappuy H, Dimet J, Rey E, Dupin N, Salmon D, Pons G, Urien S, 2005a. Weight related differences in the pharmacokinetics of abacavir in HIV-infected patients. Br. J. Clin. Pharmacol 59, 183–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jullien V, Treluyer JM, Rey E, Jaffray P, Krivine A, Moachon L, Lillo-Le Louet A, Lescoat A, Dupin N, Salmon D, Pons G, Urien S, 2005b. Population pharmacokinetics of tenofovir in human immunodeficiency virus-infected patients taking highly active antiretroviral therapy. Antimicrob. Agents Chemother 49, 3361–3366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kakuda TN, 2000. Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity. Clin. Ther 22, 685–708. [DOI] [PubMed] [Google Scholar]
- Kearney BP, Flaherty JF, Shah J, 2004. Tenofovir disoproxil fumarate: clinical pharmacology and pharmacokinetics. Clin. Pharmacokinet 43, 595–612. [DOI] [PubMed] [Google Scholar]
- Kewn S, Wang LH, Hoggard PG, Rousseau F, Hart R, MacNeela JP, Khoo SH, Back DJ, 2003. Enzymatic assay for measurement of intracellular DXG triphosphate concentrations in peripheral blood mononuclear cells from human immunodeficiency virus type 1-infected patients. Antimicrob. Agents Chemother 47, 255–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Korba BE, Schinazi RF, Cote P, Tennant BC, Gerin JL, 2000. Effect of oral administration of emtricitabine on woodchuck hepatitis virus replication in chronically infected woodchucks. Antimicrob. Agents Chemother 44, 1757–1760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuritzkes DR, Quinn JB, Benoit SL, Shugarts DL, Griffin A, Bakhtiari M, Poticha D, Eron JJ, Fallon MA, Rubin M, 1996. Drug resistance and virologic response in NUCA 3001, a randomized trial of lamivudine (3TC) versus zidovudine (ZDV) versus ZDV plus 3TC in previously untreated patients. AIDS 10, 975–981. [DOI] [PubMed] [Google Scholar]
- Lavie A, Schlichting I, Vetter IR, Konrad M, Reinstein J, Goody RS, 1997. The bottleneck in AZT activation. Nat. Med 3, 922–924. [DOI] [PubMed] [Google Scholar]
- Lee H, Hanes J, Johnson KA, 2003. Toxicity of nucleoside analogues used to treat AIDS and the selectivity of the mitochondrial DNA polymerase. Biochemistry 42, 14711–14719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lennerstrand J, Bluemling GR, Ruckstuhl M, Bennett M, Chu CK, Schinazi RF, 2006. 1-(β-D-Dioxolane)thymine (DOT) is effective against HIV-1 containing TAMs and M184V. In: Proceedings of the 13th Conference on Retroviruses and Opportunistic Infections, Denver, CO. [Google Scholar]
- Lennerstrand J, Hassani-Espili N, Pavlova A, Chu CK, Asif G, Schinazi RF, 2005. Mechanism of HIV-1 reverse transcriptase resistance to β-D-dioxolane-guanine triphosphate. In: Conference on Antimicrobial Agents and Chemotherapy, Washington DC, USA. [Google Scholar]
- Lewis W, Kohler JJ, Hosseini SH, Haase CP, Copeland WC, Bienstock RJ, Ludaway T, McNaught J, Russ R, Stuart T, Santoianni R, 2006. Antiretroviral nucleosides, deoxynucleotide carrier and mitochondrial DNA: evidence supporting the DNA pol gamma hypothesis. AIDS 20, 675–684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lipsky JJ, 1996. Antiretroviral drugs for AIDS. Lancet 348, 800–803. [DOI] [PubMed] [Google Scholar]
- Lorenzi P, Yerly S, Abderrakim K, Fathi M, Rutschmann OT, von Overbeck J, Leduc D, Perrin L, Hirschel B, 1997. Toxicity, efficacy, plasma drug concentrations and protease mutations in patients with advanced HIV infection treated with ritonavir plus saquinavir. Swiss HIV Cohort Study. AIDS 11, F95–F99. [DOI] [PubMed] [Google Scholar]
- Louie M, Hogan C, Hurley A, Simon V, Chung C, Padte N, Lamy P, Flaherty J, Coakley D, Di Mascio M, Perelson AS, Markowitz M, 2003. Determining the antiviral activity of tenofovir disoproxil fumarate in treatment-naive chronically HIV-1-infected individuals. AIDS 17, 1151–1156. [DOI] [PubMed] [Google Scholar]
- Lyseng-Williamson KA, Reynolds NA, Plosker GL, 2005. Tenofovir disoproxil fumarate: a review of its use in the management of HIV infection. Drugs 65, 413–432. [DOI] [PubMed] [Google Scholar]
- Manouilov KK, Manouilova LS, Boudinot DF, Schinazi RF, Chu CK, 1997. Biotransformation and pharmacokinetics of prodrug 9-(β-D-1,3-dioxolan-4-yl)guanine in mice. Antivir. Res 35, 187–193. [DOI] [PubMed] [Google Scholar]
- Margolis D, Mukherjee L, Hogg E, Fletcher C, Ogata-Arakaki D, Petersen T, Rusin D, Martinez A, Adams E, Mellors J, team A, 2006. A phase I/II randomised, double-blind, placebo-controlled pilot study of β-d-2 6-diaminopurine dioxolane versus DAPD+mycophenolate mofetil in treatment-experienced subjects (ACTG 5165). In: Proceedings of the 13th Conference on retroviruses and opportunistic infections, Denver, CO. [Google Scholar]
- Martin AM, Hammond E, Nolan D, Pace C, Den Boer M, Taylor L, Moore H, Martinez OP, Christiansen FT, Mallal S, 2003. Accumulation of mitochondrial DNA mutations in human immunodeficiency virus-infected patients treated with nucleoside-analogue reverse-transcriptase inhibitors. Am. J. Hum. Genet 72, 549–560 (Erratum in: Am. J. Hum. Genet. 2003, 2072, 1358). [DOI] [PMC free article] [PubMed] [Google Scholar]
- McKee EE, Bentley AT, Hatch M, Gingerich J, Susan-Resiga D, 2004. Phosphorylation of thymidine and AZT in heart mitochondria: elucidation of a novel mechanism of AZT cardiotoxicity. Cardiovasc. Toxicol 4, 155–167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mewshaw JP, Myrick FT, Wakefield DA, Hooper BJ, Harris JL, McCreedy B, Borroto-Esoda K, 2002. Dioxolane guanosine, the active form of the prodrug diaminopurine dioxolane, is a potent inhibitor of drug-resistant HIV-1 isolates from patients for whom standard nucleoside therapy fails. J. Acquir. Immune Defic. Syndr 29, 11–20. [DOI] [PubMed] [Google Scholar]
- Miller V, Stark T, Loeliger AE, Lange JM, 2002. The impact of the M184V substitution in HIV-1 reverse transcriptase on treatment response. HIV Med. 3, 135–145. [DOI] [PubMed] [Google Scholar]
- Molina JM, Peytavin G, Perusat S, Lascoux-Combes C, Serein D, Rozebaum W, Chene G, 2004. Pharmacokinetics of emtricitabine, didanosine and efavirenz administrated once-daily for the treatment of HIV-infected adults (pharmacokinetic substudy of the ANRS 091 trial). HIV Med. 5, 99–104. [DOI] [PubMed] [Google Scholar]
- Moore KH, Yuen GJ, Hussey EK, Pakes GE, Eron JJJ, Bartlett JA, 1999a. Population pharmacokinetics of lamivudine in adult human immunodeficiency virus-infected patients enrolled in two phase III clinical trials. Antimicrob. Agents Chemother 43, 3025–3029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore KHP, Barrett JE, Shaw S, Pakes GE, Churchus R, Kapoor A, Lloyd J, Barry MG, Back D, 1999b. The pharmacokinetics of lamivudine phosphorylation in peripheral blood mononuclear cells from patients infected with HIV-1. AIDS 13, 2239–2250. [DOI] [PubMed] [Google Scholar]
- Moore LF, Ascenzi MA, Tennant BC, Shi J, Schinazi RF, Boudinot FD, 1997. Stereoselective Pharmacokinetics and Metabolism of 2′-Deoxy-Fluoro-3′-Thiacytidine (FTC) in Woodchucks. American Association Scientist (AAPS), Boston, Mass, USA. [Google Scholar]
- Morse GD, Lechner JL, Santora JA, Rozek SL, 1990. Zidovudine update: 1990. DICP 24, 754–760. [DOI] [PubMed] [Google Scholar]
- Moyle G, 2000. Clinical manifestations and management of antiretroviral nucleoside analog-related mitochondrial toxicity. Clin. Ther 22, 911–936 (discussion 898). [DOI] [PubMed] [Google Scholar]
- Moyle G, 2005. Mechanisms of HIV and nucleoside reverse transcriptase inhibitor injury to mitochondria. Antivir. Ther 10 (Suppl. 2), M47–M52. [PubMed] [Google Scholar]
- Murakami E, Ray AS, Schinazi RF, Anderson KS, 2004. Investigating the effects of stereochemistry on incorporation and removal of 5-fluorocytidine analogs by mitochondrial DNA polymerase gamma: comparison of d- and l-D4FC-TP. Antivir. Res 62, 57–64. [DOI] [PubMed] [Google Scholar]
- Nijhuis M, Schuurman R, de Jong D, van Leeuwen R, Lange J, Danner S, Keulen W, de Groot T, Boucher CA, 1997. Lamivudine-resistant human immunodeficiency virus type 1 variants (184V) require multiple amino acid changes to become co-resistant to zidovudine in vivo. J. Infect. Dis 176, 398–405. [DOI] [PubMed] [Google Scholar]
- Otto MJ, 2004. New nucleoside reverse transcriptase inhibitors for the treatment of HIV infections. Curr. Opin. Pharmacol 4, 431–436. [DOI] [PubMed] [Google Scholar]
- Pereira CF, Paridaen JT, 2004. Anti-HIV drug development—an overview. Curr. Pharm. Des 10, 4005–4037. [DOI] [PubMed] [Google Scholar]
- Piliero PJ, 2004. Pharmacokinetic properties of nucleoside/nucleotide reverse transcriptase inhibitors. J. Acquir. Immune Defic. Syndr 37 (Suppl. 1), S2–S12. [DOI] [PubMed] [Google Scholar]
- Ray AS, Murakami E, Peterson CN, Shi J, Schinazi RF, Anderson KS, 2002. Interactions of enantiomers of 2′,3′-didehydro-2′,3′-dideoxy-fluorocytidine with wild type and M184V mutant HIV-1 reverse transcriptase. Antivir. Res 56, 189–205. [DOI] [PubMed] [Google Scholar]
- Ray AS, Schinazi RF, Murakami E, Bassavapathruni A, Shi J, Zorca SM, Chu CK, Anderson KS, 2003. Probing the mechanistic consequences of 5-fluorine substitution on cytidine nucleotide analogue incorporation by HIV-1 reverse transcriptase. Antivir. Chem. Chemother 14, 115–125. [DOI] [PubMed] [Google Scholar]
- Reardon JE, Miller WH, 1990. Human immunodeficiency virus reverse transcriptase. Substrate and inhibitor kinetics with thymidine 5′-triphosphate and 3′-azido-3′-deoxythymidine 5′-triphosphate. J. Biol. Chem 265, 20302–20307. [PubMed] [Google Scholar]
- Richman DD, Fischl MA, Grieco MH, Gottlieb MS, Volberding PA, Laskin OL, Leedom JM, Groopman JE, Mildvan D, Hirsch MS, et al. , 1987. The toxicity of azidothymidine (AZT) in the treatment of patients with AIDS and AIDS-related complex. A double-blind, placebo-controlled trial. N. Engl. J. Med 317, 192–197. [DOI] [PubMed] [Google Scholar]
- Robbins BL, Srinivas RV, Kim C, Bischofberger N, Fridland A, 1998. Anti-human immunodeficiency virus activity and cellular metabolism of a potential prodrug of the acyclic nucleoside phosphonate 9-R-(2-phosphonomethoxypropyl)adenine (PMPA), bis(isopropyloxy-methylcarbonyl)PMPA. Antimicrob. Agents Chemother 42, 612–617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruane PJ, DeJesus E, 2004. New nucleoside/nucleotide backbone options: a review of recent studies. J. Acquir. Immune Defic. Syndr 37, S21–S29. [DOI] [PubMed] [Google Scholar]
- Ruane PJ, Richmond GJ, DeJesus E, Hill-Zabala CE, Danehower SC, Liao Q, Johnson J, Shaefer MS, 2004. Pharmacodynamic effects of zidovudine 600 mg once/day versus 300 mg twice/day in therapy-naive patients infected with human immunodeficiency virus. Pharmacotherapy 24, 307–312. [DOI] [PubMed] [Google Scholar]
- Schinazi RF, 1991. Combined therapeutic modalities for viral infections—rationale and clinical potential In: Chou TC, Rideout DC (Eds.), Synergism and antagonism in Chemotherapy. Academic Press, Orlando, FL, pp. 110–181. [Google Scholar]
- Schinazi RF, Asif G, Detorio M, Hernandez-Santiago B, Ruckstuhl M, Yadav V, Rapp KL, Bennett M, Grier J, Xie M-Y, Lennerstrand J, Kozlowski M, Lutz S, Hurwitz SJ, Chu CK, 2005. 1-(β-D-dioxolane)thymidine (DOT) is a TK-dependent orally bioavailable nucleoside with specific activity against HIV-1 resistant variants. Antivir. Ther 10, S1–S65. [Google Scholar]
- Schinazi RF, Boudinot FD, Ibrahim SS, Manning C, McClure HM, Liotta DC, 1992. Pharmacokinetics and metabolism of racemic 2′,3′-dideoxy-5-fluoro-3′-thiacytidine in rhesus monkeys. Antimicrob. Agents Chemother 36, 2432–2438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schinazi RF, McMillan A, Lloyd JRL, Schlueter-Wirtz S, Liotta DC, Chu CK, 1997. Molecular properties of HIV-1 resistant to (+)-enantiomers and racemates of oxathiolane cytosine nucleoside and their potential for the treatment of HIV and HBV infections. Antivir. Res 34, A42. [Google Scholar]
- Schinazi RF, Mellors J, Bazmi H, Diamond S, Garber S, Gallagher K, Gelenziunas R, Klabe R, Pierce M, Rayner M, Wu J-T, Zhang H, Hammond J, Bacheler L, Manion DJ, Otto MJ, Stuyver LJ, Trainor G, Liotta DC, Erickson-Viitanen S, 2002. DPC 817: a cytidine nucleoside analog with activity against zidovudine- and lamivudine-resistant viral variants. Antimicrob. Agents Chemother 46, 1394–1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schooley RT, Ramirez-Ronda C, Lange JM, Cooper DA, Lavelle J, Lefkowitz L, Moore M, Larder BA, St Claire RL, Mulder JW, McKinnis R, Pennington KN, Harrigan PR, Kinghorn I, Steel H, Rooney JF, 1996. Virologic and immunologic benefits of initial combination therapy with zidovudine and zalcitabine or didanosine compared with zidovudine monotherapy. J. Infect. Dis 173, 1354–1366. [DOI] [PubMed] [Google Scholar]
- Seigneres B, Pichoud C, Martin P, Furman P, Trepo C, Zoulim F, 2002. Inhibitory activity of dioxolane purine analogs on wild-type and lamivudine-resistant mutants of hepadnaviruses. Hepatology 36, 710–722. [DOI] [PubMed] [Google Scholar]
- Sension MG, Bellos NC, Johnson J, Sepulveda GE, DeJesus E, Santana JL, Ames MH, Goodwin SD, 2002. Lamivudine 300 mg QD versus continued lamivudine 150 mg BID with stavudine and a protease inhibitor in suppressed patients. HIV Clin. Trials 3, 361–370. [DOI] [PubMed] [Google Scholar]
- Sharma PL, Nurpeisov V, Hernandez-Santiago B, Beltran T, Schinazi RF, 2004. Nucleoside inhibitors of human immunodeficiency virus type 1 reverse transcriptase. Curr. Top. Med. Chem 4, 895–921. [DOI] [PubMed] [Google Scholar]
- Siliciano JD, Siliciano RF, 2004. A long-term latent reservoir for HIV-1: discovery and clinical implications. J. Antimicrob. Chemother 54, 6–9. [DOI] [PubMed] [Google Scholar]
- Siliciano RF, 2005. Scientific rationale for antiretroviral therapy in 2005: viral reservoirs and resistance evolution. Top. HIV Med 13, 96–100. [PubMed] [Google Scholar]
- Solas C, Li YF, Xie M-Y, Sommadossi J-P, Zhou X-J, 1998. Intracellular nucleotides of (−)-2′,3′-deoxy-3′-thiacytidine in peripheral blood mononuclear cells of a patient infected with human immunodeficiency virus. Antimicrob. Agents Chemother 42, 2989–2995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sommadossi J-P, 1982. Cellular nucleoside pharmacokinetics and pharmacology: a potentially important determinant of antiretroviral efficacy. AIDS 3, S1–S8. [PubMed] [Google Scholar]
- Spiga MG, Weidner DA, Trentesaux C, LeBoeuf RD, Sommadossi JP, 1999. Inhibition of beta-globin gene expression by 3′-azido-3′-deoxythymidine in human erythroid progenitor cells. Antivir. Res 44, 167–177. [DOI] [PubMed] [Google Scholar]
- Staszewski S, Loveday C, Picazo JJ, Dellarnonica P, Skinhoj P, Johnson MA, Danner SA, Harrigan PR, Hill AM, Verity L, McDale H, 1996. Safety and efficacy of lamivudine–zidovudine combination therapy in zidovudine-experienced patients. A randomized controlled comparison with zidovudine monotherapy. JAMA 276, 111–117. [PubMed] [Google Scholar]
- Stein DS, Fish DG, Bilello JA, Preston SL, Martineau GL, Drusano GL, 1996. A 24-week open-label phase I/II evaluation of the HIV protease inhibitor MK-639 (indinavir). AIDS 10, 485–492. [DOI] [PubMed] [Google Scholar]
- Stuyver L, Fried MW, Van Geyt C, Rossau R, Sablon E, Schinazi RF, 2006. Dynamics of emergence and disappearance of lamivudine-associated mutations in hepatitis B virus In: Schinazi RF, Schiff ER (Eds.), Framing the knowledge of therapeutics for viral hepatitis. IHL Press, College Park, GA, pp. 505–515. [Google Scholar]
- Stuyver LJ, Lostia S, Adams M, Mathew JS, Pai BS, Grier J, Tharnish PM, Choi Y, Chong Y, Choo H, Chu CK, Otto MJ, Schinazi RF, 2002. Antiviral activities and cellular toxicities of modified 2′,3′-dideoxy-2′,3′-didehydrocytidine analogues. Antimicrob. Agents Chemother 46, 3854–3860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stuyver LJ, McBrayer TR, Schurmann D, Kravec I, Beard A, Cartee L, Schinazi RF, de la Rosa A, Murphy RL, Otto MJ, 2004. Potent antiviral effect of Reverset™ in HIV-1-infected adults following a single oral dose. Antivir. Ther 9, 529–536. [PubMed] [Google Scholar]
- Suo Z, Johnson KA, 1998. Selective inhibition of HIV-1 reverse transcriptase by an antiviral inhibitor, (R)-9-(2-phosphonylmethoxypropyl)adenine. J. Biol. Chem 273, 27250–27258. [DOI] [PubMed] [Google Scholar]
- Taylor DL, Ahmed PS, Tyms AS, Wood LJ, Kelly LA, Chambers P, Clarke J, Bedard J, Bowlin TL, Rando RF, 2000. Drug resistance and drug combination features of the human immunodeficiency virus inhibitor, BCH-10652 [(±)-2′-deoxy-3′-oxa-4′-thiocytidine, dOTC]. Antivir. Chem. Chemother 11, 291–301. [DOI] [PubMed] [Google Scholar]
- Thompson MA, Kessler HA, Eron JJ Jr., Jacobson JM, Adda N, Shen G, Zong J, Harris J, Moxham C, Rousseau FS, 2005. Short-term safety and pharmacodynamics of amdoxovir in HIV-infected patients. AIDS 19, 1607–1615. [DOI] [PubMed] [Google Scholar]
- Wang LH, Begley J, St Claire RL, Harris J, Wakeford C, Rousseau FS, 2004. Pharmacokinetic and pharmacodynamic characteristics of emtricitabine support its once daily dosing for the treatment of HIV infection. AIDS Res. Hum. Retroviruses 20, 1173–1182. [DOI] [PubMed] [Google Scholar]
- Wang LH, Chittick GE, McDowell JA, 1999. Single-dose pharmacokinetics and safety of abacavir (159U89), zidovudine, and lamivudine administrated alone and in combination in adults with human immunodeficiency virus infection. Antimicrob. Agents Chemother 43, 1708–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weidner DA, Bridges EG, Cretton EM, Sommadossi JP, 1992. Comparative effects of 3′-azido-3′-deoxythymidine and its metabolite 3′-amino-3′-deoxythymidine on hemoglobin synthesis in K-562 human leukemia cells. Mol. Pharmacol 41, 252–258. [PubMed] [Google Scholar]
- Weidner DA, Sommadossi JP, 1990. 3-Azido-3-deoxythymidine inhibits globin gene transcription in butyric acid-induced K-562 human leukemia cells. Mol. Pharmacol 38, 797–804. [PubMed] [Google Scholar]
- Weller S, Radomski KM, Lou Y, Stein DS, 2000. Population pharmacokinetics and pharmacodynamic modeling of abacavir (1592U89) from a dose-ranging, double-blind, randomized monotherapy trial with human immunodeficiency virus-infected subjects. Antimicrob. Agents Chemother 44, 2052–2060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White KL, Margot NA, Ly JK, Chen JM, Ray AS, Pavelko M, Wang R, McDermott M, Swaminathan S, Miller M, 2005. A combination of decreased NRTI incorporation and decreased excision determines the resistance profile of HIV-1 K65R RT. AIDS 19, 1751–1760. [DOI] [PubMed] [Google Scholar]
- White KL, Margot NA, Wrin T, Petropoulos CJ, Miller MD, Naeger LK, 2002. Molecular mechanisms of resistance to human immunodeficiency virus type 1 with reverse transcriptase mutations K65R and K65R+M184V and their effects on enzyme function and viral replication capacity. Antimicrob. Agents Chemother 46, 3437–3446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yeni PG, Hammer SM, Hirsch MS, Saag MS, Schechter M, Carpenter CC, Fischl MA, Gatell JM, Gazzard BG, Jacobsen DM, Katzenstein DA, Montaner JS, Richman DD, Schooley RT, Thompson MA, Vella S, Volberding PA, 2004. Treatment for adult HIV infection: 2004 recommendations of the International AIDS Society-USA Panel. JAMA 292, 251–265. [DOI] [PubMed] [Google Scholar]
- Ying C, De Clercq E, Nicholson W, Furman P, Neyts J, 2000. Inhibition of the replication of the DNA polymerase M550V mutation variant of human hepatitis B virus by adefovir, tenofovir, l-FMAU, DAPD, penciclovir and lobucavir. J. Viral Hepat 7, 161–165. [DOI] [PubMed] [Google Scholar]
- Youle M, NATAP.org, 2004. Studies of new pipeline drugs. HIV Treatment Bulletin 5. [Google Scholar]
- Yuen GJ, Lou Y, Bumgarner NF, Bishop JP, Smith GA, Otto VR, Hoelscher DD, 2004. Equivalent steady-state pharmacokinetics of lamivudine in plasma and lamivudine triphosphate within cells following administration of lamivudine at 300 mg once daily and 150 mg twice daily. Antimicrob. Agents Chemother 48, 176–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zapor MJ, Cozza KL, Wynn GH, Wortmann GW, Armstrong SC, 2004. Antiretrovirals. Part II. Focus on non-protease inhibitor antiretrovirals (NRTIs, NNRTIs, and fusion inhibitors). Psychosomatics 45, 524–535. [DOI] [PubMed] [Google Scholar]
- Zhang R, Lu Z, Diasio CR, Liu T, Soong SJ, 1993a. The time of administration of 3-azido-3-deoxythymidine (AZT) determines its host toxicity with possible relevance to AZT chemotherapy. Antimicrob. Agents Chemother 37, 1771–1776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang R, Lu Z, Liu T, Soong SJ, Diasio RB, 1993b. Circadian rhythm of rat spleen cytoplasmic thymidine kinase. Biochem. Pharmacol 45, 1115–1119. [DOI] [PubMed] [Google Scholar]
- Zhou XJ, Sheiner LB, DAquila RT, Hughes MD, Hirsch MS, Fischl MA, Johnson VA, Myers M, Sommadossi J-P, 1999. Population pharmacokinetics of nevirapine, zidovudine, and didanosine in human immunodeficiency virus-infected patients. Antimicrob. Agents Chemother 43, 121–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
