Abstract
Introduction:
There are 36.7 million people living with HIV with 20.9 million having access to antiretroviral therapy (ART). Nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) remain the ‘backbone’ of ART. However, the currently available nine NRTIs and five non-nucleoside reverse transcriptase inhibitors (NNRTIs) have significant side effects and resistance profiles.
Areas covered:
We summarize the mechanisms of resistance and other limitations of the existing NRTIs/NNRTIs. GS-9131, MK-8591, Elsulfavirine and Doravirine are four new agents that are furthest along in development.
Expert opinion:
ART development has evolved with several new promising agents. Longer-acting agents, like MK-8591 are extremely attractive to enhance drug adherence and patient satisfaction. Doravirine offers an NNRTI effective against common mutations that has fewer side effects, limitations on dosing and drug interactions. GS-9131 is very potent and active against a variety of NRTI mutants but it is too early in its development to understand its full risks and benefits. Finally, Elsulfavirine has a long half-life and preliminary data suggests fewer side effects than the most commonly used NNRTI, efavirenz. Each of these new agents shows promise and potential to improve ART in the future. The newer generation of reverse transcriptase inhibitors have longer half-lives, more favorable adverse effect profiles, and fewer drug interactions.
Keywords: HIV Reverse Transcriptase Inhibitors, GS-9191, MK-8591, Elsulfavirine, Doravirine, NNRTI, NRTI, Nucleoside reverse transcriptase inhibitors, Non-nucleoside reverse transcriptase inhibitors, HIV
1. Background
According to the latest data from UNAIDS, 76.1 million people have become infected with HIV since the start of the epidemic, and globally, 36.7 million people globally were living with HIV in 2016 [1]. Even though the number of new infections continues to decrease, it still continues to have significant morbidity and mortality, with nearly 2 million new infections, and almost 1 million people dying from AIDS related illnesses in 2016 [2]. As of June 2017, 20.9 million people living with HIV were accessing antiretroviral therapy (ART), a number which has gone up from up from 17.1 million in 2015 and 7.7 million in 2010 [3]. ART has had dramatic effects on decreasing mortality from HIV with life expectancy of HIV-infected adults in both high- and low-income countries now reaching that of the general population in those who start therapy early, are adherent and have access to healthcare services [4–6]. Given the critical importance of ART in the treatment of HIV-infected persons, it remains a crucial component of UNAIDS’s global aims to achieve by the year 2020. The 90–90-90 initiative aims to ensure that a high proportion (90%) of people are tested for HIV infection, receiving ART, and sustain viral suppression [7]. Yet, a significant portion of the world’s population, particularly adults in Western and Central Africa, the Middle East and North Africa, have severe limitations in accessing ART (36% and 24% respectively) [1]. All of these sobering figures serve to emphasize that even though the HIV epidemic has transitioned to an endemic phase, any delay or despondency in tackling its various parameters at play risks translation into more ominous outcomes [8].
ART has had tremendous impact soon evident after its introduction in 1995 leading to massive declines in mortality and morbidity, as demonstrated in the ground-breaking HIV Outpatient Study [9], eventually transforming HIV into a chronic, usually non-fatal condition in the developed world. Reverse transcriptase inhibitors remain the central ‘backbone’ of antiretroviral therapy, since the first NRTI was introduced [10] ( Zidovudine was the first NRTI which got approval by the FDA in 1987). There are two distinct types of reverse transcriptase inhibitors, the nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) and nonnucleoside reverse transcriptase inhibitors (NNRTIs). The NRTIs undergo intracellular phosphorylation, and the active triphosphate form thereafter inhibits viral replication through competition with naturally occurring purines and pyrimidines, preventing the addition of further nucleotides by reverse transcriptase (RT), and hence terminating viral DNA replication [11]. The NNRTIs bind to a hydrophobic pocket in the palm subdomain of p66, a site that is separate from the active site targeted by the NRTI class [12]. This binding of the NNRTIs leads to a stereo-chemical change in the protein, which reduces the ability of naturally occurring nucleosides to bind to the active site pocket, and hence block viral cDNA elongation [13].
There are currently nine NRTIs and five NNRTIs available. These currently require daily administration in combination with other agents to maintain viral suppression. The costs of therapy range from $400 annually in less resource rich Nations to $12,000–15,000 in resource rich Countries. The estimated costs of treating all persons living with HIV is $20 billion dollars annually [14]. The NRTIs are typically combined with a third agent (e.g., NNRTI, protease inhibitor or integrase inhibitor) for initial therapy. This remains the standard approach worldwide to therapy. The main threats affecting the use of the reverse transcriptase inhibitor classes of medications include the emergence of resistance, toxicity and intolerance of medications, and the requirement for daily administration. One theoretical advantage despite the development of resistance is impaired viral fitness. The M184V mutation to lamivudine or emitracitabine has been associated with impaired viral fitness in vivo yet there are no clinical data indicating this improves the morbidity or mortality from HIV infection [15]. In this review, we will discuss the advantages and disadvantages of currently available reverse transcriptase inhibitors and four emerging drugs within this class.
2. Medical need and current therapeutic class review
It is testament to the immense success against HIV that 20.9 million people now have access to ART, which amounts to approximately 53% of the estimated 36.7 million people living with HIV globally [3]. AIDS related deaths have fallen by 48% to 1 million since the peak of 1.9 million in 2005 [3]. The aim of ART revolves around four major parameters: to reduce HIV-associated morbidity and prolong the duration and quality of survival; to maximally and durably suppress plasma HIVi RNA replication; to prevent HIV transmission; and restore and preserve immunologic function. We have come a long way but significant challenges remain [16].
The daily requirement for an ART regimen translates into patient fatigue after years of use, and it is no surprise that in a meta-analysis that synthesized eighty-four observational studies from twenty countries, it was found that fewer than 62% of patients maintain the ≥90% adherence which is required for optimal viral suppression [17]. Pre-exposure prophylaxis (PrEP), recommended by the World Health Organization (WHO) for high-risk populations with HIV incidence of ≥3%, has adherence which ranges from 28% to 98% [18]. Adherence to ART is also greatly confounded in patients who have other medical conditions including substance abuse and psychiatric disorders [19], not to mention those who must take more than a one pill per day ART regimen [20].
The other major challenge of a life-long therapy and a mutating virus is resistance to existing HIV medications [21, 22]. In this context, the existing of pretreatment resistance becomes even more daunting of a challenge in resource-limited settings, where resistance testing is not widely available. It has been recently shown in a systematic review and meta-regression analysis of 358 datasets, representing 56,044 adults in 63 countries, that pretreatment NNRTI resistance in 2016 at about 10% in most regions -- 11% in southern Africa, 10.1% in eastern Africa, 7.2% in western and central Africa, and 9.4% in Latin America and the Caribbean [23]. Currently, there are more than 25 antiretroviral medications from six major classes available for the treatment of HIV-infected patients. For the purpose of this review, we will focus on the reverse transcriptase inhibitors.
The spectrum of activity of NRTIs includes both HIV-1 and HIV-2, and they constitute the ‘backbone’ of ART (Table 1). Of the major NRTIs, Tenofovir disoproxil fumarate (TDF) and tenofovir alafenamide (TAF) are adenosine-derived; Emtricitabine and lamivudine are cytosine analogs; Abacavir sulfate is a guanosine analog; Zidovudine and stavudine are thymidine analogs; and Didanosine, is inosine derived. All of the NRTIs have class and drug specific side effects and can select for resistance mutations. Major side effects for TDF include kidney injury and bone loss [24–27]. TAF has less renal and bone toxicity because of the lower plasma concentrations [28], but leads to higher levels of LDL and HDL cholesterol, as well as triglycerides [29]. Lamivudine has been associated with pancreatitis [30], and Emtricitabine can cause skin hyperpigmentation [31]. The use of Abacavir in patients with high viral loads (>100,000 copies) remains a concern because of higher rates of treatment failure, as well as its suspicion for being associated with higher rates of myocardial infarctions [32, 33]. Zidovudine requires twice daily dosing and its major dose-limiting toxicity is bone marrow suppression. In addition, zidovudine is associated with a myriad of adverse reactions including headache, malaise, anorexia, nausea, vomiting, lactic acidosis, and loss of limb fat. Didanosine and Stavudine are used rarely because of their adverse effect profile including neuropathy, lipodystrophy, and mitochondrial toxicity. The most commonly used combinations are available as co-formulations: tenofovir disoproxil fumarate-emtricitabine (Truvada®), abacavir-lamivudine (Epzicom®) and tenofovir alafenamide-emtricitabine (Descovy®) in the United States. All of the NRTIs can select for resistance mutations, and this occurs via two different mechanisms. The first involves a mutated viral reverse transcriptase enzyme (mutations in the N-terminal polymerase domain of the enzyme) that selectively avoids incorporating the nucleotide analogs into the DNA instead of the normal nucleotides. The most common mutations in this category include K65R, L74V, Q151M, and M184V [34–36]. The second mechanism involves thymidine analog mutations (TAMs) – essentially the mutated reverse transcriptase enacts the phosphorolytic excision of NRTIs from the 3′ end of the viral DNA chain that extends from the primer, a process referred to as “primer unblocking” [37, 38]. TAMs include M41L, D67N, K70R, L210W, T215Y/F, and K219Q/E, and they confer resistance to all NRTIs except lamivudine (3TC) and emtricitabine (FTC). The NRTIS have few clinically significant drug-drug interactions because they are not substrates, inhibitors, or inducers of hepatic cytochrome P450 (CYP) enzymes. In summary, currently available NRTIs have significant side effect profiles, known problems with resistance and must be administered on a daily or twice daily basis.
Table 1 –
Approved Nucleoside/tide Reverse Transcriptase Inhibitors
| Drug | Uses | Dosing | Key Resistance Mutations | Common Adverse Effects | Key Issues Affecting Use |
|---|---|---|---|---|---|
| Zidovudine | HIV Therapy | Twice daily | M41L, D67N, K70R, L210W, T215Y, K219Q | Nausea, vomiting Anemia Mitochondrial toxicity |
Significant Side Effects Twice Daily Dosing |
| Lamivudine / Emitricitabine | HIV Therapy PrEP |
Once daily | M184V, K65R | Nail discoloration | Not very potent antiretroviral Single mutation confers resistance Generally well tolerated |
| Tenofovir disprovoxil fumarate* | HIV Therapy PrEP |
Once daily | K65R, K70E | Bone loss Kidney dysfunction Fanconi’s syndrome |
Significant Long term toxicity Useful to treat HIV and Hepatitis B |
| Tenofovir alafenamide* | HIV Therapy | Once daily | K65R, K70E | Nausea and diarrhea | Useful to treat HIV and Hepatitis B |
| Stavudine | HIV Therapy | Twice daily | M41L, K65R, D67N, K70R, L210W, T215Y, K219Q | Mitochondrial toxicity Peripheral neuropathy Lipodystrophy |
Significant side effects |
| Didanosine | HIV Therapy | Once daily | K65R, L74V | Gastrointestinal intolerance Mitochondrial toxicity Pancreatitis |
Significant side effects Not available as fixed dose combination |
| Abacavir | HIV Therapy | Once daily | K65R, L74V, Y115F, M184V | Hypersensitivity reaction | Need for HLA B5701 testing Possible link to myocardial infarction Less effective with HIV>100,000 cpm |
Nucleotide reverse transcriptase inhibitor
PrEP=Pre-exposure Prevention Therapy
cpm=copies/mL
The NNRTIs are active against HIV-1 but lack activity against HIV-2 (Table 2). The NNRTIs are typically administered along with a dual NRTI combination. The first generation NNRTIs includes efavirenz and nevirapine; second generation NNRTIs are rilpivirine and etravirine. The first-generation agents have a low threshold to acquire resistance and only need a single mutation. One of the major mutations, K103N leads to cross-resistance to all first generation NNRTIs. The NNRTIs have a long plasma half-life and can act as perpetrators of drug interactions. Efavirenz has multiple drug interactions and needs a thorough review of concomitantly administered drugs. In addition, it has a significant side-effect profile that includes neuropsychiatric toxicity, rash, hyperlipidemia and elevated transaminases [39, 40]. The side effect profile is worse in black patients, and long term mental health consequences including suicide have overshadowed Efavirenz’ usage [41]. Nevirapine is not recommended as an initial regimen in treatment-naïve patients [42], because of its increased toxicity that includes hepatic necrosis and serious cutaneous reactions, including deaths due Stevens-Johnson Syndrome [43, 44]. Rilpivirine can be used in treatment-naïve patients with a baseline viral load of <100,000 copies/mL and CD4 count of >200 cells/mm3. It has been associated with QT interval prolongation, and as it needs gastric acid, requires administration with a meal and concomitant use of proton pump inhibitors is contraindicated. Rilpivirine also has important drug interactions and causes neurologic and psychiatric side effects, though fewer than efavirenz [45]. Etravirine is used primarily when there is a resistant virus, and its most common side effect is rash. It also needs adjustments because of its significant drug interactions.
Table 2 –
Approved Non-Nucleoside Reverse Transcriptase Inhibitors
| Drug | Uses | Dosing | Key Resistance Mutations | Common Adverse Effects | Key Issues Affecting Use |
|---|---|---|---|---|---|
| Nevirapine | HIV Therapy | Once or twice daily | Y181C, G190A, K103N | Skin Rash Hepatotoxicity |
Significant Side Effects Resistance develops easily Not available in fixed dose combination |
| Efavirenz | HIV Therapy | Once daily | K103N, Y181C, K101P, G190A | Neuropsychiatric effects Nausea/vomiting |
Significant side effects Drug-drug interactions |
| Etravirine | HIV Therapy | Twice daily | K101P, Y181C, L100I | Skin Rash Hepatotoxicity |
Twice daily dosing Intolerance of tablets Not available in fixed dose combination |
| Rilpivirine | HIV Therapy | Once daily | L100I, K101P,E138A, Y181C, Y188L | Hepatotoxicity | Dosing limitations with food Cannot use with PPIs Less effective with HIV>100,000 cpm |
PPIs=Proton Pump Inhibitors
cpm=copies/mL
Mutations conferring NNRTI resistance involve decreasing the binding of the agent to the NNRTI binding site. There are three main mechanisms: the contact of the drug could be inhibited to the NNRTI binding pocket (e.g., K103N and K101E) [46, 47]; disruption of the contact between the inhibitor and residues in the NNRTI binding domain (e.g., Y181C and Y188L) [48, 49]; or changes in the conformation or size of the NNRTI binding pocket (e.g., Y188L and G190E) [49]. Hence, the significant interactions and side-effect profiles render use of NNRTIs problematic.
3. Current research goals and scientific rationale
There is a strong scientific rationale to design of novel agents within existing classes, particularly PIs, NNRTIs and NRTIs, INSTIs with improved pharmacological properties, tolerance, and activity against drug-resistant HIV. Developing agents that are active against drug-resistance mutants is important to have sufficient options to treat those that failure standard ART. Because of the significant adverse effects of existing agents, developing safer therapy is key. Finally, because adherence to daily therapy is challenging over a lifetime, developing longer-acting agents is of paramount importance.
A major impetus to address inadequate adherence levels is the development and implementation of long-acting agents [50, 51]. The primary goal is to address the challenges of life-long drug adherence. Significant research has been focused on long-acting Cabotegravir, an integrase strand transfer inhibitor [52]; Long acting Rilpivirine, a NNRTI [53]; drug loaded inserts (Dapivirine Vaginal Ring) [54] and implants (Tenofovir Alafenamide Implant) [55]; microparticles [56]; Atazanavir with ritonavir [57]; Combinectin, BMS-986197 [58] and MK-8591 (discussed below in detail). One potentially new paradigm is combining two long-acting agents using injectable medications. This includes the NNRTI rilpivirine in combination with an integrase strand transfer inhibitor. In the randomized, phase 2b, open-label LATTE-2 study of treatment-naive adults infected with HIV-1, the two-drug combination of all-injectable, long-acting cabotegravir plus rilpivirine every 4 weeks or every 8 weeks was as effective as daily three-drug oral therapy at maintaining HIV-1 viral suppression through 96 weeks [59]. The emerging use of longer acting agents may herald a new treatment paradigm in preventing and treating HIV infection.
4. Competitive Environment
The development of new NRTIs and NNRTIs focus on improved coverage of HIV resistant mutants, fewer side effects and improvements in pharmacologic properties. The mechanism of action of new NRTIs and NNRTIs are similar to the existing ones with no major breakthrough. We review four promising agents (Table 3): GS-9131, MK 8591, Elsulfavirine and Doravirine that are most advanced in their development.
Table 3 –
Investigational Reverse Transcriptase Inhibitors
| Drug | Sponsor | Class | Clinical Trials | Dose | Activity | Resistance Features | Special Attributes |
|---|---|---|---|---|---|---|---|
| GS-9131 | Gilead | NRTI | Pre-clinical | Not available | HIV-1 HIV-2 | Active against K65R, L74V, M184V mutants or their combinations; Selects for Q151L mutation in HIV-1 with resistance | Once daily dosing predicted |
| MK-8591 | Merck | NRTI | Phase 2 studies NCT03272347* (I) NCT02217904* (II) |
0.25 mg 0.75 mg 2.25 mg |
HIV-1 HIV-2 |
Active against K65R mutants; 9-fold reduced activity with M184V mutants | Long half-life; Weekly/Monthly dosing; Role in PrEP |
| Elsulfavirine (VM-1500) |
Viriom | NNRTI | Phase 2 studies NCT02485509* (I) NCT02489487* (I) NCT02489435* (I) NCT02489461* (II) |
20 mg 40 mg |
HIV-1 | Active against K103N, Y181C mutants | Long half-life; Weekly dosing; Fewer adverse events compared to Efavirenz |
| Doravirine (MK-1439) |
Merck | NNRTI | Phase 3 studies NCT01466985* (I) NCT01632345* (II) NCT03272347* (II) NCT02652260* (II) NCT02629822* (II) NCT02275780* (III) NCT02403674* (III) NCT02397096* (III) 12 Trials Registered* |
100 mg | HIV-1 | Active against K103N, Y181C, and G190A mutants | Developed in Fixed dose combination with tenofovir and lamivudine; Limited side-effect profile; No dose restrictions with food or in patients with moderate liver disease |
Clinicaltrials.gov registration number
PrEP=Pre-exposure Prevention Therapy
Parentheses = Indicates Phase of Clinical Trial
a. GS-9131
GS-9131 is a designer NRTI under development by Gilead Sciences [60]. Building upon the nucleoside phosphonate d4AP which was discontinued from further development because of mitochondrial toxicity, a 2’-fluorine substitution modification led to development of GS-9148 and its orally bioavailable phosphonoamidate prodrug, GS-9131 [61–62]. Using the PhenoSense HIV assay to determine activity and resistance profile of GS-9131, it was shown to have potent antiretroviral activity against HIV-1 isolates of subtypes A, B, C, D, E, F, group O and N (EC50 0.29–113 nM) [63]. In addition, it also potent activity against HIV-2 (EC50 = 21 nM) with low cytotoxicity in multiple cell types including renal cells (CC50 > 100 μM) [64, 65]. The presence of RT mutations K65R, L74V, M184V or their combinations did not affect the potency of GS-9131 (EC50 fold change < 1). GS-9131 acts in a synergistic fashion in combination studies with AZT, FTC, ABC, efavirenz, the integrase inhibitors bictegravir and dolutegravir, and the protease inhibitor, lopinavir. It has additive effects with TFV and TAF [66]. However, GS-9131 has been shown to select for the very rare Q151L mutation in HIV-1 RT as a pathway to resistance, and interference experiments have shown that Q151L severely compromises binding of GS-9148-diphosphate to RT, and the 2’-fluoro group of GS-9148 may cause steric hindrance with the side chain of the Q151L mutant [67]. GS-9131 may be an attractive candidate with a potential for once daily dosing and efficacy in patients with NRTI resistance. However, with its propensity for selecting Q151L mutation, it is possible the utility of GS-9131 will be restricted.
b. MK 8591
MK-8591 (4′-ethynyl-2-fluoro-2′-deoxyadenosine [EFdA]), an NRTI, is being developed by Merck & Co [68]. It has highly potent activity against HIV-1 and HIV-2, with very favorable toxic profiles, and stability in plasma [69–70]. This agent is 10-fold more potent than tenofovir disoproxil fumarate (TDF) [71], and 1,000 times more potent than emtricitabine (FTC) [72]. What makes MK 8591 particularly interesting is its long intracellular half-life – this is due to the fluorine at position 2 of the adenine base, which essentially confers resistance to degradation by adenosine deaminase [69, 70]. It was shown to have antiviral activity for ten days in ART-naive, HIV-1-infected participants when administered as a single 10 mg dose in a phase 1b proof-of-concept clinical trial [73]. More recent data suggests that a dose as low as 0.5 mg would suppress HIV replication for at least 7 days [74]. In healthy individuals, daily doses of 0.25 mg exceed the EC50 of HIV with adequate intracellular levels in plasma, rectal and vaginal tissues [75]. Also, it was shown that long-acting parenteral formulations of MK-8591 exhibited continuous, extended-duration drug release in rodents with MK-8591 plasma levels comparable to those achieved in rhesus and humans and duration of release exceeding 6 months [76]. With possible weekly dosing, this potential drug would represent a paradigm shift in treatment of infection, helping to greatly aid in patient compliance with their HAART regimen. There is ongoing research which suggests that a single dose of MK 8591 may be able to achieve effective concentrations for up to a year [76]. Data from Macaques support the potential use of MK-8591 for HIV-1 prophylaxis in individuals at high risk of acquiring infection [77]. In summary, MK 8591 can revamp the treatment administration timing for HIV infected patients, as well as have a possible role for HIV prevention. This drug can tackle many of the problems currently encountered when treating HIV patients on HAART, ranging from poor adherence to the development of drug-resistant virus.
c. Elsulfavirine / VM-1500A
The San Diego based Biotech Company Viriom is developing Elsulfavirine or VM-1500A, an NNRTI, also known by its brand name Elpida®. It is under development as a once-daily oral regimen and there are studies underway for long acting injectable preparations of VM-1500A. It was shown to be safe in a combined Phase Ib and IIa randomized, placebo-controlled, double-blind study of VM-1500 in healthy subjects and in patients with HIV-1 infection that are antiretroviral therapy naïve (Study identifier NCT02485509) [78, 79]. In healthy participants, the half-life of VM-1500A was 8.9 days (20-mg dose) and 8.8 days (40-mg dose) [78]. In the HIV infected cohort, the half-life of the VM-1500A metabolite after 7 days was 7.4 days (20-mg dose) and 5.4 days (40-mg dose) [78]. The next Phase IIb study (Study identifier NCT02489461) compared the efficacy of Elsulfavirine to Efavirenz in combination with TDF/FTC [80]. The study showed that Elsufavirine had similar virologic control to Efavirenz, with therapy 45/55 (81%) of Elpida and 35/47 (73.7%) of EFV patients had HIV-1 RNA values <50 copies/ml at week 48 [80]. More interestingly, Elsulfavirine was significantly better tolerated than EFV-based therapy, with drug-associated adverse events being observed in 36.7% (N=22/60 participants) in the Elsulfavirine group versus 77.6% (N=45/58 participants) in the EFV group (p <0.0001). There were no allergic reactions or skin rash observed [80]. The Russian Ministry of Health granted approval of this drug in June 2017 as an oral 20 mg capsule but it has yet to be approved in the United States or Europe [81]. The company is currently working on developing long-acting, injectable formulations. In summary, Elsulfavirine appears to have a much better side effect profile compared to Efavirenz though data are limited at this point. If confirmed in additional studies this would aid in its applicability as a potential first line treatment regimen.
d. Doravirine
Doravirine, formerly MK-1439, is an NNRTI developed by Merck & Co. It is a designer drug fashioned to overcome resistance patterns which plague the NNRTIs. It was shown to have more than 30-fold improved potency compared to that of Efavirenz against K103N mutant virus and more than 9- and 4-fold better potency than Etravirine and Rilpivirine, respectively, against Y181C mutant virus, in the presence of 50% serum [82]. It has potent activity against a wide range of resistant mutant HIV viruses [83]. In a Phase IIb study involving ART-naïve HIV-1-positive patients also receiving TDF/FTC, Doravirine 100 mg daily showed potent antiretroviral activity at week 48. There were significantly fewer treatment-emergent central nervous system side effects by week 8 than those who received Efavirenz [84]. Doravirine reduced HIV viral load by about 1.3 logs in a 7-day monotherapy dose-escalation study [85]. Doravirine also demonstrated no significant alterations in atorvastatin pharmacokinetics in healthy subjects, lending credence to its usage with atorvastatin [86]. In addition, moderate hepatic impairment did not have any meaningful effect on doravirine pharmacokinetics indicating that no dose adjustment is required [87]. Food was also shown to have no clinically meaningful effect on doravirine 100 mg alone or as part of an fixed drug combination [88]. Recently, as a part of DRIVE-AHEAD, a Phase II trial, it was shown that a once-daily single tablet, fixed-dose combination of Doravirine (DOR), lamivudine, and tenofovir disoproxil fumarate was not inferior to a fixed-dose combination of efavirenz, emtricitabine, and TDF, in treatment-naïve adults infected with HIV-1 [89]. In addition, the incidence of dizziness, sleep disorders/disturbances, and altered sensorium was lower in DOR/3TC/TDF recipients than in EFV/FTC/TDF recipients (p< 0.001, p< 0.001, and p=0.033, respectively) [89]. Interestingly, fasting LDL-C and non-HDL-C were reduced by DOR/3TC/TDF and increased by EFV/FTC/TDF (both p< 0.0001) [89]. Doravirine shows promise as a first-line regimen with its activity against virus with classic NNRTI mutations, including K103N, Y181C, and G190A. In addition, its limited side-effect profile, lack of requirement for food to enhance absorption, minimal drug interactions and lack of dose adjustment requirement for moderate liver disease, makes it an attractive antiretroviral agent.
5. Potential Development Issues
Three of these agents appear to be well on their way in the drug development process. GS-9131 is a derivative of nucleoside phosphonate d4AP that had significant mitochondrial toxicity. Whether the structural similarity will affect tolerability or development is unclear. Both doravirine and elsufavirine must contend with a market place that has integrase strand transfer inhibitors as the primary initial agents used in ART. Head to head trials may be required to determine the niche for these agents. Identifying the dosing interval for MK-8591 is the key issue for this agent. In addition, the developers will need to decide if they wish to concentrate on HIV prevention or treatment.
6. Conclusion
We have presented data on four potential reverse transcriptase inhibitor antiretroviral options in the pipeline. Each of these agents has unique properties that make them potentially useful in the armamentarium to prevent and treat HIV infection. Doravirine is effective in many NNRTI resistant isolates and has fewer side effects than Efavirenz. Elsulfavirine also appears to be as effective as Efavirenz with fewer side effects in preliminary studies. Both MK-8591 and GS-9131 are earlier in the development process though each has promising pharmacologic characteristics to add to the field. The most important development in the field would be the implementation of agents with longer half-lives, spanning days, if not weeks and months. This would aid greatly in patient adherence and potential reduce the development of drug resistance. Recent data has actually shown that a four or eight week maintenance ART injection of cabotegravir plus rilpivirine had high rates of virologic response and was well tolerated through 96 weeks, with very high patient satisfaction [59]. These data signify a paradigm shift in ART administration. MK 8591 has qualities that lend to development as a long-acting agent. In summary, the newer generation of reverse transcriptase inhibitors appear to have more favorable adverse effect profiles and fewer drug interactions in preliminary studies making them attractive to administer in patients with multiple co-morbidities if confirmed in future trials.
7. Expert Opinion
Antiretroviral therapy has transformed the natural history of HIV infection with reverse transcriptase inhibitors making a substantial impact on decreasing morbidity and mortality. The past thirty years has seen the development of six classes of drugs directed at the life cycle of HIV. Over the last decade, ART has become increasingly tolerable and moved to fixed dose combinations to improve adherence. The existing paradigm includes reverse transcriptase inhibitors as a standard backbone with a third potent agent. First line ART in 2018 increasingly consists of an integrase inhibitor in combination with two NRTIs. However, NNRTIs remain an important component of first line therapy particularly in resource poor nations. The horizon will include longer-acting agents with a potential paradigm shift to intermittent administration of ART to further improve adherence. In addition, because of the side effects of NRTIs, combination therapy omitting this class of agents is emerging. Although there is data to suggest that HIV with a M184V mutation because of exposure to lamivudine or emitracitabine are less fit, there is no data to support this has a real clinical advantage. The next decade will likely continue to see shifts in treatment of HIV.
So, how can we use these new reverse transcriptase inhibitors? The two NRTIs, GS-9131 and MK-8591, must provide an advance over existing formulations. MK-8591 appears to have the most promise with a very long half-life. Clearly, there is potential as an intermittently dosed agent or provide a measure of security in those with inadequate adherence. It is attractive for both HIV prevention and treatment. However, there has not been enough human data to understand its efficacy and side effects. Furthermore, if individuals stop other agents, will the prolonged half-life and residual levels ultimately lead to the emergence of drug resistance? If there is protracted exposure to sub-therapeutic plasma concentrations, that could lead to viral resistance to not just MK 8591 but also possibly other agents in the same class. GS-9131 may be useful for treating drug resistance strains. It is valuable to have active agents from multiple classes when treating individuals with drug resistance. However, there is a long way to go in the development of this agent. The issue of whether GS-9131 has mitochondrial toxicity requires an answer before this agent comes into clinical use.
The two NNRTIs are much closer to the clinic. We need additional studies with both of these agents to see how they fit in the larger picture of ART. Integrase strand transfer inhibitors are the first choice for a potent agent to combine with NRTIs as initial therapy. Ideally, we need randomized studies to understand how these agents compare to this class of drugs. Doravirine is under development as a combination agent with TDF that has more toxicity than TAF. It seems to be unwise to proceed with development of this single fixed-dose combination given the known safety issues with TDF. Elsulfavirine has potential to be a long-acting agent. It is unclear whether there is enough of an advantage to merit much use of this agent compared to other available antiretrovirals as a first line oral agent. It is hard to imagine how Doravirine and Elsulfavirine will supplant other agents in the absence of comparisons with other first line therapies.
Finally, cost is another important consideration for all of these new agents. A cost of $40 000-$70 000 annually for long-acting ART will offer good value for patients with multiple prior failures [90]. However, to be a viable option for first- or second-line therapy, long-acting therapy costs must approach that of currently available regimens. With the recent FDA approval for ART regimen consisting of the fixed dose combination of two agents (Juluca®, a combination of dolutegravir and rilpivirine) for individuals with a suppressed viral load and on a stable regimen for at least six months, there is already a shift in the paradigm of treating HIV away from the backbone of NRTIs. Other two drug combinations are under study. It is unknown whether we can use these new agents in dual combination regimens. NRTIs and NNRTIs remain important components of ART but newer agents must offer a significant advantage to gain approval and widespread use.
Acknowledgments
Funding: This paper was funded by National Institute of Allergy and Infectious Diseases, (UM1-AI069501).
Abbreviations
- ART
Antiretroviral therapy
- NRTIs
Nucleoside/nucleotide reverse transcriptase inhibitors
- NNRTIs
Non-nucleoside reverse transcriptase inhibitors
Footnotes
Declaration of interest: C J Fichtenbaum receives research grant support to Institution from Janssen, Merck, ViiV and Gilead. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.
References
- 1.UNAIDS. FACT SHEET JULY 2017. http://www.unaids.org/sites/default/files/media_asset/UNAIDS_FactSheet_en.pdf Accessed: 12/12/17.
- 2.Dabis F and Bekker LG, We still need to beat HIV. Science, 2017. [DOI] [PubMed] [Google Scholar]
- 3.Cousins S, Long term antiretroviral injection proves effective in trial. BMJ, 2017. 358: p. j3577. [Google Scholar]
- 4.Deeks SG, Lewin SR, and Bekker LG, The end of HIV: Still a very long way to go, but progress continues. PLoS Med, 2017. 14(11): p. e1002466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Samji H, et al. , Closing the gap: increases in life expectancy among treated HIV-positive individuals in the United States and Canada. PLoS One, 2013. 8(12): p. e81355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Wada N, et al. , Cause-specific mortality among HIV-infected individuals, by CD4(+) cell count at HAART initiation, compared with HIV-uninfected individuals. AIDS, 2014. 28(2): p. 257–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Joint United Nations Programme on HIV/AIDS. 90–90–90—An ambitious treatment target to help end the AIDS epidemic. http://www.unaids.org/en/resources/documents/2017/90-90-90.
- 8.Medley GF and Vassall A, When an emerging disease becomes endemic. Science, 2017. 357(6347): p. 156–158. [DOI] [PubMed] [Google Scholar]
- 9.Palella FJ Jr., et al. , Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med, 1998. 338(13): p. 853–60. [DOI] [PubMed] [Google Scholar]
- 10.Cihlar T and Ray AS, Nucleoside and nucleotide HIV reverse transcriptase inhibitors: 25 years after zidovudine. Antiviral Res, 2010. 85(1): p. 39–58. [DOI] [PubMed] [Google Scholar]
- 11.Kakuda TN, Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity. Clin Ther, 2000. 22(6): p. 685–708. [DOI] [PubMed] [Google Scholar]
- 12.Grobler JA, et al. , HIV-1 reverse transcriptase plus-strand initiation exhibits preferential sensitivity to non-nucleoside reverse transcriptase inhibitors in vitro. J Biol Chem, 2007. 282(11): p. 8005–10. [DOI] [PubMed] [Google Scholar]
- 13.Zdanowicz MM, The pharmacology of HIV drug resistance. Am J Pharm Educ, 2006. 70(5): p. 100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wilson D and Fraser N, Who pays and why? Costs, effectiveness, and feasibility of HIV treatment as prevention. Clin Infect Dis, 2014. 59 Suppl 1: p. S28–31. [DOI] [PubMed] [Google Scholar]
- 15.Paredes R, Sagar M, Marconi VM, et al. In vivo fitness cost of the M184V mutation in multidrug-resistant human immunodeficiency virus type 1 in the absence of lamivudine. J Virol, 2011. 83 (4): p. 2038–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Este JA and Cihlar T, Current status and challenges of antiretroviral research and therapy. Antiviral Res, 2010. 85(1): p. 25–33. [DOI] [PubMed] [Google Scholar]
- 17.Ortego C, et al. , Adherence to highly active antiretroviral therapy (HAART): a meta-analysis. AIDS Behav, 2011. 15(7): p. 1381–96. [DOI] [PubMed] [Google Scholar]
- 18.Amico KR and Stirratt MJ, Adherence to preexposure prophylaxis: current, emerging, and anticipated bases of evidence. Clin Infect Dis, 2014. 59 Suppl 1: p. S55–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gonzalez JS, et al. , Depression and HIV/AIDS treatment nonadherence: a review and meta-analysis. J Acquir Immune Defic Syndr, 2011. 58(2): p. 181–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Nachega JB, et al. , Lower pill burden and once-daily antiretroviral treatment regimens for HIV infection: A meta-analysis of randomized controlled trials. Clin Infect Dis, 2014. 58(9): p. 1297–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Broder S, The development of antiretroviral therapy and its impact on the HIV-1/AIDS pandemic. Antiviral Res, 2010. 85(1): p. 1–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Tang MW and Shafer RW, HIV-1 antiretroviral resistance: scientific principles and clinical applications. Drugs, 2012. 72(9): p. e1–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Gupta RK, et al. , HIV-1 drug resistance before initiation or re-initiation of first-line antiretroviral therapy in low-income and middle-income countries: a systematic review and meta-regression analysis. Lancet Infect Dis, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zimmermann AE, et al. , Tenofovir-associated acute and chronic kidney disease: a case of multiple drug interactions. Clin Infect Dis, 2006. 42(2): p. 283–90. [DOI] [PubMed] [Google Scholar]
- 25.Lucas GM, et al. , Clinical practice guideline for the management of chronic kidney disease in patients infected with HIV: 2014 update by the HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis, 2014. 59(9): p. e96–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yin MT and Overton ET, Increasing clarity on bone loss associated with antiretroviral initiation. J Infect Dis, 2011. 203(12): p. 1705–7. [DOI] [PubMed] [Google Scholar]
- 27.Huang JS, et al. , Bone mineral density effects of randomized regimen and nucleoside reverse transcriptase inhibitor selection from ACTG A5142. HIV Clin Trials, 2013. 14(5): p. 224–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Ruane PJ, et al. , Antiviral activity, safety, and pharmacokinetics/pharmacodynamics of tenofovir alafenamide as 10-day monotherapy in HIV-1-positive adults. J Acquir Immune Defic Syndr, 2013. 63(4): p. 449–55. [DOI] [PubMed] [Google Scholar]
- 29.Sax PE, et al. , Tenofovir alafenamide versus tenofovir disoproxil fumarate, coformulated with elvitegravir, cobicistat, and emtricitabine, for initial treatment of HIV-1 infection: two randomised, double-blind, phase 3, non-inferiority trials. Lancet, 2015. 385(9987): p. 2606–15. [DOI] [PubMed] [Google Scholar]
- 30.Manfredi R and Calza L, HIV infection and the pancreas: risk factors and potential management guidelines. Int J STD AIDS, 2008. 19(2): p. 99–105. [DOI] [PubMed] [Google Scholar]
- 31.Nelson M and Schiavone M, Emtricitabine (FTC) for the treatment of HIV infection. Int J Clin Pract, 2004. 58(5): p. 504–10. [DOI] [PubMed] [Google Scholar]
- 32.Strategies for Management of Anti-Retroviral Therapy, I. and D.A.D.S. Groups, Use of nucleoside reverse transcriptase inhibitors and risk of myocardial infarction in HIV-infected patients. AIDS, 2008. 22(14): p. F17–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Group DADS, et al. , Use of nucleoside reverse transcriptase inhibitors and risk of myocardial infarction in HIV-infected patients enrolled in the D:A:D study: a multi-cohort collaboration. Lancet, 2008. 371(9622): p. 1417–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Hachiya A, et al. , K70Q adds high-level tenofovir resistance to “Q151M complex” HIV reverse transcriptase through the enhanced discrimination mechanism. PLoS One, 2011. 6(1): p. e16242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sarafianos SG, et al. , Lamivudine (3TC) resistance in HIV-1 reverse transcriptase involves steric hindrance with beta-branched amino acids. Proc Natl Acad Sci U S A, 1999. 96(18): p. 10027–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Marcelin AG, Resistance to nucleoside reverse transcriptase inhibitors, in Antiretroviral Resistance in Clinical Practice, Geretti AM, Editor. 2006: London. [PubMed] [Google Scholar]
- 37.Asahchop EL, et al. , Antiviral drug resistance and the need for development of new HIV-1 reverse transcriptase inhibitors. Antimicrob Agents Chemother, 2012. 56(10): p. 5000–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Meyer PR, et al. , A mechanism of AZT resistance: an increase in nucleotide-dependent primer unblocking by mutant HIV-1 reverse transcriptase. Mol Cell, 1999. 4(1): p. 35–43. [DOI] [PubMed] [Google Scholar]
- 39.Rihs TA, et al. , Efavirenz and chronic neuropsychiatric symptoms: a cross-sectional case control study. HIV Med, 2006. 7(8): p. 544–8. [DOI] [PubMed] [Google Scholar]
- 40.Mollan KR, et al. , Association between efavirenz as initial therapy for HIV-1 infection and increased risk for suicidal ideation or attempted or completed suicide: an analysis of trial data. Ann Intern Med, 2014. 161(1): p. 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Leutscher PD, et al. , Discontinuation of efavirenz therapy in HIV patients due to neuropsychiatric adverse effects. Scand J Infect Dis, 2013. 45(8): p. 645–51. [DOI] [PubMed] [Google Scholar]
- 42.Gunthard HF, et al. , Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2016 Recommendations of the International Antiviral Society-USA Panel. JAMA, 2016. 316(2): p. 191–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dodi F, et al. , Stevens-Johnson syndrome in HIV patients treated with nevirapine: two case reports. AIDS, 2002. 16(8): p. 1197–8. [DOI] [PubMed] [Google Scholar]
- 44.Sanne I, et al. , Severe hepatotoxicity associated with nevirapine use in HIV-infected subjects. J Infect Dis, 2005. 191(6): p. 825–9. [DOI] [PubMed] [Google Scholar]
- 45.Cohen C, et al. , Week 48 results from a randomized clinical trial of rilpivirine/emtricitabine/tenofovir disoproxil fumarate vs. efavirenz/emtricitabine/tenofovir disoproxil fumarate in treatment-naive HIV-1-infected adults. AIDS, 2014. 28(7): p. 989–97. [DOI] [PubMed] [Google Scholar]
- 46.Das K, et al. , Crystal structures of clinically relevant Lys103Asn/Tyr181Cys double mutant HIV-1 reverse transcriptase in complexes with ATP and non-nucleoside inhibitor HBY 097. J Mol Biol, 2007. 365(1): p. 77–89. [DOI] [PubMed] [Google Scholar]
- 47.Hsiou Y, et al. , The Lys103Asn mutation of HIV-1 RT: a novel mechanism of drug resistance. J Mol Biol, 2001. 309(2): p. 437–45. [DOI] [PubMed] [Google Scholar]
- 48.Ren J, et al. , Structural mechanisms of drug resistance for mutations at codons 181 and 188 in HIV-1 reverse transcriptase and the improved resilience of second generation non-nucleoside inhibitors. J Mol Biol, 2001. 312(4): p. 795–805. [DOI] [PubMed] [Google Scholar]
- 49.Das K, et al. , Crystal structures of 8-Cl and 9-Cl TIBO complexed with wild-type HIV-1 RT and 8-Cl TIBO complexed with the Tyr181Cys HIV-1 RT drug-resistant mutant. J Mol Biol, 1996. 264(5): p. 1085–100. [DOI] [PubMed] [Google Scholar]
- 50.Rusconi S, Marcotullio S, and Cingolani A, Long-acting agents for HIV infection: biological aspects, role in treatment and prevention, and patient’s perspective. New Microbiol, 2017. 40(2): p. 75–79. [PubMed] [Google Scholar]
- 51.Nyaku AN, Kelly SG, and Taiwo BO, Long-Acting Antiretrovirals: Where Are We now? Curr HIV/AIDS Rep, 2017. 14(2): p. 63–71. [DOI] [PubMed] [Google Scholar]
- 52.Whitfield T, Torkington A, and van Halsema C, Profile of cabotegravir and its potential in the treatment and prevention of HIV-1 infection: evidence to date. HIV AIDS (Auckl), 2016. 8: p. 157–164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Williams PE, Crauwels HM, and Basstanie ED, Formulation and pharmacology of long-acting rilpivirine. Curr Opin HIV AIDS, 2015. 10(4): p. 233–8. [DOI] [PubMed] [Google Scholar]
- 54.Nel A, et al. , A safety and pharmacokinetic trial assessing delivery of dapivirine from a vaginal ring in healthy women. AIDS, 2014. 28(10): p. 1479–87. [DOI] [PubMed] [Google Scholar]
- 55.Gunawardana M, et al. , Pharmacokinetics of long-acting tenofovir alafenamide (GS-7340) subdermal implant for HIV prophylaxis. Antimicrob Agents Chemother, 2015. 59(7): p. 3913–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Cortez JM Jr., et al. , Pharmacokinetics of injectable, long-acting nevirapine for HIV prophylaxis in breastfeeding infants. Antimicrob Agents Chemother, 2015. 59(1): p. 59–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Puligujja P, et al. , Pharmacodynamics of long-acting folic acid-receptor targeted ritonavir-boosted atazanavir nanoformulations. Biomaterials, 2015. 41: p. 141–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Group, M.K.D.W.Y.S.J.D.Z.L.T.M.S.Z.M.S.M.C.f.t.C.W., HIV-1 Combinectin BMS-986197: A Long-Acting Inhibitor With Multiple Modes of Action, in CROI 2016. 2016: Boston. [Google Scholar]
- 59.Margolis DA, et al. , Long-acting intramuscular cabotegravir and rilpivirine in adults with HIV-1 infection (LATTE-2): 96-week results of a randomised, open-label, phase 2b, non-inferiority trial. Lancet, 2017. 390(10101): p. 1499–1510. [DOI] [PubMed] [Google Scholar]
- 60.Mackman RL, et al. , Discovery of GS-9131: Design, synthesis and optimization of amidate prodrugs of the novel nucleoside phosphonate HIV reverse transcriptase (RT) inhibitor GS-9148. Bioorg Med Chem, 2010. 18(10): p. 3606–17. [DOI] [PubMed] [Google Scholar]
- 61.Cihlar T, et al. , Design and profiling of GS-9148, a novel nucleotide analog active against nucleoside-resistant variants of human immunodeficiency virus type 1, and its orally bioavailable phosphonoamidate prodrug, GS-9131. Antimicrob Agents Chemother, 2008. 52(2): p. 655–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ray AS, et al. , Intracellular metabolism of the nucleotide prodrug GS-9131, a potent anti-human immunodeficiency virus agent. Antimicrob Agents Chemother, 2008. 52(2): p. 648–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Boojamra CG, et al. , Synthesis and anti-HIV activity of GS-9148 (2’-Fd4AP), a novel nucleoside phosphonate HIV reverse transcriptase inhibitor. Bioorg Med Chem Lett, 2008. 18(3): p. 1120–3. [DOI] [PubMed] [Google Scholar]
- 64.White Kirsten L., Kirsten Stray NM, Yu Helen, Stepan George, Boojamra Constantine, Mackman Richard, Ray Adrian, Miller Michael D., Cihlar Tomas, GS-9131 IS A NOVEL NRTI WITH ACTIVITY AGAINST NRTI-RESISTANT HIV-1, in CROI. 2017: Seattle, Washington. [Google Scholar]
- 65.Cihlar T, et al. , Novel nucleotide human immunodeficiency virus reverse transcriptase inhibitor GS-9148 with a low nephrotoxic potential: characterization of renal transport and accumulation. Antimicrob Agents Chemother, 2009. 53(1): p. 150–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Scarth BJ, et al. , Mechanism of resistance to GS-9148 conferred by the Q151L mutation in HIV-1 reverse transcriptase. Antimicrob Agents Chemother, 2011. 55(6): p. 2662–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wu VH, et al. , MK-8591 (4’-Ethynyl-2-Fluoro-2’-Deoxyadenosine) Exhibits Potent Activity against HIV-2 Isolates and Drug-Resistant HIV-2 Mutants in Culture. Antimicrob Agents Chemother, 2017. 61(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Ohrui H, 2’-deoxy-4’-C-ethynyl-2-fluoroadenosine, a nucleoside reverse transcriptase inhibitor, is highly potent against all human immunodeficiency viruses type 1 and has low toxicity. Chem Rec, 2006. 6(3): p. 133–43. [DOI] [PubMed] [Google Scholar]
- 69.Ohrui H, et al. , 2’-Deoxy-4’-C-ethynyl-2-fluoroadenosine: a nucleoside reverse transcriptase inhibitor with highly potent activity against all HIV-1 strains, favorable toxic profiles and stability in plasma. Nucleic Acids Symp Ser (Oxf), 2006(50): p. 1–2. [DOI] [PubMed] [Google Scholar]
- 70.Kawamoto A, et al. , 2’-deoxy-4’-C-ethynyl-2-halo-adenosines active against drug-resistant human immunodeficiency virus type 1 variants. Int J Biochem Cell Biol, 2008. 40(11): p. 2410–20. [DOI] [PubMed] [Google Scholar]
- 71.Michailidis E, et al. , Hypersusceptibility mechanism of Tenofovir-resistant HIV to EFdA. Retrovirology, 2013. 10: p. 65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Stoddart CA, et al. , Oral administration of the nucleoside EFdA (4’-ethynyl-2-fluoro-2’-deoxyadenosine) provides rapid suppression of HIV viremia in humanized mice and favorable pharmacokinetic properties in mice and the rhesus macaque. Antimicrob Agents Chemother, 2015. 59(7): p. 4190–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.HIV Early Development Team, E.F.D.S.D.J.R.S.F.-B.S.Z.M.R.A.H.D.J.H.M.I.J.G.f.t., A Single Monotherapy Dose of MK-8591, a Novel NRTI, Suppresses HIV for 10 Days, in CROI 2016. 2016: Boston. [Google Scholar]
- 74.Matthews Randolph P., Deanne Jackson Rudd DS, Levine Vanessa, Fox-Bosetti Sabrina, Zhang Sandra, Robberechts Martine, Lepeleire Inge De, Huser Andreas, Hazuda Daria J., Iwamoto Marian, and Grobler Jay A. on behalf of the HIV Early Development Team. Single doses as low as 0.5 mg of the novel NRTTI MK-8591 suppress HIV for at least seven days in IAS 2017. 2017. Paris. [Google Scholar]
- 75.Mathews RP, Rudd DJ, Levine V et al. Multiple daily doses of MK-8591 as low as 0.25 mg are expected to suppress HIV. 25th Conference on Retroviruses and Opportunistic Infections, 2018; Boston MA Abstract 26. [Google Scholar]
- 76.Hazuda; for the MK-8591 Early Development Team, J.G.E.F.S.E.B.S.L.W.W.A.K.L.F.M.-T.L.M.G.M.I.D.J. Long-Acting Oral and Parenteral Dosing of MK-8591 for HIV Treatment or Prophylaxis. in CROI 2016. 2016. Boston. [Google Scholar]
- 77.Markowitz M, G. A, St. Bernard L, et al. Weekly oral MK-8591 protects male rhesus macaques against repeated low dose intrarectal challenge with SHIV109CP3.. in IAS 2017. 2017. Paris. [DOI] [PubMed] [Google Scholar]
- 78.Ratanasuwan W WP, Koryakova A, Berzins B, Bichko V, Murphy R Pharmacokinetics of VM-1500 20 mg and 40 mg in healthy and HIV-infected patients. in International AIDS Conference (AIDS). 2014. Melbourne, Australia. [Google Scholar]
- 79.Ratanasuwan W WP, Murphy RL, Bichko V. A randomized, placebo-controlled, double-blind study of VM-1500 in HIV-naive patients. in Conference on Retroviruses and Opportunistic Infections (CROI) 2014. Boston. [Google Scholar]
- 80.Robert Murphy AVK, Elena Orlova-Morozova, Firaya Nagimova, Oleg Kozirev, Tatyana Shimonava, Marina Deulina, Natalia Vostokova, Olga Zozulya, Vadim Bichko. Elsulfavirine as compared to efavirenz in combination with tdf/ftc: 48-week study. in CROI 2017. 2017. Seattle. [Google Scholar]
- 81.Al-Salama ZT, Elsulfavirine: First Global Approval. Drugs, 2017. [DOI] [PubMed] [Google Scholar]
- 82.Lai MT, et al. , In vitro characterization of MK-1439, a novel HIV-1 nonnucleoside reverse transcriptase inhibitor. Antimicrob Agents Chemother, 2014. 58(3): p. 1652–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Cote B, et al. , Discovery of MK-1439, an orally bioavailable non-nucleoside reverse transcriptase inhibitor potent against a wide range of resistant mutant HIV viruses. Bioorg Med Chem Lett, 2014. 24(3): p. 917–22. [DOI] [PubMed] [Google Scholar]
- 84.Gatell JM, et al. , Forty-eight-week efficacy and safety and early CNS tolerability of doravirine (MK-1439), a novel NNRTI, with TDF/FTC in ART-naive HIV-positive patients. J Int AIDS Soc, 2014. 17(4 Suppl 3): p. 19532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Schurmann D, et al. , A randomized, double-blind, placebo-controlled, short-term monotherapy study of doravirine in treatment-naive HIV-infected individuals. AIDS, 2016. 30(1): p. 57–63. [DOI] [PubMed] [Google Scholar]
- 86.Khalilieh S, et al. , Results of a Doravirine-Atorvastatin Drug-Drug Interaction Study. Antimicrob Agents Chemother, 2017. 61(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Khalilieh S, et al. , Moderate Hepatic Impairment Does Not Affect Doravirine Pharmacokinetics. J Clin Pharmacol, 2017. 57(6): p. 777–783. [DOI] [PubMed] [Google Scholar]
- 88.Behm MO, et al. , The Effect of Food on Doravirine Bioavailability: Results from Two Pharmacokinetic Studies in Healthy Subjects. Clin Drug Investig, 2017. 37(6): p. 571–579. [DOI] [PubMed] [Google Scholar]
- 89.Squires KE, M. J-M, Sax PE, Wong W-W, Orkin C, Sussmann O, Tutu D, Lupinacci L, Rodgers A, Xu X, Lin G, Kumar S, Nguyen B-Y, Hanna GJ, Hwang C, Martin E, for the DRIVE-AHEAD Study Group. Fixed dose combination of doravirine/lamivudine/TDF is non-inferior to efavirenz/emtricitabine/TDF in treatment-naïve adults with HIV-1 infection: week 48 results of the Phase 3 DRIVE-AHEAD study. in IAS 2017. 2017. Paris. [Google Scholar]
- 90.Ross EL, et al. , The clinical role and cost-effectiveness of long-acting antiretroviral therapy. Clin Infect Dis, 2015. 60(7): p. 1102–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
