Abstract
Dolutegravir (DTG), elvitegravir (EVG) and raltegravir (RAL), are members of latest class of antiretrovirals (ARV) available to treat human immunodeficiency virus (HIV) infection, the integrase strand transfer inhibitors (INSTI). INSTIs are potent inhibitors of the HIV integrase enzyme with IC90/95 values in the low nanogram per milliliter range and they retain antiviral activity against strains of HIV with acquired resistance to other classes of ARVs. Each of the INSTIs have unique pharmacokinetic / pharmacodynamic properties, influencing their role in clinical use in specific subsets of patients. RAL and DTG have minimal drug-drug interaction profiles, as their metabolism has minimal cytochrome P450 (CYP) involvement. Conversely, EVG metabolism occurs primarily via CYP3A4 and requires pharmacokinetic boosting to achieve systemic exposures amenable to once daily dosing. EVG and DTG have the added benefit of the availability of fixed dose combination tablets, allowing for convenient and simplified ARV regimens. RAL is the only INSTI to be listed as a preferred agent on the current United States perinatal guidelines. All three of the INSTI agents are recommended regimens for treatment-naïve individuals on the United States Adult and Adolescent HIV treatment guidelines. This review summarizes and compares the pharmacokinetics and pharmacodynamics of the INSTI agents, and describes specific pharmacokinetic considerations for special patient conditions: hepatic impairment, renal dysfunction, pregnancy and co-infections.
1.0 Introduction
There are an estimated 36 million people living with the human immunodeficiency virus (HIV) infection globally. With the advent of antiretroviral therapy (ART), HIV has become a chronic manageable condition. However, in the absence of a sterilizing cure, ART is a life-long commitment. Triple drug combinations consisting of antiretrovirals (ARVs) targeting the virus in two steps in the viral life cycle are the current standard of care for ART [1]. An HIV integrase strand transfer inhibitor (INSTI) co-administered with two nucleoside/nucleotide reverse transcriptase inhibitors (NRTI) are part of five of the six recommended regimens for ART-naïve patients in the US Department of Health and Human Services Adult and Adolescent HIV treatment guidelines [1].
The INSTI class is the newest class of drugs available to treat HIV, and targets the HIV integrase enzyme, which incorporates pro-viral HIV-1 DNA into the host cell genome. The first clinically available INSTI, raltegravir (RAL), was approved in 2007, followed by the second-generation INSTIs elvitegravir (EVG) in 2012 and dolutegravir (DTG) in 2013. In addition to their role in therapy for ART-naïve patients, INSTIs retain potency against strains of HIV that are resistant to other classes of ARVs such as protease inhibitors (PI) and non-nucleoside reverse transcriptase inhibitors (NNRTI). In this manner, INSTIs offer novel treatment options for patients with both acquired and transmitted resistance to other ARV classes.
This review summarizes the clinical pharmacokinetics (PK) of the three currently FDA approved INSTI agents for the treatment of HIV in adult patients. Because selection of optimal ART for special patient populations may be complicated by changes in ARV PK, drug-drug interactions (DDI), or overlapping adverse events, available evidence for the impact of hepatic impairment, renal dysfunction, pregnancy or co-infections on the PK of INSTIs will be summarized. Finally, the pharmacodynamics (PD) of the INSTI agents will be compared.
2.0 Pharmacokinetics of INSTIs
The PK parameters of the HIV INSTIs in healthy subjects and HIV-infected individuals are summarized in Table 1.
Table 1.
Dolutegravir 50 mg Daily | Elvitegravir 150 mg Daily | Raltegravir 400 mg Twice Daily | ||||
---|---|---|---|---|---|---|
Healthy | HIV-infected | Healthya | HIV-infectedb | Healthyc | HIV-infected | |
Cmax (μg/mL) | 6.16 (15) | 3.34 (16) | 2.66 (27.6) | 1.7 + 0.4e | Fasted 1.20 (0.8-1.81)g
Low 0.58 (0.39-0.88)g Mod 1.27 (0.84-1.91)g High 2.36 (1.57-3.56)g |
1.502 (135) |
Cmin (μg/mL) | 1.64 (25) | 0.83 (26) | 0.490 (52.9) | 0.45 ±0.26e | Fasted 0.05 (0.03-0.08)g
Low 0.42 (0.03-0.07)g Mod 0.08 (0.05-0.13)g High 0.20 (0.12-0.33)g |
0.114 (167) |
Tmax (hr) | 1.0 (0.5 to 2.0)f | 2.0 (0.97 to 4.0)f | 5 (5.0,5.5)d | 3 to 4 | 1.0 | 1.8 (0.5 to 4.0)f |
t1/2 (hr) | 13 to 14 | 11 to 12 | 11.0 (8.87,13.0)d | 8.62 (6.1 to 10.9)f | Distribution 1.0 Elimination 11.2 |
9 |
AUCx iag*hr/mL | 76.8 (19) | 43.4 (20) | 27.0 (29.4) | 23.0+7.5e | Fasted 4.44 (3.20-6.22)g
Low 2.39 (1.72-3.33)g Mod 5.02 (3.60-7.02)g High 9.42 (6.76-13.15)g |
5.84 (99) |
Protein Binding | >99% to both albumin and alpha-1-acid glycoprotein |
99.4% albumin >> alpha-1-acid glycoprotein |
76% to 83% to Albumin |
Abbreviations: Cmax, maximum plasma concentration; Cmin, minimum plasma concentration; Tmax, time to reach Cmax; t1/2, elimination half life; AUCx, area under the concentration-time curve over the dosing interval Data are presented as geometric mean (CV) unless otherwise noted.
healthy volunteers receiving EVG 150mg plus 150mg COBI.
HIV-infected data is EVG 150mg plus COBI 150 mg.
Data are presented for fasted, low, moderate and high fat meals.
Data are presented as median (Q1,Q3).
Data are presented as mean (SD)
Data are presented as median (range).
Data are presented as GM (90% CI)
2.1 Dolutegravir
DTG is available as a 50mg film coated tablet, and a fixed dose combination (FDC) tablet containing DTG 50mg, abacavir (ABC) 600mg, and lamivudine (3TC) 300mg. DTG may be given to adults as 50mg once daily for patients without INSTI-associated resistance substitutions, or 50mg twice daily for patients with known or suspected INSTI-associated resistance substitutions.
Studies describe DTG (S/GSK1349572) PK of single and multiple dose strategies with doses ranging from 2mg to 100mg in healthy volunteers. DTG was readily absorbed with a median maximum concentration (Cmax) achieved between 0.5 and 1.25 hours post dose in healthy volunteers; similar studies in HIV-infected persons found Cmax occurred within 2.5 hours [2, 3]. Low, moderate and high fat meals increased DTG area under the concentration time curve (AUC) 33%, 41%, and 66%, respectively, although current manufacturer prescribing information indicates DTG may be taken without regard to meals [4, 5]. Consistent with other INSTIs, DTG absorption is impaired by coadministration with divalent or trivalent cations, which may be overcome by dose separation. Once absorbed, DTG extensively binds (>99%) to plasma proteins, both albumin and alpha-1-acid glycoprotein [6, 3]. Bioequivalence of a FDC tablet compared with single tablet DTG 50mg and combination ABC/3TC 600mg/300mg has been demonstrated [7].
DTG exhibits bi-exponential elimination with a terminal half-life (t1/2) ranging from 13 to 15 hours in healthy volunteers, while a t1/2 of 11 to 12 hours was reported in HIV-infected individuals [3] [2]. DTG exposure, as measured by AUC, was dose proportional from 2mg to 100mg, while Cmax was slightly less than dose proportional within single dose healthy volunteer studies [3]. Repeated dose studies found AUCτ and plasma concentrations at the end of the dosing interval (Ctrough) to increase proportionally with doses in the 10mg to 50mg range, while Cmax increased slightly less than proportionally. Steady state was reached within 5 days of daily dosing in healthy volunteer, multiple dose studies. AUC, Cmax and Ctrough accumulation ratios ranged from 1.24 to 1.42, 1.16 to 1.36, and 1.29 to 1.53, respectively, within the doses evaluated [3]. Similar accumulation ratios (1.23 to 1.43) were observed in studies of HIV-infected individuals [2]. After DTG 50mg orally daily, mean steady state DTG Ctrough are ~25-fold higher than the protein adjusted 90% inhibitory concentration (IC90) in healthy volunteers [3].
In vitro, DTG was found to be a substrate for the efflux transporters P-glycoprotein (P-gp) and human breast cancer resistance protein (BCRP). In human hepatocytes, UDP-glucuronosyltransferase 1A1 (UGT1A1) was the primary enzyme responsible for DTG metabolism while cytochrome P450 (CYP) 3A4 was a minor metabolizing pathway along with minimal contribution from UGT1A3 and UGT1A9. DTG is a substrate for UGT and CYP enzymes, but it does not appear to significantly induce or inhibit these enzymes. DTG also did not inhibit multidrug resistance associated protein 2 (MRP2), organic anion transporting polypeptide (OATP) 1B1/3, organic cation transporter (OCT) 1 or the following CYPs: 1A2, 2A6, 2B6, 2C9, 2C19, 2D6, 3A4, or 2B7. Notably, DTG inhibits OCT2 (IC50=1.9 µM), which provides a basis for mild increases in serum creatinine seen in clinical use of DTG in the treatment of HIV infection. Collectively, the in vitro data demonstrate DTG’s low potential for clinically meaningful DDIs [8].
The in vivo metabolism and excretion of DTG was studied in a mass balance study of healthy male volunteers. After a 20mg oral dose of DTG, 95.6% of the dose was recovered in feces (64%) and urine (31.6%). Unchanged DTG was the primary circulating entity in blood plasma, while an inactive glucuronide (18.9%) formed via UGT1A1 was the principal metabolite recovered in urine. Minor metabolic pathways were identified via oxidation by CYP3A4 (7.9%) as well as oxidative deflourination and glutathione substitution (1.8%) [9].
Population PK analysis in treatment-naïve HIV-infected persons was performed by combining data from three studies: a proof-of-concept study (ING111521) [2], a Phase 2b study( SPRING-1, ING112276, NCT00951015) [10], and a Phase 3 study (SPRING-2, NCT01227824) [11].
Population parameter estimates, derived from data of a combined 3357 plasma samples from 563 subjects, were: apparent oral clearance (CL/F) 0.901 L/hr; apparent volume of distribution (Vd/F) 17.41 L; absorption rate constant 2.24 hr-1; and absorption lag time 0.263 hr. Weight, smoking status, age and total bilirubin were predictors of CL/F although none of these covariates were found to be clinically significant. Race and ethnicity, hepatitis B virus (HBV) and hepatitis C virus (HCV) co-infection, creatinine clearance (CLCR), albumin, alanine aminotransferase and aspartate aminotransferase did not impact the PK of DTG within the studied population [12].
DTG benefits from having relatively low PK variability when compared with other INSTI agents. In a phase IIa study investigating DTG PK in HIV-infected individuals, variability was found to be low with coefficients of variation (CV) in the 25-50% range for Cmax, Ctrough and AUCτ [3, 2].
2.2 Elvitegravir
EVG (GS9137) is available in two FDC tablets, each given once daily in adults and containing EVG 150mg, the PK enhancer cobicistat (COBI), and emtricitabine (FTC). One product includes tenofovir disoproxil fumarate (TDF), while the other includes tenofovir alafenamide (TAF). Additionally, EVG 85mg is available for use once daily in individuals receiving a ritonavir (RTV) boosted PI-based ART. The manufacturer recommends all formulations be taken with a meal [13-15].
In single dose studies of unboosted EVG at doses of 100mg, 200mg, 400mg and 800mg given orally to healthy individuals mean (%CV) Cmax concentrations of 108 (55), 160 (19), 264 (30) and 455 (33) ng/mL were reported with each respective dose. Tmax occurred 1.3-2.5 hours post dose and AUC∞ was found to be less than dose proportional [16].
During the clinical development of EVG it was recognized that co-administration of RTV, a potent CYP3A4 inhibitor used for PK boosting of PIs, resulted in substantially higher EVG exposures. EVG AUCτ were ~20 fold higher when given with RTV 100mg compared with EVG alone [17]. COBI (GS-9350), a novel, potent and selective CYP3A4 inhibitor was subsequently studied for its ability to enhance EVG PK. COBI has advantages as an ARV PK enhancer because it inherently lacks any activity against HIV-1 and is better tolerated than RTV. The relative bioavailability of EVG boosted with COBI (EVG/COBI) was compared with EVG boosted with RTV 100 mg (EVG/RTV) in a study of healthy human subjects. Relative to EVG/RTV, the geometric least-squares means ratios [90% CI] for AUCτ, Cmax, and Ctrough of EVG were 118 (110 to 126), 108 (100 to 116), and 110 (95.3 to 127), respectively, when EVG 150mg was combined with COBI 150mg[18]. Because EVG efficacy is closely linked with Ctrough concentrations, a higher dose of 150mg COBI was subsequently investigated. With the higher dose of COBI, the EVG AUCτ were bioequivalent with EVG/RTV exposures, while both Cmax and Ctrough were significantly greater than those observed with EVG/RTV. The median (Q1,Q3) t1/2 of EVG (with COBI) was 9.15 hr (7.70,12.4) compared with 3 hours when given alone [19, 18]. Collectively, these studies provided evidence for once daily dosing of EVG when given with COBI 150mg. The available EVG FDC products are both formulated with COBI 150mg [14, 15].
The oral bioavailability of EVG is significantly enhanced when it is given with a meal. A food interaction study of EVG, when given as EVG/COBI/FTC/TDF found mean AUC∞ and Cmax increased by 34% and 24%, respectively, when given with food [20]. Follow up studies in healthy Japanese males confirmed these findings, showing decreases in AUC∞ of up to 50% when EVG is taken on an empty stomach as compared with either a standard breakfast or protein rich breakfast [21].
When EVG is given orally with food, Cmax is reached in approximately 4 to 4.5 hours. Although EVG absorption is not changed with alterations in stomach pH, absorption is significantly decreased by co-administration with divalent and trivalent cations such as those found in multivitamins and antacids, which may be overcome by separating the dose times of the two products [22, 23]. EVG absorption does not appear to be significantly altered with changes in intestinal P-gp expression. Ex-vivo experiments demonstrate EVG is highly bound (>99%) to plasma proteins both albumin and alpha-1 acid glycoprotein with a preference for the former.
EVG’s metabolism occurs primarily via CYP3A4 in the liver and intestine. Additional metabolism of EVG occurs via UGT1A1 and 1A3 to primary and secondary glucuronidated metabolites. The primary metabolites of EVG have substantially reduced activity against HIV-1 and do not play a major role in EVG efficacy [24]. Mass balance studies in healthy human subjects after a radiolabeled dose of EVG 50mg plus RTV 100mg demonstrated that ~95% of the oral dose was recovered in feces while ~6.7% of the dose was recovered in urine. Untransformed EVG was the primary circulating entity in plasma, accounting for ~93% of the circulating radioactivity.
2.3 Raltegravir
RAL (MK-0518) is available in three formulations: a 400mg film coated tablet, 25mg and 50mg chewable tablets, and 100mg granules for suspension [25]. For adults, the dose of RAL is 400mg twice daily, with or without food RAL PK are distinguished within the INSTI class as having markedly high inter- and intrapatient variability. Coefficients of variation of greater than 200% have been observed from repeated plasma concentrations within individual patients on stable RAL containing ART [26]. RAL is rapidly absorbed with a Tmax of approximately 3 hours. Once absorbed, RAL is approximately 76 to 83% protein bound, mainly to albumin and not to alpha-1-acid glycoprotein [25, 27]. Secondary plasma concentration versus time peaks often occur with RAL, indicative of either enterohepatic recirculation or possibly delayed oral absorption. Similar to the other INSTIs, RAL PK is altered when given with divalent and trivalent cations, which may be overcome by dose-separation [28]. The absolute bioavailability of RAL has not been established due to the lack of a parenteral formulation, however, HIV-infected individuals have a 25-30% reduced bioavailability relative to healthy subjects [29]. Food appears to increase the amount of variability in the PK of RAL, although the mechanism is not completely clear. Studies of fasted and fed effects on RAL PK revealed significant changes in AUC. Compared with a low fat meal, a 46% decrease in AUC was observed when RAL is given in the fasted state. However, after moderate or high fat meals, AUCs were increased 13 to 212% [30]. Although substantial effects on PK are noted with meals, no significant differences have been noted in efficacy when RAL is given with or without food [25].
In healthy human subjects RAL Tmax occurred between 1 to 2 hours post dose at steady state. Terminal half-life (t1/2) was 7 to 12 hours after multiple doses of 100 mg to 800 mg twice daily. Steady state is reached in approximately two days of twice daily dosing and little accumulation occurs. Cmax is slightly less than dose proportional with observed geometric means (GM) of 4.97μg/mL and 8.77μg/mL after multiple 400mg and 800mg doses, respectively. The mean renal clearance of RAL in an adult male at 400mg twice daily at steady state is 3.63 L/hr. AUC∞ was found to be similar between adult male and female subjects [31], which is in contrast to combined data from HIV-infected individuals and healthy human subjects, where 65% higher RAL exposures were observed in females than males [29].
In a study of ART-naïve HIV-infected adults given 10 days of RAL twice daily doses of 100mg, 200mg, 400mg, or 600mg, Ctrough increased with higher doses while Cmax concentrations increased up to the 400mg dose; the 600mg dose had a slightly lower Cmax than the 400mg dose (GM 2.0 vs 1.69 μg/mL) [32]. Because of the large PK variability noted in this study, the RAL t1/2 could not be estimated. The authors did note the elimination profiles of RAL in this patient population appeared to mimic those seen in healthy volunteer studies where elimination occurred in a bi-exponential fashion with an initial t1/2 of approximately 1 hour and a terminal t1/2 of 7 to 12 hours [31-33].
A mass balance study performed in healthy human subjects after a single radiolabeled dose of 200 mg RAL indicated 51% of the dose was recovered in the feces while 32% was recovered in urine [34]. Both parent RAL and a glucuronide metabolite of RAL (M2) were present in the urine and represented 9% and 23% of the total RAL dose given. RAL was the only form of the dose recovered in the feces although it is believed RAL recovered from feces includes glucuronidated RAL, which was further hydrolyzed in bile before elimination. The major circulating entity in blood plasma was RAL that represented 70% of the total radioactivity. The remaining radioactivity could be attributed to RAL glucuronide metabolites. Mechanistic studies indicate that UGT1A1 is the major metabolic enzyme responsible for clearance of RAL in humans with minor contributions from UGT1A3 and UGT1A9. Genetic polymorphism in UGT1A1, such as the *28/28, result in lower clearance of RAL and associated higher plasma concentrations (~41% higher), however, this increase is not considered clinically significance [35].
The PK of RAL 800mg once daily were investigated in HIV-infected, treatment-naïve individuals. AUCτ was similar when RAL was given as 800 mg daily compared with 400mg twice daily [GM ratio (90% CI) 1.17 (0.8-1.72)]. However, Cmax was ~4 fold higher [3.98 (2.58-6.16)] and Ctrough was ~6 fold lower [0.15 (0.09-0.26)] with the daily dose. Once daily dosing of RAL was ultimately found to be virologically inferior to twice daily dosing (see section 4.3 for further discussion) [36, 37, 25].
3. Pharmacokinetics in Special Populations
3.1 Hepatic Impairment
Liver disease may influence ARV PK due to changes in liver blood flow or shunting, altered synthesis of plasma proteins, metabolism via CYP enzymes, and to a lesser extent, glucuronidation [38-40] . The FDA has formal guidance for evaluating the effect of hepatic impairment on drug PK [41]. Product labeling for all INSTIs recommends no dosage adjustment for patients with mild to moderate hepatic impairment (Child-Pugh A or B), but none of the labels recommend either the use of, or dosing information for, patients with severe hepatic impairment (Child-Pugh C) [25, 4, 13-15]. Unless indicated, moderate hepatic impairment was defined as a Child-Pugh Score of 7-9 for all studies described in this section. Table 2 summarizes comparative studies of INSTI PK in patients with hepatic impairment.
Table 2.
Antiretroviral | Study design | Participants | Cmax Ratio Impairment vs. Healthy |
Cmin Ratio Impairment vs. Healthy |
AUCχ Ratio Impairment vs. Healthy |
---|---|---|---|---|---|
Moderate hepatic impairment | |||||
Dolutegravira
[43] |
Parallel-group, single dose, PK evaluation over 72 hours HIV seronegative volunteers |
Moderate hepatic impairment:b n=8 Matched, healthy volunteers: n=8 |
1.02 (0.754, 1.37) | na | 1.05 (0.745, 1.49) |
Elvitegravirc [45] | Parallel-group, steady state, PK evaluation over 96 hours HIV seronegative volunteers EVG given as EVG/c/TDF/FTC given daily for 10 days |
Moderate hepatic impairment:b n=10 Matched, healthy volunteers: n=10 |
EVG: 1.41 (1.09, 1.83) COBI: 0.861 (0.654, 1.13) |
EVG: 1.80 (1.11, 2.91) COBI: 2.08 (1.17, 3.68) |
EVG: 1.35 (1.03, 1.77) COBI: 0.997 (0.76, 1.31) |
Raltegravirc [46] | Parallel-group, single dose, PK evaluation over 96 hours HIV seronegative volunteers |
Moderate hepatic impairment:b n=8 Matched, healthy volunteers: n=8 |
0.63 (0.23, 1.70) |
1.26 (0.65, 2.43) |
0.86 (0.41, 1.77) |
Raltegravird [47] | Parallel-group, steady state, Ctrough evaluation HIV-infected patients with and without HCV co-infection |
HIV/HCV co-infection:e
n=13 HIV-infected: n=16 |
na | 1.16 (0.56, 1.39) |
na |
Raltegravirc [48] | Parallel-group, steady state, PK evaluation over a 12-hour dosing interval HIV-infected patients with HCV co-infection |
HIV/HCV co-infection with advanced cirrhosis:f n=5 HIV/HCV co-infection without cirrhosis:g n=5 |
1.15 (0.55, 2.43) |
6.58 (2.92, 14.85) |
1.72 (1.02, 2.92) |
Severe renal impairment | |||||
Dolutegravira [51] | Parallel-group, single dose, PK evaluation over 72 hours HIV-seronegative volunteers |
CLCRl<30ml/min: n=8 Healthy volunteers: n=8 |
0.774 ( 0.532, 1.13) |
0.566 (0.352, 0.908) |
0.6 (0.37, 0.975) |
Raltegravirc [46] | Parallel-group, single dose, PK evaluation over 96 hours HIV-seronegative volunteers |
CLCR<30ml/min: n=10 Healthy volunteers: n=10 |
0.68 (0.35, 1.32) |
1.28 (0.79, 2,06) |
0.85 (0.49, 1.49) |
Abbreviations: ART, antiretroviral therapy; AUC∞, area under the concentration time curve; Cmax maximum concentration observed; Cmin minimum concentration observed; COBI, cobicistat; CLCR, estimated creatinine clearance; DTG, dolutegravir; EVG, elvitegravir; EVG/c/TDF/FTC, elvitegravir/cobicistat/tenofovir disoproxil fumarate/emtricitabine co-formulated tablet; HCV, hepatitis C virus; HIV, human immunodeficiency virus; na, not available.
Data presented as geometric least squares mean ratio (90% confidence interval).
Moderate hepatic impairment was defined as Child-Pugh score 7-9.
Data presented as geometric mean ratio (90% confidence interval).
Data presented as geometric mean ratio (95% confidence interval).
Patients with mild or moderate hepatic impairment, all without cirrhosis.
Advanced cirrhosis based on biopsy, Metavir score F4.
Non-cirrhosis based on biopsy, Metavir score F0-F1.
3.1.1 Dolutegravir
The Phase II/III efficacy trials for DTG included 71 patients with HIV/HCV and 27 patients with HIV/HBV coinfection without cirrhosis. Population PK in the HCV coinfected patients indicated no clinically relevant effect on DTG PK, but the evaluation was limited for HIV/HBV coinfected patients due to sample size [42, 12]. One study evaluated DTG in HIV-seronegative participants with moderate hepatic impairment compared with healthy participants [43]. After a single dose of DTG 50 mg, no difference was observed in PK parameters for total DTG concentrations. Unbound DTG concentrations were 48-106% higher in the participants with hepatic impairment compared with healthy participants at two time points post-dose. Higher unbound fractions were correlated with a higher Child-Pugh score, lower albumin concentrations, and increased bilirubin concentrations (all P<0.001).
3.1.2 Elvitegravir
Population PK evaluations from the Phase II/III efficacy trials in HIV-infected patients with HBV or HCV coinfection identified no impact of coinfection without cirrhosis on EVG PK when combined with COBI (n=24) [44] or RTV (n=56) [13]. The impact of hepatic impairment on steady state exposure of EVG was assessed in HIV-seronegative volunteers receiving EVG/COBI/TDF/FTC [45]. EVG exposure was 35% higher in participants with moderate hepatic impairment. Overall COBI exposure, EVG free fraction, and COBI free fraction was similar between groups.
3.1.3 Raltegravir
The PK of RAL 400mg after a single dose was evaluated in HIV-seronegative volunteers with or without moderate hepatic insufficiency [46]. No clinically significant differences were observed in any of the RAL PK parameters. In another study in HIV-infected patients, no difference in RAL Ctrough was observed in HIV/HCV co-infected persons without cirrhosis compared with HIV monoinfected persons after 4 weeks of RAL 400mg twice daily [47].
Unique to RAL, published data describe RAL exposure in HIV/HCV coinfected individuals with cirrhosis. HIV/HCV coinfected patients with and without advanced cirrhosis received RAL 400mg twice daily for five days in combination with their existing PI-based ART regimen [48]. The patients with advanced cirrhosis had 72% higher total RAL exposure and a notably higher Ctrough. The authors reported no adverse events or RAL treatment-discontinuation during the study period.
RAL PK was also evaluated in HIV-infected persons with end stage liver disease (ESLD) and in patients post-liver transplantation [49]. Ten HIV-infected individuals with ESLD (MELD score ≥15) were switched from effective ART (HIV-RNA < 50 copies/mL) to RAL-based ART. After one month on therapy, total RAL exposure was higher in patients with ESLD (median area under the concentration-time curve during a 9 hour time interval (AUC9): 33.5 mcg*hr/mL) than historical controls with normal hepatic function. In these 10 persons, RAL was well tolerated and all participants remained virologically suppressed on ART. Consistent with RAL PK in other populations, high variability was observed in total and unbound RAL concentrations (AUC9 interpatient CV%, 95% and 91%, respectively). The same study reports RAL PK in five HIV-infected persons who underwent liver transplantation and initiated RAL-based ART post-operatively. After one month, the total and unbound RAL exposures were similar to those observed in persons with ESLD.
3.2 Renal Impairment
Because none of the INSTIs are primarily eliminated by renal excretion, a significant impact on INSTI exposure is not expected in persons with renal impairment. Because renal impairment can influence hepatic metabolism and drug transporters, a renal impairment study is required for chronically administered drugs [50]. Table 2 summarizes the comparative studies conducted in severe renal impairment. Unless indicated, severe renal impairment was defined as an CLCR<30 mL/min for all studies discussed in this section.
3.2.1 Dolutegravir
Unexpectedly, total plasma DTG exposure was 40% lower following a single dose of DTG 50mg in HIV-seronegative volunteers with severe renal impairment compared with matched controls [51]. Population PK modeling from the Phase III clinical trial did not find an association between the degree of mild or moderate renal impairment and total DTG exposure, and the reason for lower DTG concentrations during severe renal insufficiency is unclear [42]. Given these results, paired with the PD of DTG (see section 4.1 for further discussion), DTG may be given without dosage adjustment for INSTI-naïve persons with mild, moderate, or severe renal impairment. However, due to this observed decrease in DTG exposure, caution is warranted for those with severe renal impairment and known or suspected INSTI-resistance mutations [4]. DTG has not been evaluated in persons receiving renal replacement therapy, however due to high protein binding (>99%), it is not expected to be removed significantly [4, 51].
3.2.2 Elvitegravir
The EVG/COBI/TDF/FTC coformulated tablet is not recommended for initial therapy in persons with a CLCR ≤70 ml/min due to an imbalance of patients on EVG/COBI/TDF/FTC developing renal adverse events in trials of clinical efficacy (9 vs. 3 persons on comparator regimens) [44]. In addition, EVG/COBI/TDF/FTC should be discontinued in persons whose CLCR falls below 50 ml/min during therapy due to the coformulation with TDF, because TDF requires a dose reduction at CLCR below this threshold. Given these restrictions, there are no PK data available for this formulation in persons with any degree of renal insufficiency [44].
EVG/COBI/TAF/FTC may be used for patients with moderate renal insufficiency (CLCR 30-69 ml/min). The safety and efficacy of EVG/COBI/TAF/FTC was evaluated in 242 patients with stable, moderate renal insufficiency (median CLCR 56 ml/min), and intensive PK was conducted in 30 participants between weeks 4-8 [52]. EVG and COBI exposure (AUC 27.1 and 9.92 mcg*h/mL, respectively) was similar to historical data [44], and was comparable between persons with CLCR above or below 50 ml/min.
Minimal data are available for EVG when co-administered with RTV. Data on file with the company indicate that no clinically relevant differences in EVG PK were observed in participants with severe renal impairment when EVG is boosted with RTV [13] . Removal of EVG by renal replacement therapy is expected to be low due to high EVG protein binding (98-99%).
3.2.3 Raltegravir
No differences in PK parameters were observed in one evaluation of single dose RAL 400mg in HIV-seronegative participants with severe renal insufficiency [46]. The extent of drug removed by renal replacement therapy is expected to be minimal due to high plasma protein binding, and this is confirmed in case reports to date [53, 54].
3.3 Pregnancy
Similar to non-pregnant adults, ART is recommended for all HIV-infected pregnant women by the U.S. Perinatal HIV treatment guidelines [55]. In addition to the known efficacy, safety, and tolerability of INSTIs in the treatment of non-pregnant adults, INSTIs may play an important role in managing HIV infection during late pregnancy due to the rapid viral decay observed with INSTI therapy. Based on accumulating clinical and PK data, RAL was included as a preferred option for ARV-naïve pregnant women in the 2015 update of the U.S. Perinatal HIV guidelines in 2015 [55].
Physiologic changes during pregnancy are known to impact the PK of some ARVs, resulting in dose adjustment recommendations for select ARVs during pregnancy in order to maintain optimal PK exposure. Specific metabolic pathways that may influence INSTI metabolism include induction of UGT1A1 and CYP3A4 during pregnancy [56, 57]. Therefore, guidelines recommend an assessment of ARV PK during pregnancy prior to routine clinical use [55]. Published data on INSTI PK in pregnant women are summarized in Table 3. In addition to maternal influences on ARV PK, fetal exposure to ARVs during the antepartum period is also important for the prevention of HIV transmission. McCormack and Best recently published a comprehensive review of ARV placental transfer [58]; therefore, only in vivo information published since this recent review is described herein.
Table 3.
Antiretroviral | Study design | Participants | AUCt (μg*hr/mL) | Cmin (μg/mL) | Comments |
---|---|---|---|---|---|
Dolutegravira
[59] |
Longitudinal, steady state, intensive PK sampling over 24 hours |
2nd trimester: n=9 | 58.4 (47.6 – 64.5) |
0.86 (0.78 – 1.05) |
No statistically significant differences were observed. Individual AUCx above the 10th percentile of non-pregnant adults (37.5 mcg/mL):
|
3rd trimester: n=15 |
48.7 (40.3 – 57.6) |
1.03 (0.87 – 1.24) |
|||
Postpartum: n=10 |
71.1 58.0 – 83.1) |
0.70 (0.60 – 0.86) |
|||
Raltegravirb [62] | Longitudinal, steady state, intensive PK sampling over 12 hours after an observed dose while fasting |
2nd trimester: n=16 |
6.6 (2.1 – 18.5) |
0.047 (<0.01 – 0.162) |
AUCx was statistically lower in both the 2nd and 3rd trimester compared to postpartum. Cmin was statistically lower in the 2nd trimester compared to postpartum. No differences were observed between 2nd and 3rd trimesters. |
3rd trimester: n=41 |
5.4 (1.4 – 35.4) |
0.057 (<0.01 – 0.607) |
|||
Postpartum: n=38 |
11.6 (1.6 – 39.9) |
0.0474 (<0.01 – 0.917) |
|||
Raltegravirc [63] | Longitudinal, steady state, intensive PK sampling over 12 hours after an observed dose with breakfast |
3rd trimester: n=21 |
5.0 (3.56, 7.01) |
0.077 (0.043, 0.137) |
90% confidence intervals of the geometric mean ratios (3nd trimester:postpartum) of all PK parameters included 1, except AUCτ [0.71 (0.53 - 0.96)]. 11 of 17 patients (65%) with paired visits had lower exposure during the 3rd trimester vs. postpartum. |
Postpartum: n=18 |
7.11 (4.91, 10.31) |
0.120 (0.074, 0.193) |
Abbreviations: AUCx, area under the concentration time curve for the dosing interval; Cmin, minimum concentration observed; PK, pharmacokinetic.
Data are presented as geometric mean (interquartile range).
Data are presented as median (range).
Data are presented as geometric mean (95% confidence interval).
3.3.1 Dolutegravir
Perinatal treatment guidelines do not recommend, or make specific dosing recommendations for DTG use in ARV-naïve pregnant women [55]. Since the guideline’s last update, the first PK evaluation of DTG in 21 pregnant women was presented [59]. DTG exposure was lower during pregnancy compared with postpartum in the same group of women. Although DTG PK was variable, the authors concluded the postpartum PK was similar to historical data [4], suggesting the postpartum exposure was an appropriate control group. In the same study, the exposure to, and elimination of, DTG was assessed in 10 infants over 5-9 days after delivery. The median concentration 6.9 hours after delivery was 1.96 (IQR 1.42 – 2.48) µg/mL, demonstrating significant maternal:fetal transfer of DTG; the t1/2 was 34.5 hours, in contrast to 14 hours reported in historical adult patients [4].
3.3.2 Elvitegravir
No systematic evaluation of EVG PK during pregnancy is available; therefore, it is not recommended by U.S. treatment guidelines [55]. The first case report of EVG use during pregnancy was recently published, administered as EVG/COBI/TDF/FTC once daily [60]. The woman’s AUCτ of EVG and COBI were measured at 34 weeks gestational age and 6 weeks postpartum. EVG AUCτ was similar during and after pregnancy (data not provided); however, the patient’s AUCτ postpartum was lower than historical data. The Ctrough after observed drug intake was 60% lower during pregnancy compared with postpartum, and was below the study defined target concentration 0.13 µg/mL [61]. Exposure to COBI (AUCτ) was 44% lower during pregnancy compared with postpartum, which was similar to historical data. At delivery, the EVG maternal and cord plasma concentrations of EVG were both 0.3 µg/mL; however, COBI was below the lower limit of quantitation (LLQ, 0.03 µg/mL) in both samples.
3.3.3 Raltegravir
Similar to non-pregnant adults, RAL exhibits high PK variability during pregnancy when given either with or without food, as observed in two studies of pregnant and postpartum women. In the first, RAL was given on an empty stomach to 42 women during pregnancy [62]. RAL exposure (AUCτ) was approximately 50% lower during both the 2nd and 3rd trimesters of pregnancy compared with postpartum. The authors selected a target Ctrough concentration of 0.035 µg/mL based on the 10th percentile value in nonpregnant historical controls [25]; 69%, 80%, and 79% of participants were above this Ctrough target during the 2nd trimester, 3rd trimester, and postpartum, respectively, with a notably high variability (<0.01 – 0.917 µg/mL).
More recently, RAL was given with a meal (650 kcal; 30 g fat) during the 3rd trimester (median gestational age 33 weeks) and postpartum (median 5 weeks) in 22 women [63]. Overall, RAL AUCτ and Ctrough were 29% and 36% lower during pregnancy compared with postpartum; significant variability was observed in all PK parameters. Three participants (13.6%) failed to achieve virologic suppression before delivery. Of those, one person’s Ctrough during the 3rd trimester was below the desired threshold established by the authors [0.020 µg/mL, based on the comparison of RAL 800mg once daily to 400mg twice daily [64] (see section 4.3 for further discussion)]. No other patient fell below this threshold during or after pregnancy. Fetal exposure to RAL was evaluated in nine paired cord and maternal blood samples at delivery (median 10 hours post-last RAL dose). The ratio of RAL in cord/maternal blood was 1.21 (IQR 1.02-2.17), similar to prior reports [58].
Given the available data, high interpatient variability, and the virologic efficacy observed in these studies (92% [62] and 86% [63] virologic suppression at delivery), U.S. treatment guidelines currently recommended RAL 400mg twice daily without dose adjustment in ARV-naïve pregnant women [55].
3.4 Co-Infections
PK evidence in both healthy volunteers and individuals co-infected with HIV and tuberculosis (TB) have shown substantial reductions in RAL exposure when given in combination with rifampicin. Two studies have identified approximately 40% lower RAL AUC when combined with rifampicin, presumably from the UGT1A1 induction effect of rifampicin [66, 35]. PD outcomes when combining RAL and rifampicin have also been studied. In individuals co-infected with HIV/TB, the ANRS 12 180 (REFLATE TB) study investigated the use of RAL-based ART in individuals receiving drug-sensitive TB treatment including rifampicin. In an analysis of 153 individuals randomized to either RAL 400mg twice daily, RAL 800mg twice daily or EFV 600mg once daily, all in combination with TDF/3TC, RAL 400mg twice daily performed similarly to either dose-escalated RAL 800mg twice daily or EFV-based regimens: week 24 virologic suppression rates were RAL 400mg, 76%; RAL 800mg, 78%; and EFV, 63% [65]. Studies investigating the use of INSTI based ART in HIV and hepatitis C virus (HCV) co-infected individuals are lacking. Drug-drug interaction data in healthy volunteers suggest minimal effects of newer HCV direct acting antivirals (DAA) on RAL and DTG PK [1]; however, effects of both disease state and treatment of HCV on INSTI PD remain to be investigated. PK and PD data are deficient for use of INSTI’s in individuals co-infected with HIV and malaria. Based on known pathways of metabolism for the INSTI agents, DDIs between RAL and DTG are unlikely with antimalarial agents, while DDIs with EVG/c or EVG/r may be expected.
4. Pharmacodynamics
Table 4 provides a comparison of select PD characteristics for the three INSTIs.
Table 4.
Dolutegravir | Elvitegravir | Raltegravir | |
---|---|---|---|
IC50 (ng/mL) IC90 (ng/mL) IC95 (ng/mL) |
16 64 na |
7.2 na 44.9 |
na na 14.7 |
EC50 (ng/mL) EC90 (ng/mL) |
36 324 |
14 126 |
na na |
IQ (Ctrough / IC90 or 95) |
17 | 10 | 8 |
Abbreviations: Ctrough, measured concentration at the end of the dosing interval; IC, protein-binding adjusted concentration inhibiting viral replication by 50, 90 or 95% in vitro; EC, concentration producing 50 or 90% effect (reduction of HIV-RNA) in vivo; IQ, inhibitory quotient; na, not available.
4.1 Dolutegravir
The PD characteristics of DTG have been generated through studies when given as monotherapy, and in combination with NRTIs to treatment-naïve, and treatment-experienced persons with and without documented resistance to RAL and EVG. DTG monotherapy in doses of 2, 10 and 50mg once daily for 10 days was given to 35 INSTI-naïve HIV-infected persons not receiving ARV therapy [67]. Monotherapy was associated with a reduction in plasma HIV-RNA ranging from 1.5 log10 for 2mg, 2.03 for 10mg and 2.46 for 50mg. Seven of 10 recipients of 50mg once daily had plasma HIV-RNA < 50 copies/mL at day 11. Plasma HIV-RNA reductions (log10 change) were best predicted by DTG Ctrough following a maximum effect (Emax) relationship. The estimated EC50 was 36 ng/mL; with a Hill Factor of 1, a concentration of 324 ng/mL was associated with 90% of Emax. The geometric mean Ctrough observed for the three doses were 2mg, 40 ng/mL; 10mg, 190 ng/mL and 50mg, 830 ng/mL. These data indicate that DTG monotherapy at 50mg once daily produced concentrations well on the plateau portion of the exposure-response relationship. A subsequent dose ranging study evaluated DTG (10mg, 25mg or 50mg once daily) or efavirenz (EFV), in combination with TDF/FTC or ABC/3TC in ARV-naïve persons [10]. At week 48, the proportion of participants who had HIV-RNA < 50 copies/mL were 91%, 88% and 90% for the DTG 10mg, 25mg and 50mg groups, respectively, and 82% for those receiving EFV. The comparability of virologic responses across the three different DTG doses suggests that additive to synergistic anti-HIV responses were achieved when DTG was combined with other ARVs.
DTG 50mg once daily was compared with twice-daily RAL in 822 treatment-naïve adults [11]. Both were given with investigator selected ABC/3TC or TDF/FTC. At the 48-week primary endpoint, 88% of DTG recipients and 85% of RAL recipients had HIV-RNA < 50 copies/mL. At 96 weeks, 81% of DTG compared with 76% of RAL recipients had HIV-RNA < 50 copies/mL. Virologic non-response occurred in 5% of those randomized to DTG and in 10% of those to RAL. In a similar design, once daily DTG (50mg) was compared with once daily DRV/RTV, each given with investigator selected ABC/3TC or TDF/FTC, in 484 ARV-naïve persons [68]. At the week 48 primary endpoint, 90% of DTG recipients and 83% of DRV/RTV recipients had HIV-RNA < 50 copies/mL. DTG was statistically superior (p=0.025) to DRV/RTV. Drug-limiting adverse events were less frequent in the DTG recipients: 2% of DTG recipients and 4% of DRV/RTV recipients discontinued therapy for adverse events.
The safety and efficacy of DTG (n=354) was compared with RAL (n=361) in INSTI-naïve but ARV-experienced persons with at least two-class resistance [69]. A total of 715 persons were enrolled. At week 48, 71% of DTG recipients vs. 64% of RAL recipients had plasma HIV-RNA < 50 copies/mL (p=0.03). Additionally, significantly fewer DTG recipients had virologic failure with treatment-emergent INSTI resistance (4 vs. 17, p=0.003). In this study, the geometric mean DTG Ctrough across 335 subjects was 850 ng/mL [42]. Those who had DTG Ctrough in the lowest quartile (median Ctrough, 260 ng/mL) had the lowest virologic response rates (HIV-RNA <50 copies/mL at week 24): 76% in lowest quartile vs. 81-87% across the 2nd through 4th quartiles.
Subjects with DTG Ctrough in the lowest quartile were more likely to have concentrations below the lower limit of quantification (LLQ) likely reflecting poor adherence and/or receiving CYP3A inducers; the median DTG Ctrough in the lowest quartile is consistent with a 10mg once daily dose.
DTG PK were shown to be less than dose proportional in healthy human volunteers when increasing from 50mg to 100mg daily, for this reason the dose of 50mg twice daily was investigated in an attempt to increase DTG exposures in patients with prior INSTI resistance [70]. 183 treatment-experienced persons with documented resistance to RAL and EVG were enrolled in an evaluation of DTG at 50mg twice daily, first in a functional 7 day monotherapy phase followed by a second phase with optimized ARV background [70]. At week 24, 69% of subjects had plasma HIV-RNA <50 copies/mL. The overall geometric mean DTG Ctrough was 2330 ng/mL, and was similar between those who achieved HIV-RNA <50 copies/mL and those who did not (2420 ng/mL vs. 2120 ng/mL). A clear relationship was observed between the DTG inhibitory quotient (IQ; the ratio of DTG Ctrough to baseline susceptibility of the virus to DTG, or IC50) and the percent of subjects at week 24 with HIV-RNA < 50 copies/mL [42]. However, this relationship was strongly driven by differences in IC50, which ranged from 5110 ng/mL in the first quartile to 710 µg/mL in the 4th quartile, compared with DTG Ctrough that ranged from 1840 to 3860 ng/mL in the 1st to 4th quartiles, respectively [42].
4.2 Elvitegravir
Forty HIV-infected, ARV-naïve and experienced persons not currently on therapy received varying doses of EVG with and without RTV as monotherapy to evaluate PKPD [71]. EVG concentrations were highest when combined with RTV. EVG Ctrough were strongly associated with anti-HIV effect (log10 change in plasma HIV-RNA), where Emax was a 2.32 log10 reduction in HIV-RNA, EC50 was 14 ng/mL and EC90 was 126 ng/mL. Subsequently, 278 HIV-infected persons with HIV-RNA ≥1000 copies/mL and ≥1 PI resistance mutation were randomized to EVG or to a comparator RTV-boosted PI [72]. Three different doses of EVG, all given once daily with 100 mg of RTV were evaluated: 20mg, 50mg and 125mg. The 20 mg EVG arm was stopped at week 8 by a data safety monitoring board because that arm was associated with a higher rate of virologic failure. For the primary endpoint (change in HIV-RNA from baseline to week 24), the two remaining EVG arms (50mg and 125mg) were non-inferior to the comparator PI arm (−1.44 and −1.66 log10 reduction in HIV-RNA vs. −1.19 log10 reduction in HIV-RNA). Mean EVG Ctrough in the three arms were: 20mg, 67 ng/mL; 50mg, 211 ng/mL; and 125mg, 263 ng/mL [44]. Ctrough with the 20 mg EVG dose was less than the protein binding adjusted IC95 of 45 ng/mL, providing a reason for why this dose was subtherapeutic and a rationale to maintain concentrations above this threshold. The EVG 125mg with RTV 100mg arm achieved a statistically greater decrease in HIV-RNA at week 24 compared with the PI arm, which formed the basis for the selection of this dose (and level of systemic exposure) for evaluation in phase III studies.
Two phase III, randomized, double-blind trials compared the efficacy EVG with EFV or ATV/RTV, all given with TDF/FTC [61, 73]. EVG 150mg was given once daily with COBI 150mg. This dose combination (in a FDC with TDF and FTC) was shown to achieve similar concentrations to EVG 125mg with RTV 100mg. Combined these two studies enrolled 1408 treatment-naïve adults. In both studies, for the primary outcome of proportion of subjects with HIV-RNA <50 copies/mL, EVG/COBI was non-inferior to EFV (87.6% vs. 84.1%) and to ATV/RTV (89.5% vs. 86.8%). PD relationships were investigated in an analysis performed by the FDA, where patients were separated into deciles according to EVG Ctrough and the percentage of subjects with HIV-RNA <50 copies/mL was compared [44]. The median Ctrough in the lowest decile was 156 ng/mL and the virologic success rate was 87%. Virologic success in deciles with higher trough concentrations (234–916 ng/mL) ranged between 84–97%. This analysis indicates that virologic success was flat across the range of EVG trough concentrations achieved with the 150mg dose. A trend for different rates of virologic success was observed in participants with baseline viral load <100,000 copies/mL (92%) compared with those who had >500,000 copies/mL (79%). Collectively, these phase III trials confirm the efficacy of the 150mg once daily dose of EVG/COBI predicted by the exposure-response relationship.
4.3 Raltegravir
RAL is known to exhibit extremely high intra- and interpatient variability in plasma concentrations. For example, Ctrough in participants taking 400 mg twice daily ranged from 5.3 to 4067 ng/mL [74, 75]. This variability has clouded the ability to elucidate clear, concentration-effect relationships. A 10-day monotherapy trial in ARV-naïve persons is illustrative. 28 individuals received RAL doses of 100, 200, 400 and 600mg twice daily for 10 days [76]. Median AUC values were 2.3, 5.0, 8.2 and 7.2 µg*h/mL for the 100, 200, 400 and 600mg doses, respectively; median Ctrough were: 22, 65, 72 and 90 ng/mL, respectively. The ranges for Ctrough at the 400mg twice-daily dose were 29 to 118 ng/mL, completely encompassing the median Ctrough range across the dose range of 100 to 600mg twice daily. This interpatient variability in PK concentrations contributes to variability in virologic responses. In a phase II, randomized dose ranging study of 200mg, 400mg and 600mg of RAL twice daily, with an optimized background regimen in treatment-experienced persons, the proportions of participants with HIV-RNA <50 copies/mL at week 24 were 63%, 48%, and 56%, respectively [77]. These data illustrate that high interpatient PK variability, which contributes to the lack of dose proportionality and overlapping concentrations across a range of doses, obscures a quantitative understanding of a concentration-response relationship. The flatness of the response relationship was also observed in the large, pivotal, placebo-controlled phase III trial of RAL with optimized background therapy in persons in whom ARV therapy had failed with triple class drug resistance [75]. An exposure-response analysis conducted by the FDA found that virologic success (HIV-RNA <400 copies/mL) was similar, 77%, for patients with the lower median Ctrough of 33 ng/mL and the higher median Ctrough of 483 ng/mL [74].
The existence of an exposure-response relationship for RAL was clearly shown in a trial of once daily compared with twice-daily RAL dosing. 775 ARV-naïve persons were randomized to receive RAL 800mg once daily or 400mg twice daily, both with TDF/FTC [36]. A significantly lower proportion of participants who received RAL once daily had HIV-RNA <50 copies/mL at week 48 than did those who received 400mg twice-daily (83% vs. 89%). An exposure-response analysis found trough RAL concentrations with once daily dosing correlated with virologic response [64]. Geometric mean RAL Ctrough was 37 ng/mL for once daily dosing compared with 169 ng/mL for twice-daily dosing. A greater proportion of participants who received once daily had trough concentrations below 14 ng/mL (protein binding adjusted IC95): 42% vs. 14%. Participants who had RAL trough concentrations in the lowest quartile (median Ctrough 12.5 ng/mL) had a clear fall off in virologic response, with <80% achieving HIV-RNA <50 copies/mL. These data clearly indicate an underlying exposure-response relationship exists for RAL, and provide strong support that trough concentrations should be above the protein binding adjusted IC95 to achieve the optimal response. Additionally, they are illustrative that the shape of the concentration curve (Cmax to Cmin) can affect PD.
5. Interpretations and Conclusions
The most distinguishing PK characteristic of the INSTIs is the difference in hepatic metabolism: EVG is primarily metabolized by CYP3A4, while DTG and RAL by UGT1A1. EVG must be given with a PK enhancer (booster), usually COBI because of the availability of coformulations. As such, EVG/COBI has a higher likelihood to be a perpetrator of drug-drug interactions. The extensive clinical PK data derived from using RTV as a PK enhancer with PIs, however, largely applies to management strategies for COBI. DTG (in ARV-naïve persons) and EVG/COBI are administered once daily, while RAL requires twice-daily administration. RAL is also distinguished by a substantially higher degree of intra- and interpatient PK variability. There are differences in the PK characteristics for all three INSTIs between healthy volunteers and HIV-infected individuals, and this illustrates the importance of developing clinical PKPD data in the intended population, as discussed elsewhere [78]. Given its longer duration of clinical use, the most evidence supports standard dosing of RAL in patients with hepatic impairment (moderate through advanced cirrhosis), severe renal impairment, and during pregnancy. DTG and EVG may be used at standard doses in patients with moderate hepatic impairment. In patients with severe renal dysfunction, standard dose DTG is recommended for patients without INSTI resistance; however, DTG is not recommended for patients with INSTI resistance due to unexpectedly lower concentrations in patients with severe renal dysfunction. EVG/COBI is not recommended for patients with severe renal dysfunction, but EVG/RTV may be used without dose adjustment. Additional data are required before DTG or EVG are recommended for use in pregnant women.
In vitro IC50, 90, 95 values indicate the hierarchy of potency (most potent = lowest IC) is RAL > EVG > DTG. The EC values derived from studies in HIV-infected persons, and only available for EVG and DTG, are consistent with predictions based on in vitro potency that a lower IC value should translate into a lower EC value: the EC50, 90 for DTG > EVG. The IQ is the ratio of drug concentration in any biologic fluid (e.g., plasma, CSF) divided by an in vitro inhibitory concentration, thus integrating in vivo PK and in vitro PD (i.e. how much drug you have to how much drug you need) [79]. As discussed by the FDA, the IQ is considered important in ARV drug development because a high IQ indicates sufficient drug can be achieved, which may minimize the emergence of viral resistance, and can inform the selection of doses for phase III and IV studies, as well as for different patient populations [80]. At approved doses, the hierarchy of IQ values is DTG > EVG > RAL, and there are supporting correlates from clinical trials. DTG was shown non-inferior to RAL in ARV-naïve persons and superior to RAL in ARV-experienced but INSTI-naïve [11, 69]. These findings are consistent with DTG having a higher IQ value than RAL (17 vs. 8). For EVG, a dose of 20mg once daily (with RTV) was inferior to doses of 50 and 125mg [72]. The IQ of a 20mg dose would be approximately 1.5 compared with an IQ for the 150mg dose of 10. Once daily dosing of 800mg of RAL was shown to be inferior to 400mg twice daily [36]. The IQ for the once daily regimen (Ctrough divided by IC95) would be 2.5 compared with a value of 8 for twice daily.
In clinical studies in ARV-naïve persons, at 48-week endpoints, RAL and EVG demonstrated non-inferiority compared with EFV: 86.1% of RAL recipients vs. 81.9% of EFV achieved HIV-RNA < 50 copies/mL; 87.6% of EVG vs. 84.1% of EFV achieved < 50 copies/mL of HIV-RNA [81, 73]. DTG was shown to be statistically superior to EFV in ARV-naïve subjects at 48-weeks: 88% of DTG compared with 81% of EFV recipients had HIV-RNA < 50 copies/mL [82]. DTG was also shown to be statistically superior to DRV/RTV in ARV-naïve subjects at 48-weeks, and in the only head-to-head trial of two INSTIs in ARV-naïve persons, DTG was non-inferior to RAL (88% of DTG compared with 85% of RAL recipients had HIV-RNA at 48 weeks < 50 copies/mL) [83, 68].
Current treatment guidelines in the United States presently recommend INSTI-based regimens as among those preferred for the ARV-naïve individual [1]. There is evidence to support that the use of INSTIs has improved viral suppression rates in the United States. A study of 31,055 persons living with HIV and receiving care at eight clinical sites across the United States examined rates of viral suppression from 1997 to 2015 [84]. The percent of these individuals with undetectable viral load increased from 30% in 1997 to 87% in 2014. In multivariable analyses of participants on ART after 2010, older age, white race, male sex, better adherence, and INSTI use was associated with an undetectable viral load. These findings are concordant with the clinical PKPD profile of these agents: convenient dosing, availability in coformulations with other ARVs, high IQ values conferring high potency and rapid drops in viral load, safe, and well tolerated.
Key Points.
Integrase strand transfer inhibitors (INSTIs) represent the newest class of antiretrovirals to treat HIV infection.
The three currently approved INSTIs have unique pharmacokinetic and pharmacodynamic profiles, providing advantages for certain individuals and in special population such as those with renal and hepatic impairment or who are pregnant.
INSTIs achieve high inhibitory quotients in vivo, rapidly decrease viral load, and are safe and well tolerated.
There are emerging data to support that the use of INSTIs has improved viral suppression rates in the United States.
Acknowledgments
Funding
We acknowledge support from the following grants from the National Institutes of Health: 1R01HD085887-01A1 (to KS) and RO1 AI124965-01 and UM1AI06701 (to CVF).
Footnotes
Compliance with Ethical Standards
Conflicts of Interest
Anthony T. Podany, Kimberly K. Scarsi and Courtney V. Fletcher have no conflicts of interest that are directly relevant to the content of this review.
References
- 1.Department of Health and Human Services (DHHS) Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. Department of Health and Human Services. http://aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf. Accessed: 9 Feb 2016. 2016. http://aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf. . Accessed February 9, 2016.
- 2.Min S, Sloan L, DeJesus E, Hawkins T, McCurdy L, Song I, et al. Antiviral activity, safety, and pharmacokinetics/pharmacodynamics of dolutegravir as 10-day monotherapy in HIV-1-infected adults. Aids. 2011;25(14):1737–45. doi: 10.1097/QAD.0b013e32834a1dd9. [DOI] [PubMed] [Google Scholar]
- 3.Min S, Song I, Borland J, Chen S, Lou Y, Fujiwara T, et al. Pharmacokinetics and safety of S/GSK1349572, a next-generation HIV integrase inhibitor, in healthy volunteers. Antimicrobial agents and chemotherapy. 2010;54(1):254–8. doi: 10.1128/AAC.00842-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Tivicay® . Viiv Healthcare; Research Triangle Park, NC: Aug, 2015. [package insert] [Google Scholar]
- 5.Song I, Borland J, Chen S, Patel P, Wajima T, Peppercorn A, et al. Effect of food on the pharmacokinetics of the integrase inhibitor dolutegravir. Antimicrobial agents and chemotherapy. 2012;56(3):1627–9. doi: 10.1128/AAC.05739-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Kobayashi M, Yoshinaga T, Seki T, Wakasa-Morimoto C, Brown KW, Ferris R, et al. In vitro antiretroviral properties of S/GSK1349572, a next-generation HIV integrase inhibitor. Antimicrobial agents and chemotherapy. 2011;55(2):813–21. doi: 10.1128/AAC.01209-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Weller S, Chen S, Borland J, Savina P, Wynne B, Piscitelli SC. Bioequivalence of a dolutegravir, abacavir, and lamivudine fixed-dose combination tablet and the effect of food. JAIDS Journal of Acquired Immune Deficiency Syndromes. 2014;66(4):393–8. doi: 10.1097/QAI.0000000000000193. [DOI] [PubMed] [Google Scholar]
- 8.Reese MJ, Savina PM, Generaux GT, Tracey H, Humphreys JE, Kanaoka E, et al. In vitro investigations into the roles of drug transporters and metabolizing enzymes in the disposition and drug interactions of dolutegravir, a HIV integrase inhibitor. Drug Metabolism and Disposition. 2013;41(2):353–61. doi: 10.1124/dmd.112.048918. [DOI] [PubMed] [Google Scholar]
- 9.Castellino S, Moss L, Wagner D, Borland J, Song I, Chen S, et al. Metabolism, excretion, and mass balance of the HIV-1 integrase inhibitor dolutegravir in humans. Antimicrobial agents and chemotherapy. 2013;57(8):3536–46. doi: 10.1128/AAC.00292-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.van Lunzen J, Maggiolo F, Arribas JR, Rakhmanova A, Yeni P, Young B, et al. Once daily dolutegravir (S/GSK1349572) in combination therapy in antiretroviral-naive adults with HIV: planned interim 48 week results from SPRING-1, a dose-ranging, randomised, phase 2b trial. Lancet Infect Dis. 2012;12(2):111–8. doi: 10.1016/S1473-3099(11)70290-0. doi:10.1016/S1473-3099(11)70290-0. [DOI] [PubMed] [Google Scholar]
- 11.Raffi F, Jaeger H, Quiros-Roldan E, Albrecht H, Belonosova E, Gatell JM, et al. Once-daily dolutegravir versus twice-daily raltegravir in antiretroviral-naive adults with HIV-1 infection (SPRING-2 study): 96 week results from a randomised, double-blind, non-inferiority trial. Lancet Infect Dis. 2013;13(11):927–35. doi: 10.1016/S1473-3099(13)70257-3. doi:10.1016/S1473-3099(13)70257-3. [DOI] [PubMed] [Google Scholar]
- 12.Zhang J, Hayes S, Sadler BM, Minto I, Brandt J, Piscitelli S, et al. Population pharmacokinetics of dolutegravir in HIV-infected treatment-naive patients. British journal of clinical pharmacology. 2015;80(3):502–14. doi: 10.1111/bcp.12639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Vitekta® . Gilead Sciences, Inc.; Foster City, CA: Jul, 2015. [package insert] [Google Scholar]
- 14.Stribild® . Gilead Sciences, Inc.; Foster City, CA: Jul, 2015. [package insert] [Google Scholar]
- 15.Genvoya® . Gilead Sciences, Inc.; Foster City, CA: Nov, 2015. [package insert] [Google Scholar]
- 16.Kawaguchi I, Ishikawa T, Ishibashi M, Irie S, Kakee A, editors. Safety and pharmacokinetics of single oral dose of JTK-303/GS-9137, a novel HIV integrase inhibitor, in healthy volunteers; 13th conference on retroviruses and opportunistic infections.2006. [Google Scholar]
- 17.Kearney B, Mathias A, Zhong L, editors. Pharmacokinetics/pharmacodynamics of GS-9137 an HIV integrase inhibitor in treatment-naive and experienced patients. International Workshop on Clinical Pharmacology of HIV Therapy. 2006 [Google Scholar]
- 18.German P, Warren D, West S, Hui J, Kearney BP. Pharmacokinetics and bioavailability of an integrase and novel pharmacoenhancer-containing single-tablet fixed-dose combination regimen for the treatment of HIV. JAIDS Journal of Acquired Immune Deficiency Syndromes. 2010;55(3):323–9. doi: 10.1097/QAI.0b013e3181eb376b. [DOI] [PubMed] [Google Scholar]
- 19.DeJesus E, Berger D, Markowitz M, Cohen C, Hawkins T, Ruane P, et al. Antiviral activity, pharmacokinetics, and dose response of the HIV-1 integrase inhibitor GS-9137 (JTK-303) in treatment-naive and treatment-experienced patients. JAIDS Journal of Acquired Immune Deficiency Syndromes. 2006;43(1):1–5. doi: 10.1097/01.qai.0000233308.82860.2f. [DOI] [PubMed] [Google Scholar]
- 20.German P, Warren D, Wei L, Zhong L, Hui J, Kearney B, editors. Effect of food on pharmacokinetics of elvitegravir, emtricitabine, tenofovir DF and the pharmacoenhancer GS-9350 as a fixed-dose combination tablet; 49th Interscience Conference on Antimicrobial Agents and Chemotherapy (ICAAC); San Francisco, USA. 2009. [Google Scholar]
- 21.Shiomi M, Matsuki S, Ikeda A, Ishikawa T, Nishino N, Kimura M, et al. Effects of a protein-rich drink or a standard meal on the pharmacokinetics of elvitegravir, cobicistat, emtricitabine and tenofovir in healthy Japanese male subjects: A randomized, three-way crossover study. The Journal of Clinical Pharmacology. 2014;54(6):640–8. doi: 10.1002/jcph.283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Patel P, Song I, Borland J, Patel A, Lou Y, Chen S, et al. Pharmacokinetics of the HIV integrase inhibitor S/GSK1349572 co-administered with acid-reducing agents and multivitamins in healthy volunteers. Journal of antimicrobial chemotherapy. 2011:dkr139. doi: 10.1093/jac/dkr139. [DOI] [PubMed] [Google Scholar]
- 23.Ramanathan S, Shen G, Hinkle J, Enejosa J, Kearney B, editors. Pharmacokinetic evaluation of drug interactions with ritonavir-boosted HIV integrase inhibitor GS-9137 (elvitegravir) and acid-reducing agents; 8th International Workshop on Clinical Pharmacology of HIV Therapy.2007. [Google Scholar]
- 24.Ramanathan S, Wright M, West S, Kearney B, editors. Pharmacokinetics, metabolism, and excretion of ritonavir-boosted GS-9137 (Elvitegravir) Eighth International Workshop on Clinical Pharmacology of HIV Therapy. 2007 [Google Scholar]
- 25.Isentress® . Merck & Co., Inc. February; Whitehouse Station, NJ: 2015. [package insert] [Google Scholar]
- 26.Siccardi M, D'Avolio A, Rodriguez-Novoa S, Cuenca L, Simiele M, Baietto L, et al. Intrapatient and interpatient pharmacokinetic variability of raltegravir in the clinical setting. Therapeutic drug monitoring. 2012;34(2):232–5. doi: 10.1097/FTD.0b013e31824aa50a. [DOI] [PubMed] [Google Scholar]
- 27.Barau C, Furlan V, Yazdanpanah Y, Fagard C, Molina J-M, Taburet A-M, et al. Characterization of binding of raltegravir to plasma proteins. Antimicrobial agents and chemotherapy. 2013;57(10):5147–50. doi: 10.1128/AAC.00625-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kiser JJ, Bumpass JB, Meditz AL, Anderson PL, Bushman L, Ray M, et al. Effect of antacids on the pharmacokinetics of raltegravir in human immunodeficiency virus-seronegative volunteers. Antimicrobial agents and chemotherapy. 2010;54(12):4999–5003. doi: 10.1128/AAC.00636-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Arab-Alameddine M, Fayet-Mello A, Lubomirov R, Neely M, di Iulio J, Owen A, et al. Population pharmacokinetic analysis and pharmacogenetics of raltegravir in HIV-positive and healthy individuals. Antimicrobial agents and chemotherapy. 2012;56(6):2959–66. doi: 10.1128/AAC.05424-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Brainard DM, Friedman EJ, Jin B, Breidinger SA, Tillan MD, Wenning LA, et al. Effect of Low-, Moderate-, and High-Fat Meals on Raltegravir Pharmacokinetics. The Journal of Clinical Pharmacology. 2011;51(3):422–7. doi: 10.1177/0091270010367652. [DOI] [PubMed] [Google Scholar]
- 31.Iwamoto M, Wenning L, Petry A, Laethem M, De Smet M, Kost J, et al. Safety, tolerability, and pharmacokinetics of raltegravir after single and multiple doses in healthy subjects. Clinical Pharmacology & Therapeutics. 2008;83(2):293–9. doi: 10.1038/sj.clpt.6100281. [DOI] [PubMed] [Google Scholar]
- 32.Markowitz M, Morales-Ramirez JO, Nguyen B-Y, Kovacs CM, Steigbigel RT, Cooper DA, et al. Antiretroviral activity, pharmacokinetics, and tolerability of MK-0518, a novel inhibitor of HIV-1 integrase, dosed as monotherapy for 10 days in treatment-naive HIV-1-infected individuals. JAIDS Journal of Acquired Immune Deficiency Syndromes. 2006;43(5):509–15. doi: 10.1097/QAI.0b013e31802b4956. [DOI] [PubMed] [Google Scholar]
- 33.Petry A, Wenning L, Laethem M, De Smet M, Kost J, Merschman S, et al., editors. Safety, tolerability, and pharmacokinetics after single and multiple doses of MK-0518 in healthy subjects; 46th Interscience Conference on Antimicrobial Agents and Chemotherapy.2006. [Google Scholar]
- 34.Kassahun K, McIntosh I, Cui D, Hreniuk D, Merschman S, Lasseter K, et al. Metabolism and disposition in humans of raltegravir (MK-0518), an anti-AIDS drug targeting the human immunodeficiency virus 1 integrase enzyme. Drug Metabolism and Disposition. 2007;35(9):1657–63. doi: 10.1124/dmd.107.016196. [DOI] [PubMed] [Google Scholar]
- 35.Wenning L, Petry A, Kost J, Jin B, Breidinger S, DeLepeleire I, et al. Pharmacokinetics of raltegravir in individuals with UGT1A1 polymorphisms. Clinical Pharmacology & Therapeutics. 2009;85(6):623–7. doi: 10.1038/clpt.2009.12. [DOI] [PubMed] [Google Scholar]
- 36.Eron JJ, Jr., Rockstroh JK, Reynes J, Andrade-Villanueva J, Ramalho-Madruga JV, Bekker LG, et al. Raltegravir once daily or twice daily in previously untreated patients with HIV-1: a randomised, active-controlled, phase 3 non-inferiority trial. Lancet Infect Dis. 2011;11(12):907–15. doi: 10.1016/S1473-3099(11)70196-7. doi:10.1016/S1473-3099(11)70196-7. [DOI] [PubMed] [Google Scholar]
- 37.Rizk ML, Hang Y, Luo W-L, Su J, Zhao J, Campbell H, et al. Pharmacokinetics and pharmacodynamics of once-daily versus twice-daily raltegravir in treatment-naive HIV-infected patients. Antimicrobial agents and chemotherapy. 2012;56(6):3101–6. doi: 10.1128/AAC.06417-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Debinski HS, Lee CS, Danks JA, Mackenzie PI, Desmond PV. Localization of uridine 5'- diphosphate-glucuronosyltransferase in human liver injury. Gastroenterology. 1995;108(5):1464–9. doi: 10.1016/0016-5085(95)90695-9. [DOI] [PubMed] [Google Scholar]
- 39.Furlan V, Demirdjian S, Bourdon O, Magdalou J, Taburet AM. Glucuronidation of drugs by hepatic microsomes derived from healthy and cirrhotic human livers. J Pharmacol Exp Ther. 1999;289(2):1169–75. [PubMed] [Google Scholar]
- 40.George J, Murray M, Byth K, Farrell GC. Differential alterations of cytochrome P450 proteins in livers from patients with severe chronic liver disease. Hepatology (Baltimore, Md) 1995;21(1):120–8. [PubMed] [Google Scholar]
- 41.Food and Drug Administration Pharmacokinetics in patients with impaired hepatic function: study design, data analysis and impact on dosing and labeling. http://www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/guidances/ucm072123.pdf. Accessed 5 Mar 2016.
- 42.Food and Drug Administration (FDA) Dolutegravir. 2013 http://www.accessdata.fda.gov/drugsatfda_docs/nda/2013/204790Orig1s000ClinPharmR.pdf . Accessed February 18, 2016.
- 43.Song IH, Borland J, Savina PM, Chen S, Patel P, Wajima T, et al. Pharmacokinetics of Single-Dose Dolutegravir in HIV-Seronegative Subjects With Moderate Hepatic Impairment Compared to Healthy Matched Controls. Clinical pharmacology in drug development. 2013;2(4):342–8. doi: 10.1002/cpdd.55. doi:10.1002/cpdd.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Food and Drug Administration (FDA) Elvitegravir. 2011 http://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/203100Orig1s000ClinPharmR.pdf. Accessed February 18, 2016.
- 45.Custodio JM, Rhee M, Shen G, Ling KH, Kearney BP, Ramanathan S. Pharmacokinetics and safety of boosted elvitegravir in subjects with hepatic impairment. Antimicrob Agents Chemother. 2014;58(5):2564–9. doi: 10.1128/AAC.02180-13. doi:10.1128/aac.02180-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Iwamoto M, Hanley WD, Petry AS, Friedman EJ, Kost JT, Breidinger SA, et al. Lack of a clinically important effect of moderate hepatic insufficiency and severe renal insufficiency on raltegravir pharmacokinetics. Antimicrob Agents Chemother. 2009;53(5):1747–52. doi: 10.1128/AAC.01194-08. doi:10.1128/aac.01194-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Calza L, Danese I, Colangeli V, Manfredi R, Magistrelli E, Verucchi G, et al. Plasma concentrations of efavirenz, darunavir/ritonavir and raltegravir in HIV-HCV-coinfected patients without liver cirrhosis in comparison with HIV-monoinfected patients. Infectious diseases (London, England) 2015;47(9):625–36. doi: 10.3109/23744235.2015.1034169. doi:10.3109/23744235.2015.1034169. [DOI] [PubMed] [Google Scholar]
- 48.Hernandez-Novoa B, Moreno A, Perez-Elias MJ, Quereda C, Dronda F, Casado JL, et al. Raltegravir pharmacokinetics in HIV/HCV-coinfected patients with advanced liver cirrhosis (Child-Pugh C) The Journal of antimicrobial chemotherapy. 2014;69(2):471–5. doi: 10.1093/jac/dkt386. doi:10.1093/jac/dkt386. [DOI] [PubMed] [Google Scholar]
- 49.Barau C, Braun J, Vincent C, Haim-Boukobza S, Molina JM, Miailhes P, et al. Pharmacokinetic study of raltegravir in HIV-infected patients with end-stage liver disease: the LIVERAL-ANRS 148 study. Clin Infect Dis. 2014;59(8):1177–84. doi: 10.1093/cid/ciu515. doi:10.1093/cid/ciu515. [DOI] [PubMed] [Google Scholar]
- 50.Food and Drug Administration Pharmacokinetics in patients with impaired renal function: study design, data analysis and impact on dosing and labeling. 2010 Mar; http://www.fda.gov/downloads/Drugs/…/Guidances/UCM204959.pdf. Accessed 7 Mar 2016.
- 51.Weller S, Borland J, Chen S, Johnson M, Savina P, Wynne B, et al. Pharmacokinetics of dolutegravir in HIV-seronegative subjects with severe renal impairment. European journal of clinical pharmacology. 2014;70(1):29–35. doi: 10.1007/s00228-013-1590-9. doi:10.1007/s00228-013-1590-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Pozniak A, Arribas JR, Gathe J, Gupta SK, Post FA, Bloch M, et al. Switching to tenofovir alafenamide, coformulated with elvitegravir, cobicistat, and emtricitabine, in HIV-infected patients with renal impairment: 48 week results from a single-arm, multi-center, open-label, Phase 3 study. J Acquir Immune Defic Syndr. 2015 doi: 10.1097/QAI.0000000000000908. doi:10.1097/qai.0000000000000908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Molto J, Sanz-Moreno J, Valle M, Cedeno S, Bonal J, Bouarich H, et al. Minimal removal of raltegravir by hemodialysis in HIV-infected patients with end-stage renal disease. Antimicrob Agents Chemother. 2010;54(7):3047–8. doi: 10.1128/AAC.00363-10. doi:10.1128/AAC.00363-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Giguere P, la Porte C, Zhang G, Cameron B. Pharmacokinetics of darunavir, etravirine and raltegravir in an HIV-infected patient on haemodialysis. Aids. 2009;23(6):740–2. doi: 10.1097/QAD.0b013e328328f79d. doi:10.1097/QAD.0b013e328328f79d. [DOI] [PubMed] [Google Scholar]
- 55.Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission Recommendations for Use of Antiretroviral Drugs in Pregnant HIV-1-Infected Women for Maternal Health and Interventions to Reduce Perinatal HIV Transmission in the United States. Available at http://aidsinfo.nih.gov/contentfiles/lvguidelines/PerinatalGL.pdf. Accessed 8 Mar 2016.
- 56.Colbers A, Greupink R, Burger D. Pharmacological considerations on the use of antiretrovirals in pregnancy. Curr Opin Infect Dis. 2013;26(6):575–88. doi: 10.1097/QCO.0000000000000017. doi:10.1097/QCO.0000000000000017. [DOI] [PubMed] [Google Scholar]
- 57.Jeong H, Choi S, Song JW, Chen H, Fischer JH. Regulation of UDP-glucuronosyltransferase (UGT) 1A1 by progesterone and its impact on labetalol elimination. Xenobiotica. 2008;38(1):62–75. doi: 10.1080/00498250701744633. doi:10.1080/00498250701744633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.McCormack SA, Best BM. Protecting the fetus against HIV infection: a systematic review of placental transfer of antiretrovirals. Clin Pharmacokinet. 2014;53(11):989–1004. doi: 10.1007/s40262-014-0185-7. doi:10.1007/s40262-014-0185-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Mulligan N, Best BM, Capparelli E, Stek A, Barr E, Smith E, Chakhtoura N, Wang J, Burchett S, Mirochnick M. CROI 2016. Boston, MA: Feb 22-25, 2016. Dolutegravir pharmacokinetics in HIV-infected pregnant and postpartum women. Abstract #438. [Google Scholar]
- 60.Schalkwijk S, Colbers A, Konopnicki D, Greupink R, Russel FG, Burger D, et al. First reported use of elvitegravir and cobicistat during pregnancy. AIDS. 2016;30(5):807–8. doi: 10.1097/QAD.0000000000000976. doi:10.1097/QAD.0000000000000976. [DOI] [PubMed] [Google Scholar]
- 61.DeJesus E, Rockstroh JK, Henry K, Molina JM, Gathe J, Ramanathan S, et al. Co-formulated elvitegravir, cobicistat, emtricitabine, and tenofovir disoproxil fumarate versus ritonavir-boosted atazanavir plus co-formulated emtricitabine and tenofovir disoproxil fumarate for initial treatment of HIV-1 infection: a randomised, double-blind, phase 3, non-inferiority trial. Lancet. 2012;379(9835):2429–38. doi: 10.1016/S0140-6736(12)60918-0. doi:10.1016/S0140-6736(12)60918-0. [DOI] [PubMed] [Google Scholar]
- 62.Watts DH, Stek A, Best BM, Wang J, Capparelli EV, Cressey TR, et al. Raltegravir pharmacokinetics during pregnancy. J Acquir Immune Defic Syndr. 2014;67(4):375–81. doi: 10.1097/QAI.0000000000000318. doi:10.1097/qai.0000000000000318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Blonk MI, Colbers AP, Hidalgo-Tenorio C, Kabeya K, Weizsacker K, Haberl AE, et al. Raltegravir in HIV-1-Infected Pregnant Women: Pharmacokinetics, Safety, and Efficacy. Clin Infect Dis. 2015;61(5):809–16. doi: 10.1093/cid/civ366. doi:10.1093/cid/civ366. [DOI] [PubMed] [Google Scholar]
- 64.Rizk ML, Hang Y, Luo WL, Su J, Zhao J, Campbell H, et al. Pharmacokinetics and pharmacodynamics of once-daily versus twice-daily raltegravir in treatment-naive HIV-infected patients. Antimicrob Agents Chemother. 2012;56(6):3101–6. doi: 10.1128/AAC.06417-11. doi:10.1128/AAC.06417-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Grinsztejn B, De Castro N, Arnold V, Veloso VG, Morgado M, Pilotto JH, et al. Raltegravir for the treatment of patients co-infected with HIV and tuberculosis (ANRS 12 180 Reflate TB): a multicentre, phase 2, non-comparative, open-label, randomised trial. The Lancet infectious diseases. 2014;14(6):459–67. doi: 10.1016/S1473-3099(14)70711-X. [DOI] [PubMed] [Google Scholar]
- 66.Burger DM, Magis-Escurra C, van den Berk GE, Gelinck LB. Pharmacokinetics of double-dose raltegravir in two patients with HIV infection and tuberculosis. AIDS. 2010;24(2):328–30. doi: 10.1097/QAD.0b013e3283350f08. [DOI] [PubMed] [Google Scholar]
- 67.Min S, Sloan L, DeJesus E, Hawkins T, McCurdy L, Song I, et al. Antiviral activity, safety, and pharmacokinetics/pharmacodynamics of dolutegravir as 10-day monotherapy in HIV-1-infected adults. AIDS. 2011;25(14):1737–45. doi: 10.1097/QAD.0b013e32834a1dd9. doi:10.1097/QAD.0b013e32834a1dd9. [DOI] [PubMed] [Google Scholar]
- 68.Clotet B, Feinberg J, van Lunzen J, Khuong-Josses MA, Antinori A, Dumitru I, et al. Once-daily dolutegravir versus darunavir plus ritonavir in antiretroviral-naive adults with HIV-1 infection (FLAMINGO): 48 week results from the randomised open-label phase 3b study. Lancet. 2014;383(9936):2222–31. doi: 10.1016/S0140-6736(14)60084-2. doi:10.1016/S0140-6736(14)60084-2. [DOI] [PubMed] [Google Scholar]
- 69.Cahn P, Pozniak AL, Mingrone H, Shuldyakov A, Brites C, Andrade-Villanueva JF, et al. Dolutegravir versus raltegravir in antiretroviral-experienced, integrase-inhibitor-naive adults with HIV: week 48 results from the randomised, double-blind, non-inferiority SAILING study. Lancet. 2013;382(9893):700–8. doi: 10.1016/S0140-6736(13)61221-0. doi:10.1016/S0140-6736(13)61221-0. [DOI] [PubMed] [Google Scholar]
- 70.Castagna A, Maggiolo F, Penco G, Wright D, Mills A, Grossberg R, et al. Dolutegravir in antiretroviral-experienced patients with raltegravir- and/or elvitegravir-resistant HIV-1: 24-week results of the phase III VIKING-3 study. J Infect Dis. 2014;210(3):354–62. doi: 10.1093/infdis/jiu051. doi:10.1093/infdis/jiu051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.DeJesus E, Berger D, Markowitz M, Cohen C, Hawkins T, Ruane P, et al. Antiviral activity, pharmacokinetics, and dose response of the HIV-1 integrase inhibitor GS-9137 (JTK-303) in treatment-naive and treatment-experienced patients. J Acquir Immune Defic Syndr. 2006;43(1):1–5. doi: 10.1097/01.qai.0000233308.82860.2f. doi:10.1097/01.qai.0000233308.82860.2f. [DOI] [PubMed] [Google Scholar]
- 72.Zolopa AR, Berger DS, Lampiris H, Zhong L, Chuck SL, Enejosa JV, et al. Activity of elvitegravir, a once-daily integrase inhibitor, against resistant HIV Type 1: results of a phase 2, randomized, controlled, dose-ranging clinical trial. J Infect Dis. 2010;201(6):814–22. doi: 10.1086/650698. doi:10.1086/650698. [DOI] [PubMed] [Google Scholar]
- 73.Sax PE, DeJesus E, Mills A, Zolopa A, Cohen C, Wohl D, et al. Co-formulated elvitegravir, cobicistat, emtricitabine, and tenofovir versus co-formulated efavirenz, emtricitabine, and tenofovir for initial treatment of HIV-1 infection: a randomised, double-blind, phase 3 trial, analysis of results after 48 weeks. Lancet. 2012;379(9835):2439–48. doi: 10.1016/S0140-6736(12)60917-9. doi:10.1016/S0140-6736(12)60917-9. [DOI] [PubMed] [Google Scholar]
- 74.Food and Drug Administration (FDA) Raltegravir. 2007 Part 1. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2007/022145_ClinPharmR_P1.pdf Part 2. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2007/022145_ClinPharmR_P2.pdf. Accessed February 18, 2016.
- 75.Steigbigel RT, Cooper DA, Kumar PN, Eron JE, Schechter M, Markowitz M, et al. Raltegravir with optimized background therapy for resistant HIV-1 infection. N Engl J Med. 2008;359(4):339–54. doi: 10.1056/NEJMoa0708975. doi:10.1056/NEJMoa0708975. [DOI] [PubMed] [Google Scholar]
- 76.Markowitz M, Morales-Ramirez JO, Nguyen BY, Kovacs CM, Steigbigel RT, Cooper DA, et al. Antiretroviral activity, pharmacokinetics, and tolerability of MK-0518, a novel inhibitor of HIV-1 integrase, dosed as monotherapy for 10 days in treatment-naive HIV-1-infected individuals. J Acquir Immune Defic Syndr. 2006;43(5):509–15. doi: 10.1097/QAI.0b013e31802b4956. doi:10.1097/QAI.0b013e31802b4956. [DOI] [PubMed] [Google Scholar]
- 77.Grinsztejn B, Nguyen BY, Katlama C, Gatell JM, Lazzarin A, Vittecoq D, et al. Safety and efficacy of the HIV-1 integrase inhibitor raltegravir (MK-0518) in treatment-experienced patients with multidrug-resistant virus: a phase II randomised controlled trial. Lancet. 2007;369(9569):1261–9. doi: 10.1016/S0140-6736(07)60597-2. doi:10.1016/S0140-6736(07)60597-2. [DOI] [PubMed] [Google Scholar]
- 78.Fletcher CV. Drug interactions should be evaluated in patients. Clin Pharmacol Ther. 2010;88(5):585–7. doi: 10.1038/clpt.2010.213. doi:10.1038/clpt.2010.213. [DOI] [PubMed] [Google Scholar]
- 79.Fletcher CV. Editorial commentary: cerebrospinal fluid inhibitory quotients of antiretroviral drugs. Clin Infect Dis. 2015;60(2):318–20. doi: 10.1093/cid/ciu775. doi:10.1093/cid/ciu775. [DOI] [PubMed] [Google Scholar]
- 80.Food and Drug Administration (FDA) Guidance for Industry, Antiviral Product Development, Conducting and Submitting Virology Studies to the Agency. 2006 http://www.fda.gov/OHRMS/DOCKETS/98fr/05d-0183-gdl0002-01.pdf. Accessed February 18, 2016.
- 81.Lennox JL, DeJesus E, Lazzarin A, Pollard RB, Madruga JV, Berger DS, et al. Safety and efficacy of raltegravir-based versus efavirenz-based combination therapy in treatment-naive patients with HIV-1 infection: a multicentre, double-blind randomised controlled trial. Lancet. 2009;374(9692):796–806. doi: 10.1016/S0140-6736(09)60918-1. doi:10.1016/S0140-6736(09)60918-1. [DOI] [PubMed] [Google Scholar]
- 82.Walmsley SL, Antela A, Clumeck N, Duiculescu D, Eberhard A, Gutierrez F, et al. Dolutegravir plus abacavir-lamivudine for the treatment of HIV-1 infection. N Engl J Med. 2013;369(19):1807–18. doi: 10.1056/NEJMoa1215541. doi:10.1056/NEJMoa1215541. [DOI] [PubMed] [Google Scholar]
- 83.Raffi F, Rachlis A, Stellbrink HJ, Hardy WD, Torti C, Orkin C, et al. Once-daily dolutegravir versus raltegravir in antiretroviral-naive adults with HIV-1 infection: 48 week results from the randomised, double-blind, non-inferiority SPRING-2 study. Lancet. 2013;381(9868):735–43. doi: 10.1016/S0140-6736(12)61853-4. doi:10.1016/S0140-6736(12)61853-4. [DOI] [PubMed] [Google Scholar]
- 84.Simoni JM, Nance RM, Delaney JAC, Wilson I, Aunon F, Safren SA, et al. Conference on Retroviruses and Opportunistic Infections (CROI) Boston: Feb 22, 2016. HIV viral load in US clinics over time: trends and predictions from CNICS. MA2016. [Google Scholar]