Abstract
The human immunodeficiency virus (HIV) causes acquired immumodeficiency syndrome (AIDS), one of the worst global pandemic. The virus infects human CD4 T cells and macrophages, and causes CD4 depletion. HIV enters target cells through the binding of the viral envelope glycoprotein to CD4 and the chemokine coreceptor, CXCR4 or CCR5. In particular, the CCR5-utilizing viruses predominate in the blood during the disease course. CCR5 is expressed on the surface of various immune cells including macrophages, monocytes, microglia, dendric cells, and active memory CD4 T cells. In the human population, the CCR5 genomic mutation, CCR5Δ32, is associated with relative resistance to HIV. These findings paved the way for the discovery and development of CCR5 inhibitors to block HIV transmission and replication. Maraviroc, discovered as a CCR5 antagonist, is the only CCR5 inhibitor that has been approved by both US FDA and the European Medicines Agency (EMA) for treating HIV/AIDS patients. In this review, we summarize the medicinal chemistry and clinical studies of Maraviroc.
Keywords: Antiretroviral therapy, Clinical study, HIV-1, HIV-2, Medicinal chemistry, Microbicide, SAR
Introduction
The human immunodeficiency virus (HIV) infection leads to acquired immumodeficiency syndrome (AIDS), which afflicts an estimated 35.5 million people globally. HIV enters the host cells through the fusion of the lipid membrane of the virus with that of the host cells. The fusion is triggered by the interaction of the viral surface protein (gp120) with specific host cell surface receptors, CD4 and the chemokine coreceptor CXCR4 or CCR5 [1–9].
The importance of the chemokine coreceptors in HIV infection and pathogenesis has been demonstrated by the findings that a mutant allele of the CCR5 gene, CCR5Δ32, confers relative resistance to HIV infection [10]. CCR5 is expressed on the surface of various immune cells including macrophages, monocytes, microglia, dendric cells, active memory CD4 T cells. Given the natural existence of CCR5 deletion in the human population, it is feasible to target CCR5 to block HIV infection. Therefore, the development of CCR5 antagonists has become a high research priority in recent years.
Maraviroc is a CCR5 antagonist that inhibits the entry of HIV into host cells by blocking gp120 interaction with CCR5 [11]. Currently, Maraviroc is the only CCR5 inhibitor that has been approved by both US FDA and the European Medicines Agency (EMA) for the treatment of antiretroviral drug-experienced and -naive patients [12, 13]. The approval of Maraviroc for clinical use was based on the Maraviroc plus Optimized Therapy in Viremic Antiretroviral Treatment Experienced patients (MOTIVATE) 1 and MOTIVATE 2 studies [14]. In this review, we mainly focus on the medicinal chemistry of Maraviroc and its recent clinical applications in combination with other antiretroviral drugs. We also review the development of Maraviroc as an anti-HIV microbicide.
1. Medicinal Chemistry of Maraviroc
Maraviroc (Figure 1) was discovered in 2005 by Pfizer global research and development team. It was identified through the combination of high-throughput screen (HTS), high throughput binding assay, and medicinal chemistry [15].
Figure 1.
Structure of Maraviroc
It took approximately two and a half years of development from the initial high throughput screening, during which 965 analogues were synthesized through medicinal chemistry. Maraviroc was finally identified with the desirable antiviral activity, metabolic stability, absorption, and hERG inhibition. It took another two more years of clinical studies to have Maraviroc approved by FDA for the treatment of HIV/AIDS. Currently, research is still ongoing to develop Maraviroc-related analogues with improved pharmacokinetics. In the following section, we review the medicinal chemistry in the discovery of Maraviroc and Maraviroc-related analogues.
1.1 Discovery of Maraviroc
In the discovery of Maraviroc, nearly 1,000 analogues were synthesized, from which Maraviroc emerged as the best drug candidate [16, 17]. Several important steps leading to the identification of Maraviroc have been taken:
1.1.1 Identification of the hits and the lead compounds
A chemokine radioligand-binding assay was first used to screen the Pfizer compound library to identify small-molecule CCR5 ligands. A number of hits were identified from the HTS and then triaged. Four hits remained in the pool after taking into account of target affinity, ligand efficiency, and toxicity. Then, two of them, the imidazopyridine 1 (UK-107,543) (MIP-1β IC50 0.4 μM) and 2 (UK-179,645) (MIP-1β IC50 1.1 μM) (Figure 2), were chosen as the start points of an intensive medicinal chemistry program for further optimization [15].
Figure 2.

Compound 1 & 2 Structures
However, neither of the two hits could be considered as the lead compound because of the lack of desirable properties such as a low molecular weight, high binding affinity, and potent antiviral activity. Therefore, hit-to-lead studies were taken as the second step.
To develop the lead compound, the most attractive features of the two hits were combined to produce novel and selective antagonists, in which compound 3 became the focus of the structure-activity relationship (SAR) investigations. Compound 3 exhibited good chemokine receptor binding (MIP-1β IC50 45 nM) and antiviral activity. SAR studies of different amide substituents were firstly started. SAR suggested that the amide substituent interacts with a predominantly lipophilic binding site in the CCR5 receptor, and the cyclobutyl amide analogue 4 was the most potent one in this series (Table 1). Further studies established the (s) enantiomer of cyclobutyl amide (s)-4 was the active isomer. Then, starting from compound (s)-4, by keeping the cyclobutyl substituent constant, a series of piperidine analogues were generated, among which, the tropane derivatives 5 and 6 (exo and endo isomers) were found to be highly potent inhibitors of viral replication. The piperidine ring in 6 was forced into a boat conformation, the resulting structure overlaps well with that of the exo substituted analogues. Compound 5 possessed the enhanced binding affinity and improved antiviral activity than the two hit compounds, and it was also a potent ligand for the hERG channel, displaying 80% inhibition of the binding of tritiated dofetilid when screened at 300 nM. The endo isomer 6 was essentially equipotent to 5. They were chosen as lead compounds for further optimization.
Table 3.
Cell fusion and antiviral activities for maraviroc and related analogues 9–12.
| ||
|---|---|---|
| Compound | Fusion IC50a | AV IC90b |
| Maraviroc | 0.2 nM | 0.7 nM |
| 9 | 7.5 nM | Inactive |
| 10 | 14 nM | _ |
| 11 | 48 nM | 310 nM |
| 12 | 5 nM | _ |
Cell fusion assay
Antiviral activity-concentration required to inhibit replication of HIVBal in PM-1 cells by 90%
1.1.2 Lead Optimizations
In the development of Maraviroc, Price et al focused on hERG liability and used the [3H]-dofetilide binding assay to enable critical SAR analysis [16, 17]. They started with the lead compound 5, which was a potent ligand for the CCR5 receptor and also a potent ligand for the HERG channel. The study on the compound 7 with the oxygen bridgehead having a profound effect on the basicity of the nitrogen demonstrated that the change of the basicity/steric environment of the central nitrogen atom has had little effects on the compounds’ affinity for the hERG channel (Table 2). So they decided to focus on the optimization of benzimidazole moiety [16, 17].
Table 4.
Activities of compounds 13 and 14.
| ||||
|---|---|---|---|---|
| Compound | Fusion IC50 nM | logD7.4 | HLM (μl/min/mg of microsomal protein) | hERG IC50, nM |
| 13 | 37.5 | 1.2 | <7 | 849 |
| 14 | 2.0 | 1.9 | <7 | 4000 |
Docking of compound 5 into the model of hERG channel indicates that the phenyl group of the benzimidazole overlaps perfectly with a lipophilic binding area, which suggested alternative expressions for the benzimidazole would reduce affinity for the hERG channel. Complete replacement of the benzimidazole rather than modification was chosen as the best strategy. Compound 8 was found to have good levels of antiviral potency but with some increased affinity for the hERG channel compared to compound 5. It was found that the affinity for the hERG channel had started to return with increased lipophilicity in this series, and the triazole series showed a superior profile to the benzimidazole [16, 17].
Based on the model of the hERG channel, modification of the cyclobutyl amide turned to reduce the affinity for the channel. After parallel chemistry in the final step amide coupling, this work was rapidly performed, and within the triazole series, the 4,4-fifluorocyclohexyl group showed no activity for the hERG channel due to the steric demands of the cyclohexyl group and the dipole generated by the difluoro moiety. Eventually, Maraviroc was discovered with potent antiviral profile and lack of affinity for the hERG channel.
Maraviroc possesses excellent antiviral potency, reasonable microsomal stability, high selectivity over potential ion channel effects, good aqueous solubility, and no significant binding to the HERG potassium channel at 1,000 nM.
To summarize, rapid ability to generate high quality data in a well validated assay and excellent pharmacophore models of the hERG channel eventually led to the discovery of Maraviroc [16, 17].
1.2 Maraviroc-related analogues
Maraviroc contains a tertiary amine (tropane), two hydrophobic groups, and one heteroaryl group, and these four pharmacophore elements are found in most CCR5 antaganists.
1.2.1 Tertiary amine (tropane) analogues
The tropane bridge often gave a potent activity in cell-based gp160 fusion assay, and was proved beneficial for antiviral activities due to steric hindrance around the central basic center and restrained conformational flexibility of the molecule [15]. For example, the antiviral activity was lost when the bridge was removed from Maraviroc to give 9 [18]. (Table 3)
Table 5.
RANTES binding inhibition and antiviral activity of exo- 15(a–d), endo- 15(a–d), and 16(a–d).
| Compound | Binding (nM)a | Antiviral (nM)b |
|---|---|---|
| Maraviroc | 1.4 | 2.8 |
| Exo-15a | 35 | 40 |
| Exo-15b | 6 | 200 |
| Exo-15c | 43 | ≥625 |
| Exo-15d | _ | 81 |
| Endo-15a | 18 | 210 |
| Endo-15b | 6 | 74 |
| Endo-15c | 30 | ≥625 |
| Endo-15d | 8 | 41 |
| 16a | 7 | 62 |
| 16b | 5 | 24 |
| 16c | 13 | 220 |
| 16d | 5 | 5 |
Binding inhibition (IC50) of [128I]-RANTES to CCR5-expressing CHO cells.
Replication inhibition (IC50) of R5 HIVNLBal in JC53-BL cells.
Barber et al. proposed that similar steric hindrance and conformational effects could also be achieved through the addition of an alpha methyl branch onto the 3-amino-3-phenyl-propyl side chain. For example, removal of the tropane bridge and the ethyl substituent from compound 10 to give 11 resulted in a loss of fourfold in potency. However, the alpha-methyl derivative 12 increased the potency about threefold over the equivalent tropane 10 [18] (Table 3). With further exploration and SAR studies on the 4-position of piperidine of 12, replacement of the tropane core of maraviroc by piperidine with a branched N-substituent yielded a new class of CCR5 antagonist [18], among which compound 13 and 14 with 1,3,4-triazoles showed good whole cell antiviral activity, metabolic stability, and in the case of 13, only weak activity at the hERG ion channel (Table 4). Further investigation is ongoing for this series of compounds.
Table 6.
IC50 values for compounds 17–20 in a MIP 1-β binding assay.
| |
|---|---|
| Compound | IC50 (μM) |
| 17 | 0.97 |
| 18 | 0.31 |
| 19 | 0.031 |
| 20 | 0.004 |
Lemoine et al [19] evaluated the effects of replacement of 3-amino-8-azabicyclo [3.2.1] octane in Maraviroc. The tertiary amine as part of the rigid 8-azabicyclo [3.2.1] octane bicyclic system was replaced with the 5-amino-3-azabicyclo[3.3.0] octane. The pKa values of the tertiary amine of the 4-substituted dimethyl 1,2,4-triazole derived from 3-aminotropane, and that derived from the bicyclic exo or endo 5-amino-3-azabicyclo[3.3.0] octanes were calculated to be virtually the same (9.47 ± 0.48, 9.42 ± 0.48, respectively), and the only difference between the two ring systems would be structural and conformational [19]. Then, a series of compound 15 in both sub-series (exo or endo) were prepared to study the replacement effects, by comparing to the series of compound 16 with the tropane core (Figure 3).
Figure 3.
Structures of Compounds 15 and 16.
It was concluded that the bicyclic exo and endo 5-amino-3-azabicyclo[3.3.0] octanes were effective but not equal replacements for the conformationally restricted tropane in Maraviroc (Table 5) [19].
Table 7.
Viral entry inhibition of compounds 20–24, IC50 (μM).
| |||||
|---|---|---|---|---|---|
| Compound | Virus | ||||
| JRCSF | ASM80 | Ba-L | 97-ZA-003 | RU570 | |
| 20 | 0.118 | 0.058 | 0.048 | 0.029 | 0.028 |
| Maraviroc | 0.005 | 0.006 | 0.004 | 0.008 | 0.002 |
| 21 | 0.009 | 0.016 | 0.028 | 0.009 | 0.005 |
| 22 | 0.009 | 0.011 | 0.062 | 0.017 | 0.007 |
| 23 | 0.015 | 0.120 | 1.048 | 0.116 | 0.090 |
| 24 | 0.003 | 0.005 | 0.008 | 0.005 | 0.002 |
More SAR exploration would be needed for further optimization of both the endo and the exo-sub-series, however no other compounds were reported in this new series, likely because of the decreased potency in comparison with Maraviroc.
1.2.2 Tropane core retained analogues
1.2.2.1 N-ethyl spacer analogues
Ernst and coworkers identified compound 17, which contained the attractive features of reported templates, including those from maraviroc [20]. From the starting compound 17 (IC50 = 0.97 μM), the N-ethyl spacer was elongated by one carbon to give a compound 18 which showed three fold higher potency (IC50 = 0.31 μM). Modification of the cyclohexylamide moiety of 18 to a urea resulted in compound 19 with a 10-fold increase in potency (Table 6).
Then, the SAR studies of compound 19, containing two different features, elongated N-ethyl spacer and urea functionality instead of amide functionality compared to Maraviroc, were done through introduction of different urea derivatives, modifications of the triazole moiety, and introduction of fluorine atom to the phenyl ring. Representative compounds are summarized in Table 6 and 7. The data showed the para-substituted phenyl urea group gave high potency in chemokine binding assay, and the series of compounds were represented by compound 20 (Table 6). Incorporation of a polar side chain to triazole moiety in an appropriate orientation improved the viral entry inhibition potency, and compound 21 possessed potent antiviral activity after triazole was replaced with imidazole. Meta-Fluoro-substitution at the phenyl ring of 22 appeared to be tolerated to the viral entry inhibition, however, para-fluoro-substituted analogue 23 just retained one tenth of potency compared to compound 21. With theses modifications, a series of substituted imidazopiperidine-tropane CCR5 antagonists were yielded, and the antiviral properties were optimized through a luciferase-reporter phenotypic viral entry assay [21]. Compound 24 was identified with attractive pharmocokinetic, selectivity, and antiviral properties. Further studies with 24 are ongoing [20].
1.2.2.2 Hydrophobic group analogues
Through robust chemical approaches toward putative CCR5 antagonist scaffolds and evaluation of analogues in the125I-[MIP-1β] binding and Ba-L-Hos antiviral assays, a serie of 4,4-disubstituted pireridine-based CCR5 antagonists were discovered [22, 23]. Among them, compound 25 and 26 showed high potency and bioavailability (Table 8), which were used as lead molecules for further optimization. Through hERG and pharmacokinetics SAR studies, a druglike compound 27 was obtained with improved potency and bioavilability [24].
Table 8.
Activities of compounds 25–27.
| ||||||
|---|---|---|---|---|---|---|
| Compound | pIC50 CCR5 binding [MIP-1β] | Ba-L-Hos pIC50 | hERG patch clamp pIC50 | QTc (%) | RAT PO DNAUC [ng.h/mL] | DOG PO DNAUC [ng.h/mL] |
| 25 | 7.8 | 7.80 | 5.7 | 9.4 | 16 | - |
| 26 | 9.45 | 8.01 | 6.0 | 6.5 | 144 | - |
| 27 | 8.37 | 8.46 | 4.72 | 1.2 | 272 | 170 |
By replacing the piperidine ring of 4,4-disubstituted pireridine-based CCR5 antagonists with cyclohexylamine (Figure 4), a series of 4,4-disubstituted cyclohexylamine based CCR5 antagonists were developed.
Figure 4.

4,4-Disubstituted cyclohexylamine based CCR5 antagonists SAR Study.
SAR studies were done on the linker and amide moiety indicated in Figure. A representative compound 28 (Figure 5) was obtained with pronounced activity against HIV-1 (HOS IC50 = 58 nM) [25].
Figure 5.

Structure of Compound 28
1.2.2.3 Heteroaryl analogues
Maraviroc is dosed b.i.d. clinically and has moderate bioavailability in man with food effects on oral exposure [26, 27]. To address this, a new series of imidazopiperidine CCR5 antagonists were developed, and they retained the antiviral profile and hERG activity of Maraviroc [28]. Moreover, they possessed improved absorption profiles in rat and dog. In the development of the new series of CCR5 antagonists, the medicinal chemistry started with lead compound 29 (Figure 6). As a lead compound, 29 had several important features. For examples, the acetyl of the amide moiety of 29 minimizes the lipophilic interactions to a lipophilic region of the hERG pharmacophore, and therefore reduced affinity to the hERG channel; the tropane core, retained the same as Maraviroc, ensured excellent antiviral activity and overcame problems such as CYP inhibition [11]; the benzimidazole retained compound 29 with potent activity in the gp160 fusion assay and moderate activity in a hERG binding assay. Similar to the SAR study strategies used by Ernest et al, the benzimidazole moiety of 29 was converted to imidazolepiperidine for the increased polarity, the fluorine atom was introduced to the meta position of phenyl ring for the improved Caco-2 profile. Then, a serie of imidazolepiperidine and isomeric imidazolepiperidine CCR5 antagonists were developed, among which 30 (PF-232798) was screened with complete oral absorption in rat and dog and demonstrated improvement for in vitro metabolic stability in comparison with Maraviroc (Table 9). Compound 30 has shown improvement for in vitro metabolic stability and antiviral activity against HIV BaL in PBMCs in comparison with Maraviroc. Furthermore, compound 30 retained antiviral activity against a Maraviroc-resistant HIV-1 strain. Thus, compound 30 was chosen for further clinical studies, based on a favorable profile in preclinical safety studies. Phase II clinical studies of compound 30 are ongoing [28].
Figure 6.

Structures of Compound 29& 30
Table 9.
Profiles of 30 and Maraviroc
| Compound | Antiviral Activity | Pharmacokinetic property | Human, Rat, Dog Liver microsome Assays | hERG channel activity | Permeability | ||||
|---|---|---|---|---|---|---|---|---|---|
| Gp 160 IC50 (nM) | AV IC90 (nM) | Rat absorption | Dog absorption | HLM Clint | RLM Clint | DLM Clint | Caco-2 AB/BA | ||
| Maraviroc | 0.2 | 3.2 | 20 | 70 | 49 | < 6 | < 9 | 0% @ 300 nM | < 1/12 |
| 30 | < 0.1 | 2.0 | complete | complete | 7 | < 6 | < 9 | IC50 12 μM | 2/8 |
1.2.2.4 Maraviroc-hybrid molecules
A serial of small molecule CCR5 antagonists were designed and synthesized through fragment assembly, based on the structures of Maraviroc and the compound 31 (Figure 7), which was identified through structure-based virtual screening. Representative Compound 32 (Figure 7) had an IC50 value of 0.233 μM [29]. Preliminary SARs showed that the 4,4-difluorocyclohexanecarbonyl is important to maintain the antagonistic activity, and the activity could be significantly different with different heterocyclic aryl groups. Although this serie of compounds did not show more potent activity compared with Maraviroc, the novel scaffold could be applied in the development of Maraviroc-derived second generation of CCR5 antagonists [29].
Figure 7.

Structures of Compound 31 & 32
To summarize, among those Maraviroc-related analogues, compound 30 (PF-232798) is under clinical phase II study, and 27 (GSK 163929) has completed with the preclinical characterization, and further progression to the clinic is ongoing. The SAR in the development of Maraviroc and its analogues may provide useful information in the discovery of other drugs for the treatment of HIV.
2. Clinical studies
Maraviroc blocks the binding of gp120 to CCR5 receptor selectively and reversibly. This interaction prevents the conformational changes necessary for the M-tropic HIV entry into host cells [15]. Maraviroc has been a drug to treat HIV/AIDS in the market for several years, and the clinical studies have been continuing, and had been reviewed previously [13, 30–33]. Here, we mainly focus on recent clinical applications of Maraviroc in combination with other antiretroviral drugs and as an anti-HIV microbicide.
2.1 Determination of HIV tropism
Maraviroc is effective to prevent the R5-tropic viruses to enter the cell, however, X4 or dual/mixed populations are unaffected by the drug. It is necessary to determinate the viral tropism prior to initiating therapy.
Before prescribing Maraviroc, it is mandatory to determine viral tropism. In current clinical practice, Trofile® and its new version (ES-Trofile®) assays are predominantly performed. Genotypic methods are developing in Europe as an alternative to ES-Trofile®[34]. Maraviroc clinical test (MCT) is a clinical approach to determine the treatment with Maraviroc. MCT could be considered as an additional method before CCR5-antagonist prescription, since the rate of the virological success increased in a combined antiviral therapy after MCT [35].
2.2 Pharmacokinetics
The pharmacokinetics of Maraviroc was evaluated when administered at 150 mg once daily in combination with lopinavir/ritonavir in HIV-positive treatment-naive patients [36]. Lopinavir and ritonavir exposure were not affected by the coadministration of Maraviroc, and all PK values were comparable to what was previously reported. The study showed that 150 mg dosing of Maraviroc once daily compared to twice daily seems to be pharmacologically enough [36].
The pharmacokinetic profile of a novel nucleoside-sparing combination antiretroviral regimen, which is an attractive option for the treatment of HIV infection, was established by Mora-Peris and the coworkers [37]. Eligible participants were the HIV–infected subjects on stable once-daily antiretroviral therapy receiving 245/200 mg of tenofovir/emtricitabine plus 800/100 mg of darunavir/ritonavir. Maraviroc once daily was added to the current combination antiretroviral therapy during study period 1 (days 1–10), tenofavir/emtricitabine was discontinued in study period 2 (days 11–20). No clinical safety concerns were observed, and it was found that Maraviroc exposure only depends on ritonavir exposure.
2.3 Clinical efficacy and safety
The efficacy of Maraviroc in treating HIV-1 infection was evaluated by a parallel, randomized, two double-blind, and placebo-controlled Phase III studies: MOTIVATE 1 and 2 [14]. MOTIVATE 1 was conducted in Canada and the USA, whereas MOTIVATE 2 was conducted in Australia, Europe and the USA. In this study, 1049 patients were received Maraviroc doses equivalent to 300 mg once daily, 300 mg twice daily, or placebo in a 2:2:1 ratio in addition to optimized background therapy (OBT). The duration of the trial was 48 weeks.
Maraviroc treatment had a significantly better virological and immunological response at week 48 compared with placebo. There is an approximately 1 log10 decrease in HIV-1 RNA levels from baseline in Maraviroc plus OBT group as compared with OBT alone (p<0.001). Viral load was undetectable (<50 copies/mL) in participants treated with Maraviroc once or twice daily compared with patients receiving placebo (p<0.001). The CD4 cell count was significantly higher in the Maraviroc once daily (116 cell/μL) and Maraviroc twice (124 cells/μL) than in the placebo group (61 cells/μL) (p<0.001) [14].
The efficacy of Maraviroc was also studied in patients who were infected with dual/mixed X 4 viruses. In this study, Maraviroc did not show significant benefit on the virologic outcomes, although the CD4 T cell count was slightly increased in Maraviro plus OBT arm [38]. In other words, Maraciroc has no benefit to those patients with dual/mixed-trpic virus infection.
The efficacy and tolerability of Maraviroc as part of a variety of cART regimens have been well studied in TE patients with R5 HIV-1 infection in two Phase 2b/3 studies [14, 39, 40]]. Additional safety and immunovirologic ativity data of Maraviroc in regimens were obtained after newer agents such as darunavir, raltegravir, and etravirine were clinically available [41]. It was found that Maraviroc was well tolerated over 96 weeks as a combination with different agents, and treatment with Maraviroc in combination with OBT was associated with significantly greater immunovirologic activity versus placebo plus OBT [40, 41].
The immunologic effectiveness of Maraviroc-and raltegravir-containing regimens (R+M+) versus raltegravir-based regimens containing no Maraviroc (R+M−) were compared, and no difference was observed in terms of the rate of change in CD4 count over 24 months of follow-up [42]. The results contribute to the knowledge regarding the optimal use of Maraviroc in the treatment of HIV-infected patients from an immunological perspective [42].
Maraviroc and raltegravir are new antiretroviral drugs for treating HIV infection, and their uses are important in patients with intolerance or resistance to other antiretroviral agents. The correlation was evaluated between a short therapeutic regimen (raltegravir, Maraviroc, and a protease inhibitor fosamprenavir) and liver diseases. The serum creatine kinase level increased overwhelmingly during the treatment. However, no information is available regarding this correlation [43].
Salvage therapy with raltegravir, Maraviroc and etravirine regimen showed excellent CD4 recovery and HIV-RNA suppression at 24 weeks. This combination regimen treatment is safe and efficacious in the management of drug-experienced patients [43].
Overall, the safety profile of Maraviroc in clinical trials was acceptable. There were no significant differences in liver functional abnormalities between the study arms in the MOTIVATE trials. The most common adverse events, such as diarrhea, fatigue, fever, headache, nausea and upper respiratory, were not found in the Maraviroc arms.
2.4 Resistance
There are two possible ways in which HIV-1 can escape the effect of Maraviroc: X4 variants bening already in the virus population that could result in a HIV coreceptor switch; viral R5 tropism mutations that permit CCR5 binding despite the presence of Maraviroc [44, 45]. The main mechanism of Maraviroc resistance was found to be that the resistant virus can use Maraviroc-bound CCR5 receptor as a result of selection of multiple mutations in the V3 loop of gp120 [46]. Given that Maraviroc-resistant HIV-1 can bind to CCR5 in both the normal conformation and the maraviroc-bound conformation, increasing the dose of Maraviroc does not increase the inhibition of HIV. This intrinsic viral resistance to the drug is uncommon, and it is not easy to give a recommendation in clinical trials for characterizing Maraviroc resistance. Thus, testing viral tropism is important when Maraviroc is considered as an alternate drug in anti-HIV treatment.
2.5 Applications in Combination Therapy
Maraviroc was studied to substitute the third drug of antiretroviral treatment in aviraemic subjects infected with R5 HIV, and it was found switching the third drug to Maraviroc was safe, efficacious, and with improved lipid parameters [47].
Maraviroc was used in combination with tacrolimus in the setting of post-hepatic transplantation. Although it was just one clinical application, there were emerging data supporting the potential use of Maraviroc for prolonging graft survival [48].
A study in mice showed Maraviroc could reduce the antherosclerotic progression, and reversed the proinflammatory profile in mice with ritonavir-induced inflammation [49]. Although large clinical studies are needed, it is fair to say Maraviroc could benefit HIV-positive patients with residual chronic inflammation [49].
Diaz-Delfin et al. studied the effects of Maroviroc during and after the differentiation of human adipocytes in culture, and they concluded that Maraviroc did not alter adipocyte differentiation, but showed anti-inflammatory properties, which suggested that Maraviroc could be beneficial by minimizing adverse effects on adipose tissue development, metabolism, and inflammation [50].
2.6 Interaction between alcohol and Maraviroc
Many HIV patients think they should not take medication when they drink alcohol. However, resistance could be developed after frequently missed medication. The study from Gruber et al showed that alcohol did not affect Maraviroc pharmacokinetics, so patients should not miss Maraviroc treatment even though alcohol is consumed [51].
2.7 Studies in HIV-2 Treatment
HIV-2 infection undergoes a long asymptomatic phase and AIDS development. Maraviroc is active in vitro against CCR5-tropic HIV-2 [52, 53]. Visseaux and coworkers the first time assessed that Maraviroc is active in vitro against R5 HIV-2 clinical isolates through a phonotypic peripheral blood mononuclear cell-based test [52]. Borrego and coworkers studied the susceptibility of primary HIV-2 to Maraviroc along with entry inhibitors. They found reduced sensitivity of R5 variants to Maraviroc, indicating that higher dosages of Maraviroc might be needed for the treatment of HIV-2 than HIV-1, and the treatment should be adjusted to the HIV-2 disease courses. There is a growing interest in using Maraviroc to treat HIV-2-infected patients. Maraviroc was combined with foscarnet as a salvage therapy in HIV-2-infected patients with antiretroviral treatment failure [54]. Armstrong-James et al reported that an HIV-2 patient responded positively to a raltegravir- and Maraviroc-based theraphy, while resistant to protease inhibitor, nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs) and nonnucleoside reverse transcriptase inhibitors (NNRTIs) [55]. Caixas et al described a long-term successful control of one HIV-2-infected patient using Maraviroc [56].
2.8 Application in Microbicide Development
An effective vaginal microbicide can reduce HIV-1 transmission to women. Maraviroc is a highly potential microbicide candidate in clinical development. Veazey et al firstly tested the ability of Maraviroc as a vaginal microbicide to prevent transmission using a stringent model that involves challenge of rhesus macaques with a high dose of SHIV-162P3, a CCR5-using virus [57]. Maraviroc provided dose- and time-dependent protection against challenge with SHIV-162P3. The pharmacokinetics and efficacy were assessed, and a high degree of correlation between PK and efficacy was observed [58]. Later, Neff et al. tested Maraviroc as microbicide in RAG-hu humanized mouse model [59]. Female RAG-hu mice were challenged vaginally with HIV-1 after intravaginal application of the maraviroc gel. It was found that Maraviroc gel treated mice were fully protected against HIV-1 challenge, while the placebo gel treated mice all became infected [59]. Their findings demonstrated Maraviroc as a candidate in the development of vaginal microbicides [57, 59].
Malcolm et al. [60] described the sustained release of Maraviroc from matrix-type silicone elastomer vaginal rings after tests in aqueous gel formulations [58]. Their founding could help design macaque challenge experiments and ring performance during human female menstrual cycle [60].
Fetherston et al. reported a combination microbicide formulation containing both dapivirine and Maraviroc, in the form of a silicone elastomer vaginal ring [61]. A silicone elastomer matrix-type ring vaginal formulation containing 25 mg dapivirinr and 100 mg Maraviroc has been developed and evaluated in Phase I clinical trial [61]. Maraviroc, combined with dendrimers, showed synergistic profile against CCR5 and dual tropic HIV-1 [62]. Vaccines and microbicides containing Maraviroc may protect better when used together than separately [63]. These evaluations supported the development of combinatorial microbicides to fight against HIV spread.
In Summary, Maroviroc is a potent new antiretroviral drug that has no cross-resistance with other drugs. It is also a valuable drug additional to the current drugs available in the combination against HIV.
Conclusions
Maraviroc is the only HIV-1 CCR5-based entry inhibitor to date approved by FDA. The study of its clinical benefits is still continuing. In this article, we reviewed the medicinal chemistry and clinical applications of Maraviroc as an anti-HIV drug. We also reviewed the development of Maraviroc as a microbicide. Ongoing research and development of Maraviroc-derivatives have showed improved potency and pharmocokinetics over Maraviroc, suggesting that similar class of CCR5 inhibitors will likely in clinical treatment of HIV infection in the near future.
Table 1.
MIP-1β inhibitory activity and antiviral activity of compound 3–6.
| ||
|---|---|---|
| Compound | MIP-1βIC50 (nM) | Antiviral activity |
| 3 | 45 | 210 nM IC50 |
| 4 | 40 | 75 nM IC50 |
| (s)-4 | 20 | 73 nM IC50 |
| 5 | 2 | 13 nM IC90 |
| 6 | 6 | 3 nM IC90 |
Table 2.
Antiviral activity and hERG channel activity of compound 7, 8 and Maraviroc.
| ||
|---|---|---|
| Compound | Antiviral IC90 (nM) | hERG channel inhibition |
| 7 | 39 | 70% @ 300 nM |
| 8 | 8 | 30% @ 300 nM |
| Maraviroc | 2 | 0% @ 300 nM |
Acknowledgments
This work is supported in part by US Public Health Service grant 1R01MH102144 from NIMH to Y. W.
Footnotes
Conflict of Interest:
Declared none.
References
- 1.Klatzmann D, Champagne E, Chamaret S, Gruest J, Guetard D, Hercend T, Gluckman JC, Montagnier L. T-lymphocyte T4 molecule behaves as the receptor for human retrovirus LAV. Nature. 1984;312(5996):767–768. doi: 10.1038/312767a0. [DOI] [PubMed] [Google Scholar]
- 2.Ashorn PA, Berger EA, Moss B. Human immunodeficiency virus envelope glycoprotein/CD4-mediated fusion of nonprimate cells with human cells. J Virol. 1990;64(5):2149–2156. doi: 10.1128/jvi.64.5.2149-2156.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Feng Y, Broder CC, Kennedy PE, Berger EA. HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science. 1996;272(5263):872–877. doi: 10.1126/science.272.5263.872. [DOI] [PubMed] [Google Scholar]
- 4.Cocchi F, DeVico AL, Garzino-Demo A, Arya SK, Gallo RC, Lusso P. Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+ T cells. Science. 1995;270(5243):1811–1815. doi: 10.1126/science.270.5243.1811. [DOI] [PubMed] [Google Scholar]
- 5.Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, Murphy PM, Berger EA. CC CKR5: a RANTES, MIP-1alpha, MIP-1beta receptor as a fusion cofactor for macrophage-tropic HIV-1. Science. 1996;272(5270):1955–1958. doi: 10.1126/science.272.5270.1955. [DOI] [PubMed] [Google Scholar]
- 6.Choe H, Farzan M, Sun Y, Sullivan N, Rollins B, Ponath PD, Wu L, Mackay CR, LaRosa G, Newman W, Gerard N, Gerard C, Sodroski J. The beta-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell. 1996;85(7):1135–1148. doi: 10.1016/s0092-8674(00)81313-6. [DOI] [PubMed] [Google Scholar]
- 7.Deng H, Liu R, Ellmeier W, Choe S, Unutmaz D, Burkhart M, Di Marzio P, Marmon S, Sutton RE, Hill CM, Davis CB, Peiper SC, Schall TJ, Littman DR, Landau NR. Identification of a major co-receptor for primary isolates of HIV-1. Nature. 1996;381(6584):661–666. doi: 10.1038/381661a0. [DOI] [PubMed] [Google Scholar]
- 8.Doranz BJ, Rucker J, Yi Y, Smyth RJ, Samson M, Peiper SC, Parmentier M, Collman RG, Doms RW. A dual-tropic primary HIV-1 isolate that uses fusin and the beta-chemokine receptors CKR-5, CKR-3, and CKR-2b as fusion cofactors. Cell. 1996;85(7):1149–1158. doi: 10.1016/s0092-8674(00)81314-8. [DOI] [PubMed] [Google Scholar]
- 9.Dragic T, Litwin V, Allaway GP, Martin SR, Huang Y, Nagashima KA, Cayanan C, Maddon PJ, Koup RA, Moore JP, Paxton WA. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature. 1996;381(6584):667–673. doi: 10.1038/381667a0. [DOI] [PubMed] [Google Scholar]
- 10.Dean M, Carrington M, Winkler C, Huttley GA, Smith MW, Allikmets R, Goedert JJ, Buchbinder SP, Vittinghoff E, Gomperts E, Donfield S, Vlahov D, Kaslow R, Saah A, Rinaldo C, Detels R, O’Brien SJ. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Francisco City Cohort, ALIVE Study. Science. 1996;273(5283):1856–1862. doi: 10.1126/science.273.5283.1856. [DOI] [PubMed] [Google Scholar]
- 11.Wood A, Armour D. The discovery of the CCR5 receptor antagonist, UK-427,857, a new agent for the treatment of HIV infection and AIDS. Prog Med Chem. 2005;43:239–271. doi: 10.1016/S0079-6468(05)43007-6. [DOI] [PubMed] [Google Scholar]
- 12.Meanwell NA, Kadow JF. Maraviroc, a chemokine CCR5 receptor antagonist for the treatment of HIV infection and AIDS. Curr Opin Investig Drugs. 2007;8(8):669–681. [PubMed] [Google Scholar]
- 13.Dorr P, Perros M. CCR5 inhibitors in HIV-1 therapy. Expert Opin Drug Discov. 2008;3(11):1345–1361. doi: 10.1517/17460441.3.11.1345. [DOI] [PubMed] [Google Scholar]
- 14.Gulick RM, Lalezari J, Goodrich J, Clumeck N, DeJesus E, Horban A, Nadler J, Clotet B, Karlsson A, Wohlfeiler M, Montana JB, McHale M, Sullivan J, Ridgway C, Felstead S, Dunne MW, van der Ryst E, Mayer H. Maraviroc for previously treated patients with R5 HIV-1 infection. N Engl J Med. 2008;359(14):1429–1441. doi: 10.1056/NEJMoa0803152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Dorr P, Westby M, Dobbs S, Griffin P, Irvine B, Macartney M, Mori J, Rickett G, Smith-Burchnell C, Napier C, Webster R, Armour D, Price D, Stammen B, Wood A, Perros M. Maraviroc (UK-427,857): a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity. Antimicrob Agents Chemother. 2005;49(11):4721–4732. doi: 10.1128/AAC.49.11.4721-4732.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Price DA, Armour D, de Groot M, Leishman D, Napier C, Perros M, Stammen BL, Wood A. Overcoming HERG affinity in the discovery of the CCR5 antagonist maraviroc. Bioorg Med Chem Lett. 2006;16 (17):4633–4637. doi: 10.1016/j.bmcl.2006.06.012. [DOI] [PubMed] [Google Scholar]
- 17.Price DA, Armour D, de Groot M, Leishman D, Napier C, Perros M, Stammen BL, Wood A. Overcoming hERG affinity in the discovery of maraviroc; a CCR5 antagonist for the treatment of HIV. Curr Top Med Chem. 2008;8(13):1140–1151. doi: 10.2174/156802608785700007. [DOI] [PubMed] [Google Scholar]
- 18.Barber CG, Blakemore DC, Chiva JY, Eastwood RL, Middleton DS, Paradowski KA. 1-Amido-1-phenyl-3-piperidinylbutanes - CCR5 antagonists for the treatment of HIV. Part 1 Bioorg Med Chem Lett. 2009;19(4):1075–1079. doi: 10.1016/j.bmcl.2009.01.009. [DOI] [PubMed] [Google Scholar]
- 19.Lemoine RC, Petersen AC, Setti L, Baldinger T, Wanner J, Jekle A, Heilek G, deRosier A, Ji C, Rotstein DM. Evaluation of a 3-amino-8-azabicyclo[3.1]octane replacement in the CCR5 antagonist maraviroc. Bioorg Med Chem Lett. 2010;20(5):1674–1676. doi: 10.1016/j.bmcl.2010.01.080. [DOI] [PubMed] [Google Scholar]
- 20.Ernst J, Dahl R, Lum C, Sebo L, Urban J, Miller SG, Lundstrom J. Anti-HIV-1 entry optimization of novel imidazopiperidine-tropane CCR5 antagonists. Bioorg Med Chem Lett. 2008;18(4):1498–1501. doi: 10.1016/j.bmcl.2007.12.058. [DOI] [PubMed] [Google Scholar]
- 21.Coakley E, Petropoulos CJ, Whitcomb JM. Assessing chemokine co-receptor usage in HIV. Curr Opin Infect Dis. 2005;18(1):9–15. doi: 10.1097/00001432-200502000-00003. [DOI] [PubMed] [Google Scholar]
- 22.Kazmierski WM, Aquino C, Chauder BA, Deanda F, Ferris R, Jones-Hertzog DK, Kenakin T, Koble CS, Watson C, Wheelan P, Yang H, Youngman M. Discovery of bioavailable 4,4-disubstituted piperidines as potent ligands of the chemokine receptor 5 and inhibitors of the human immunodeficiency virus-1. J Med Chem. 2008;51(20):6538–6546. doi: 10.1021/jm800598a. [DOI] [PubMed] [Google Scholar]
- 23.Duan M, Aquino C, Ferris R, Kazmierski WM, Kenakin T, Koble C, Wheelan P, Watson C, Youngman M. [2-(4-Phenyl-4-piperidinyl)ethyl]amine based CCR5 antagonists: derivatizations at the N-terminal of the piperidine ring. Bioorg Med Chem Lett. 2009;19(6):1610–1613. doi: 10.1016/j.bmcl.2009.02.014. [DOI] [PubMed] [Google Scholar]
- 24.Kazmierski WM, Anderson DL, Aquino C, Chauder BA, Duan M, Ferris R, Kenakin T, Koble CS, Lang DG, McIntyre MS, Peckham J, Watson C, Wheelan P, Spaltenstein A, Wire MB, Svolto A, Youngman M. Novel 4,4-disubstituted piperidine-based C-C chemokine receptor-5 inhibitors with high potency against human immunodeficiency virus-1 and an improved human ether-a-go-go related gene (hERG) profile. J Med Chem. 2011;54(11):3756–3767. doi: 10.1021/jm200279v. [DOI] [PubMed] [Google Scholar]
- 25.Duan M, Aquino C, Dorsey GF, Jr, Ferris R, Kazmierski WM. 4, 4-Disubstituted cyclohexylamine based CCR5 chemokine receptor antagonists as anti-HIV-1 agents. Bioorg Med Chem Lett. 2009;19(17):4988–4992. doi: 10.1016/j.bmcl.2009.07.074. [DOI] [PubMed] [Google Scholar]
- 26.Abel S, Russell D, Whitlock LA, Ridgway CE, Nedderman AN, Walker DK. Assessment of the absorption, metabolism and absolute bioavailability of maraviroc in healthy male subjects. Br J Clin Pharmacol. 2008;65 (Suppl 1):60–67. doi: 10.1111/j.1365-2125.2008.03137.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Chan PL, Weatherley B, McFadyen L. A population pharmacokinetic meta-analysis of maraviroc in healthy volunteers and asymptomatic HIV-infected subjects. Br J Clin Pharmacol. 2008;65 (Suppl 1):76–85. doi: 10.1111/j.1365-2125.2008.03139.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Stupple PA, Batchelor DV, Corless M, Dorr PK, Ellis D, Fenwick DR, Galan SR, Jones RM, Mason HJ, Middleton DS, Perros M, Perruccio F, Platts MY, Pryde DC, Rodrigues D, Smith NN, Stephenson PT, Webster R, Westby M, Wood A. An imidazopiperidine series of CCR5 antagonists for the treatment of HIV: the discovery of N-{(1S)-1-(3-fluorophenyl)-3-[(3-endo)-3-(5-isobutyryl-2-methyl-4,5,6,7-tetrahydr o-1H-imidazo[4,5-c]pyridin-1-yl)-8-azabicyclo[3.1]oct-8-yl]propyl}acetamide (PF-232798) J Med Chem. 2010;54(1):67–77. doi: 10.1021/jm100978n. [DOI] [PubMed] [Google Scholar]
- 29.Liu Y, Zhou E, Yu K, Zhu J, Zhang Y, Xie X, Li J, Jiang H. Discovery of a novel CCR5 antagonist lead compound through fragment assembly. Molecules. 2008;13(10):2426–2441. doi: 10.3390/molecules13102426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Emmelkamp JM, Rockstroh JK. Maraviroc, risks and benefits: a review of the clinical literature. Expert Opin Drug Saf. 2008;7(5):559–569. doi: 10.1517/14740338.7.5.559. [DOI] [PubMed] [Google Scholar]
- 31.Gilliam BL, Riedel DJ, Redfield RR. Clinical use of CCR5 inhibitors in HIV and beyond. J Transl Med. 2011;9 (Suppl 1):S9. doi: 10.1186/1479-5876-9-S1-S9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Haqqani AA, Tilton JC. Entry inhibitors and their use in the treatment of HIV-1 infection. Antiviral Res. 2013;98(2):158–170. doi: 10.1016/j.antiviral.2013.03.017. [DOI] [PubMed] [Google Scholar]
- 33.Henrich TJ, Kuritzkes DR. HIV-1 entry inhibitors: recent development and clinical use. Curr Opin Virol. 2013;3(1):51–57. doi: 10.1016/j.coviro.2012.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Gutierrez F, Rodriguez JC, Garcia F, Poveda E. Methods for determination of HIV tropism and their clinical use. Enferm Infecc Microbiol Clin. 2012;29 (Suppl 5):45–50. doi: 10.1016/S0213-005X(11)70043-X. [DOI] [PubMed] [Google Scholar]
- 35.Genebat M, Pulido I, Romero-Sanchez MC, Gonzalez-Serna A, Ferrando-Martinez S, Machmach K, Pacheco YM, Munoz-Fernandez MA, Ruiz-Mateos E, Leal M. Patients on a combined antiretroviral therapy after maraviroc clinical test show no immunovirological impairment. Antiviral Res. 2012;95(3):207–211. doi: 10.1016/j.antiviral.2012.06.007. [DOI] [PubMed] [Google Scholar]
- 36.Calcagno A, Nozza S, Gonzalez de Requena D, Galli A, D’Avolio A, Simiele M, Chiappetta S, Di Perri G, Lazzarin A, Bonora S. Pharmacokinetics of maraviroc administered at 150 mg once daily in association with lopinavir/ritonavir in HIV-positive treatment-naive patients. J Antimicrob Chemother. 2013;68(7):1686–1688. doi: 10.1093/jac/dkt074. [DOI] [PubMed] [Google Scholar]
- 37.Mora-Peris B, Croucher A, Else LJ, Vera JH, Khoo S, Scullard G, Back D, Winston A. Pharmacokinetic profile and safety of 150 mg of maraviroc dosed with 800/100 mg of darunavir/ritonavir all once daily, with and without nucleoside analogues, in HIV-infected subjects. J Antimicrob Chemother. 2013;68(6):1348–1353. doi: 10.1093/jac/dkt006. [DOI] [PubMed] [Google Scholar]
- 38.Goodrich JM, Saag M, van der Ryst E, et al. 48-week safety and efficacy of maraviroc, a novel CCR5 antagonist in combination with optimized background therapy (OBT) for the treatment of antiretroviral-experienced patients infected with dual/mixed-tropic HIV-1 [abstract LB-2]. 45th Annual Meeting of the Infectious Diseases Society of America; San Diego, California. October 4–7, 2007. [Google Scholar]
- 39.Fatkenheuer G, Nelson M, Lazzarin A, Konourina I, Hoepelman AI, Lampiris H, Hirschel B, Tebas P, Raffi F, Trottier B, Bellos N, Saag M, Cooper DA, Westby M, Tawadrous M, Sullivan JF, Ridgway C, Dunne MW, Felstead S, Mayer H, van der Ryst E. Subgroup analyses of maraviroc in previously treated R5 HIV-1 infection. N Engl J Med. 2008;359(14):1442–1455. doi: 10.1056/NEJMoa0803154. [DOI] [PubMed] [Google Scholar]
- 40.Hardy WD, Gulick RM, Mayer H, Fatkenheuer G, Nelson M, Heera J, Rajicic N, Goodrich J. Two-year safety and virologic efficacy of maraviroc in treatment-experienced patients with CCR5-tropic HIV-1 infection: 96-week combined analysis of MOTIVATE 1 and 2. J Acquir Immune Defic Syndr. 2010;55(5):558–564. doi: 10.1097/QAI.0b013e3181ee3d82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Furtado J, Madruga JV, Bicudo EL, da Eira M, Lopes MI, Netto EM, Santini-Oliveira M, Leite OH, Machado AA, Tupinambas U, de Andrade Neto JL, Lima MP, de Pedro RJ, Miranda AF, Lewi DS, Santos BR, Portsmouth S, Wajsbrot DB, Cassoli LM. Safety and immunovirologic outcomes with maraviroc combination regimens in patients with a history of past treatment failures and virologic resistance in Brazil: an open-label, multicenter phase 3b study. AIDS Res Hum Retroviruses. 2013;29(9):1203–1210. doi: 10.1089/AID.2012.0330. [DOI] [PubMed] [Google Scholar]
- 42.Antoniou T, Smith G, Su D, Raboud JM, Lee D, Kovacs C, Brunetta J, Fletcher D, Crouzat F, Loutfy M. Immunologic Effectiveness of Maraviroc- and Raltegravir-Containing Regimens (R+M+) versus Raltegravir-Based Regimens That Do Not Include Maraviroc (R+M-) J Int Assoc Physicians AIDS Care (Chic) 2012;11(3):192–197. doi: 10.1177/1545109711424967. [DOI] [PubMed] [Google Scholar]
- 43.Degli Antoni A, Weimer LE, Manfredi R, Fragola V, Ferrari C. A reduced grade of liver fibro-steatosis after raltegravir, maraviroc and fosamprenavir in an HIV/HCV co-infected patient with chronic hepatitis, cardiomyopathy, intolerance to nelfinavir and a marked increase of serum creatine phosphokinase levels probably related to integrase inhibitor use. West Indian Med J. 2013;61(9):932–936. doi: 10.7727/wimj.2011.194. [DOI] [PubMed] [Google Scholar]
- 44.Leonard JT, Roy K. The HIV entry inhibitors revisited. Curr Med Chem. 2006;13(8):911–934. doi: 10.2174/092986706776361030. [DOI] [PubMed] [Google Scholar]
- 45.MacArthur RD, Novak RM. Reviews of anti-infective agents: maraviroc: the first of a new class of antiretroviral agents. Clin Infect Dis. 2008;47(2):236–241. doi: 10.1086/589289. [DOI] [PubMed] [Google Scholar]
- 46.Westby M, Smith-Burchnell C, Mori J, Lewis M, Mosley M, Stockdale M, Dorr P, Ciaramella G, Perros M. Reduced maximal inhibition in phenotypic susceptibility assays indicates that viral strains resistant to the CCR5 antagonist maraviroc utilize inhibitor-bound receptor for entry. J Virol. 2007;81(5):2359–2371. doi: 10.1128/JVI.02006-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Bonjoch A, Pou C, Perez-Alvarez N, Bellido R, Casadella M, Puig J, Noguera-Julian M, Clotet B, Negredo E, Paredes R. Switching the third drug of antiretroviral therapy to maraviroc in aviraemic subjects: a pilot, prospective, randomized clinical trial. J Antimicrob Chemother. 2013;68(6):1382–1387. doi: 10.1093/jac/dks539. [DOI] [PubMed] [Google Scholar]
- 48.Dufty NE, Gilleran G, Hawkins D, Else LJ, Taylor S. Pharmacokinetic interaction of maraviroc with tacrolimus in a patient coinfected with HIV and hepatitis B virus following hepatic transplant due to hepatocellular carcinoma. J Antimicrob Chemother. 2012;68(4):972–974. doi: 10.1093/jac/dks486. [DOI] [PubMed] [Google Scholar]
- 49.Cipriani S, Francisci D, Mencarelli A, Renga B, Schiaroli E, D’Amore C, Baldelli F, Fiorucci S. Efficacy of the CCR5 antagonist maraviroc in reducing early, ritonavir-induced atherogenesis and advanced plaque progression in mice. Circulation. 2013;127(21):2114–2124. doi: 10.1161/CIRCULATIONAHA.113.001278. [DOI] [PubMed] [Google Scholar]
- 50.Diaz-Delfin J, Domingo P, Giralt M, Villarroya F. Maraviroc reduces cytokine expression and secretion in human adipose cells without altering adipogenic differentiation. Cytokine. 2013;61(3):808–815. doi: 10.1016/j.cyto.2012.12.013. [DOI] [PubMed] [Google Scholar]
- 51.Gruber VA, Rainey PM, Lum PJ, Beatty GW, Aweeka FT, McCance-Katz EF. Interactions between Alcohol and the HIV Entry Inhibitor Maraviroc. J Int Assoc Provid AIDS Care. 2013 doi: 10.1177/2325957413495567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Visseaux B, Charpentier C, Hurtado-Nedelec M, Storto A, Antoine R, Peytavin G, Damond F, Matheron S, Brun-Vezinet F, Descamps D. In vitro phenotypic susceptibility of HIV-2 clinical isolates to CCR5 inhibitors. Antimicrob Agents Chemother. 2011;56(1):137–139. doi: 10.1128/AAC.05313-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Borrego P, Calado R, Marcelino JM, Bartolo I, Rocha C, Cavaco-Silva P, Doroana M, Antunes F, Maltez F, Caixas U, Barroso H, Taveira N. Baseline susceptibility of primary HIV-2 to entry inhibitors. Antivir Ther. 2012;17(3):565–570. doi: 10.3851/IMP1996. [DOI] [PubMed] [Google Scholar]
- 54.Stegmann S, Manea ME, Charpentier C, Damond F, Karmochkine M, Laureillard D, Si-Mohamed A, Weiss L, Piketty C. Foscarnet as salvage therapy in HIV-2-infected patient with antiretroviral treatment failure. J Clin Virol. 2009;47(1):79–81. doi: 10.1016/j.jcv.2009.11.008. [DOI] [PubMed] [Google Scholar]
- 55.Armstrong-James D, Stebbing J, Scourfield A, Smit E, Ferns B, Pillay D, Nelson M. Clinical outcome in resistant HIV-2 infection treated with raltegravir and maraviroc. Antiviral Res. 2010;86(2):224–226. doi: 10.1016/j.antiviral.2010.02.324. [DOI] [PubMed] [Google Scholar]
- 56.Caixas U, Ferreira J, Marinho AT, Faustino I, Grilo NM, Lampreia F, Germano I, Monteiro EC, Pereira SA. Long-term maraviroc use as salvage therapy in HIV-2 infection. J Antimicrob Chemother. 2012;67 (10):2538–2539. doi: 10.1093/jac/dks240. [DOI] [PubMed] [Google Scholar]
- 57.Veazey RS, Ketas TJ, Dufour J, Moroney-Rasmussen T, Green LC, Klasse PJ, Moore JP. Protection of rhesus macaques from vaginal infection by vaginally delivered maraviroc, an inhibitor of HIV-1 entry via the CCR5 co-receptor. J Infect Dis. 2010;202(5):739–744. doi: 10.1086/655661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Malcolm RK, Forbes CJ, Geer L, Veazey RS, Goldman L, Klasse PJ, Moore JP. Pharmacokinetics and efficacy of a vaginally administered maraviroc gel in rhesus macaques. J Antimicrob Chemother. 2012;68(3):678–683. doi: 10.1093/jac/dks422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Neff CP, Kurisu T, Ndolo T, Fox K, Akkina R. A topical microbicide gel formulation of CCR5 antagonist maraviroc prevents HIV-1 vaginal transmission in humanized RAG-hu mice. PLoS One. 2011;6(6):e20209. doi: 10.1371/journal.pone.0020209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Malcolm RK, Veazey RS, Geer L, Lowry D, Fetherston SM, Murphy DJ, Boyd P, Major I, Shattock RJ, Klasse PJ, Doyle LA, Rasmussen KK, Goldman L, Ketas TJ, Moore JP. Sustained release of the CCR5 inhibitors CMPD167 and maraviroc from vaginal rings in rhesus macaques. Antimicrob Agents Chemother. 2012;56(5):2251–2258. doi: 10.1128/AAC.05810-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Fetherston SM, Boyd P, McCoy CF, McBride MC, Edwards KL, Ampofo S, Malcolm RK. A silicone elastomer vaginal ring for HIV prevention containing two microbicides with different mechanisms of action. Eur J Pharm Sci. 2012;48(3):406–415. doi: 10.1016/j.ejps.2012.12.002. [DOI] [PubMed] [Google Scholar]
- 62.Cordoba EV, Arnaiz E, De La Mata FJ, Gomez R, Leal M, Pion M, Munoz-Fernandez MA. Synergistic activity of carbosilane dendrimers in combination with maraviroc against HIV in vitro. AIDS. 2013 doi: 10.1097/QAD.0b013e328361fa4a. [DOI] [PubMed] [Google Scholar]
- 63.Barouch DH, Klasse PJ, Dufour J, Veazey RS, Moore JP. Macaque studies of vaccine and microbicide combinations for preventing HIV-1 sexual transmission. P Natl Acad Sci USA. 2012;109(22):8694–8698. doi: 10.1073/pnas.1203183109. [DOI] [PMC free article] [PubMed] [Google Scholar]


