Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Mar 2.
Published in final edited form as: Curr Opin HIV AIDS. 2020 Jan;15(1):33–41. doi: 10.1097/COH.0000000000000591

Long-acting implants to treat and prevent HIV infection

Ethel D Weld a, Charles Flexner a,b
PMCID: PMC7050620  NIHMSID: NIHMS1556156  PMID: 31764198

Abstract

Purpose of review

Subcutaneous implants are a promising technology to enable long-acting parenteral delivery of antiretroviral drugs (ARV) because they may be able to provide protective drugs concentrations for a year or longer following a single implant. The present review covers the current status of preclinical and clinical development of antiretroviral implants.

Recent findings

Over the past three decades, subcutaneous implants have been widely used for long-acting hormonal contraception and the treatment of hormonally-driven malignancies. They are economical and scalable to manufacture, but require special procedures for insertion and removal. They are generally well tolerated, and can remain in place for up to five years. As long-acting delivery of ARV would confer significant advantages, a few investigational implants are under development for the delivery of ARV; most remain at preclinical stages of development. Islatravir, a potent nucleoside analog reverse transcriptase translocation inhibitor that shows particular promise, has entered clinical testing in implant form. Investigational implants containing tenofovir alafenamide and nevirapine, and entecavir (for hepatitis B virus) have been developed and tested in animal models, with varying degrees of success. There is also burgeoning interest in bioerodable implant formulations of established ARVs.

Summary

LARV implants are a promising new technology, but are in early stages of clinical development. Their potential advantages include more consistent and predictable drug release than that provided by intramuscular injections, the possibility of combining several partner drugs into one implant, and the fact that implants can be removed in the case of a desire to stop treatment or the development of adverse events.

Keywords: bioerodable implants, HIV, inert polymer implants, long-acting antiretroviral implants, nonbioerodible implants

INTRODUCTION

Great advances over the past three decades have led to the development and optimization of antiretrovirals that are highly potent, safe, and well tolerated. There is, however, no currently available ARV drug or formulation with a dosing frequency less than once daily. Nonadherence to daily oral medication remains the most significant barrier to long-term suppression of HIV replication and prevention of the emergence of drug-resistant virus [1]. People who experience even short periods of nonadherence are in danger of losing the health benefits of antiretroviral therapy and narrowing their choices for future treatment or prevention.

ARV dosing strategies that are less frequent than daily, enabled by the development of long-acting products and formulations, have emerged in recent years as a promising approach to adherence support [3■]. Injections of ARVs at infrequent intervals, for example every few months, would dramatically lessen the adherence concerns for those having difficulty taking once-daily pills [4]. Both injectable and implantable parenteral strategies have the advantage of avoiding hepatic first-pass metabolism and degradation in the digestive tract, while also lessening the extreme peaks and valleys which can be associated with oral administration. In general, implant formulations avoid the high peaks and ‘dose dumping’ that has occasionally been associated with intramuscular injections. An additional potential benefit of long-acting parenteral formulations would be their use in the many clinical situations where oral medications cannot be used temporarily or permanently, for example because of gastrointestinal surgery, malabsorption, swallowing dysfunction, pill aversion, or neurologic disease [5■]. In areas of the world where visible antiretroviral use is heavily stigmatized, these formulations have the potential to remove such stigma, as the use of these agents is not outwardly visible and does not involve or require pill bottles.

Long-acting injectable nanoformulations of cabotegravir and rilpivirine are already in phase III clinical trials for HIV treatment, and cabotegravir is in advanced clinical development for HIV preexposure prophylaxis; these formulations are extensively reviewed elsewhere in this volume of COHA. A critical drawback of the injectable long-acting ARVs is that they are nondialyzable and cannot be removed from the body after injection. They can maintain detectable concentrations of drug in the bloodstream for longer than a year in some cases [2]. If such formulations were to provoke an adverse effect, that adverse effect must be endured for an extended period of time; as drug concentrations decay and become subtherapeutic, drug resistance can occur.

Implants and similar sustained release technologies have been developed to deliver drugs used in the treatment and prevention of a variety of medical conditions other than HIV (see Table 1). Implant indications include hormonal contraception, prostate cancer, coronary artery disease, and cytomegalovirus retinitis, among others. Since 2013, over 53 million contraceptive implants have been distributed in the developing world, and major global gains in rates of modern contraception are attributable to increased uptake of contraceptive implants, particularly in sub-Saharan Africa [3■,4]. In some parts of the word, implants now represent half of all contraceptive methods used among women [3■]. Broadly, there are two categories of implants: matrix-style and reservoir-style. Matrix-style implants involve drug dispersed in a polymer which slowly biodegrades, controlling the rate at which the drug is released systemically. Reservoir-based implants involve a core of specially formulated drug which is encased in a polymer barrier, the properties of which control the rate of drug release. Implants have several important advantages over injectable drug delivery strategies, broadly and in the setting of HIV (see Table 2).

Table 1.

Properties of selected implants for non-HIV indicationsa

Drug and indication Materials Dose (mg/day) Duration Size
Etonorgestrel (Implanon; Nexplanon) Hormonal contraception Ethylene vinyl acetate (EVA) copolymer, barium sulfate, magnesium stearate 65 mg (0.06 mg/day) 3 years 2 × 40 m m (1 rod)
Levonorgestrel (Norplant; Jadelle) Hormonal contraception Dimethylsiloxane/methylvinylsiloxane core in thin- walled silicone (provided w. disposable trocar) 150 mg (0.03–0.04 mg/day (p 12 months) 5 years 2.5 × 43 mm (2 rods)
Leuprolide acetate (Viadur) Hormone-responsive prostate cancer Dimethyl sulfoxide, osmotic tablets 65 mg (0.17 mg/day) 1 year 4 × 45 mm (wt. 1.1 g)
Risperidone subcutaneous implant [3234] Schizophrenia Risperidone 375, 48, 720, or 960 mg (2, 2.7, 4, and 5.3 mg/day, respectively) 6 months Not mentioned in primary publication
Buprenorphine (Probuphine) (subcutaneous) Matrix-style implant for maintenance treatment of opioid addiction Drug released through four individual poly(ethylene-vinyl acetate) (EVA) rods 80 mg buprenorphine hydrochloride (0.44 mg/day) 6 months 2.5 × 26 mm
Ganciclovir (Vitrasert) (intravitreal) Cytomegalovirus retinitis Ganciclovir, magnesium stearate, PVA and EVA polymer coating 4.5 mq (1.40 μ/h, ranqe from 0.5 to 2.88 μ/h) 5–8 months 2.5 × 1 mm
Dexamethasone (Ozurdex) (intravitreal) Macular edema Dexamethasone, NOVADUR solid polymer (PLGA matrix, degrades to water and CO2) 0.7mg (3.9 meg/ day) 6 months 1 × 0.5 mm
Fluocinolone acetonide (Retisert) (intravitreal) Posterior uveitis not due to infection Fluocinolone Acetonide + magnesium stearate, microcrystalline cellulose, encased in silicone elastomer cup w polyvinyl alcohol membrane and orifice 0.59 mg (initial rate 0.6 mcg/day ×30 days, then 0.3–0.4 meg/ day thereafter) -linear release characteristics; no bolus release initially 30 months 3 × 2 × 5 mm

Note: Intravitreal implants are surgically placed into the posterior segment of the eye via a pars plana incision.

EFdA, 4’-ethynyl-2-fluoro-2’-deoxyadenosine; EVA, ethylene vinyl acetate; NMP, N-methyl-2-pyrrolidone; PCL, poly (ε-caprolactone); PLGA, biodegradable polylactic co-glycolic acid; PVA, polyvinyl alcohol.

a

Refs: Package Inserts: Viadur, Probuphine, Vitrasert, Ozurdex, Retisert, Nexplanon, Jadelle, Implanon, Norplant.

Table 2.

Potential advantages and disadvantages of antiretroviral implants as compared to injectables

Advantages Disadvantages
Swift/easy removal at the end of treatment or in setting of adverse effects Specialized device w need for training/sterility/ equipment/procedure for insertion & removal
No oral lead-in required Minor surgical procedure required to remove
No oral TDF/FTC needed to protect during subtherapeutic PK ‘tail’ Must be removed at the end of product lifespan
Lower dose/day Impossible to discern from palpation how long the device has been in place
Can remain in place for years (require less interaction with healthcare system) Can migrate from original insertion site to a place where palpation is difficult (esp. in beagles)
More consistent and predictable drug release kinetics Regulated as both a drug and a device
PK properties may not depend on injection site More complex uptake into generic marketplaces
Palpable under skin indicating its presence Visibility (arm) & possible stigma
Radio-opaque for visualization in case of unintended subcutaneous migration
Biodegradable versions also possible
Avoid high injection volumes

This review will address the state of the field of implant technology for the delivery of antiretroviral drugs (ARVs), with an emphasis on the ability of these drug delivery systems to provide effective concentrations of medications for a period of years, based on extrapolation from implant systems in current use for conditions other than HIV infection.

LONG-ACTING ANTIRETROVIRAL IMPLANTS IN PRECLINICAL AND CLINICAL DEVELOPMENT

ARV agents that are most suitable for implant formulation and delivery are those with exceptionally high antiviral potency. Because the allowable mass dose of active pharmaceutical ingredient that can loaded into a single implant rod is small, the agent or agents involved must have extraordinary capacity to inhibit viral replication at low concentrations, otherwise drug delivery by implant becomes infeasible even if one considers inserting multiple rods at the same time. Transdermal strategies (such as drug delivery via matrix-based adhesive patch or reservoir-based patches) generally depend on the drug having adequate properties for passively permeating through skin—these include low molecular weight of less than 500 Da, melting point less than 2508C, and moderate log P [1,2,3■,5■].

In contrast to most ARVs, hormones used in contraception are exceedingly potent. For perspective, a single 3-year etonogestrel (Nexplanon) implant delivers an average daily dose of etonogestrel of only 62 μg (0.06 mg) per day [6], and a single levonorgestrel (Jadelle) 75 mg implant delivers an expected daily dose of 82 μg (0.08 mg) over the 5-year insertion period [7]. In the case of the levonorgestrel implant, the release rate is known to slow over the first 2 years after insertion, dropping from approximately 100 μg/day during the first month to 40 μg/day at 12 months and 30 mg/day after month 24 [7] (see Table 1 for more details). Few marketed drugs have a daily dose of active pharmaceutical ingredient this low. The most potent approved antiretroviral drugs, tenofovir alafenamide and rilpivirine, have daily oral doses of 25 mg/day.

Several antiretroviral compounds have now been identified whose in vitro potency approaches that seen with drugs used in implants marketed for other indications. These are reviewed below, and a summary of candidate ARV implants is provided in Table 3.

Table 3.

Properties of selected ARV implants

Implant type Model Drug name Size Dose (mg/day) Duration of levels/activity
Nonbioerodable
1. Granulated drug core, PVA coating, permeable silicone tubes [35] Rats Nevirapine (NVP) 90 days
2. Pure drug powder core, platinum micro-perforated silicone tubing, PVA coating [11] Beagle dogs Tenofovir alafenamide (TAF) 1.9 × 4.0 mm In vivo release rate: 1.07 mg/day (Human doses down to 0.15 mg/day) 40 days
3. Refiliable nonpolymer nanochannel delivery implant (NDI)[15■]
  • Titanium drug reservoir

  • Silicone nanochannel membrane

  • Twoi sealable refillable silicone drug loading ports

Rhesus macaques Tenofovir alafenamide (TAF) and Emtricitabine (FTC) FTC: 43 mm × 28.5 mm × 8.7 mm; 250 nm nanochannel TAF: 5mm × 20mm × 12.3 mm; 20 nm nano-channel In vitro: TAF: 0.21±0.03 mg/ day FTC: 2.67±0.35 mg/day 83 days (TFV-DP); 28 days (FTC-DP)
4. Silicone-based transdermal matrix-based (drug-in-adhesive) patch w polyi so butylene adhesives [5■] Dermatomed human cadaver skin Tenofovir alafenamide (TAF) 7 × 7 cm Permeation flux of 7 μg/cm2/h (extrapolates to 8.4 mg TAF/ day) 1 week (in vitro)
5. Biocompatible polymer blended with entecavir via hot melt extrudates and polymer coated tablets (both administered subcutaneously) [16] Rats (Wistar han) Entecavir (ETV) Dose 350 mg/kg 87 days
6. Titanium osmotic mini-pump system (Medici DDS™) [36] TDF-FTC ′match-stick sized′ 6 months to 12 months
7. Biodegradable and nonbiodegradable matrix-based polymer with islatravir [37■■]
  • HME process: barrel temp above melting point for polymer but below melting temp for drug→ solid crystalline drug in polymer matrix

Rats, NHP Islatravir (ISL) aka EFdA (MK-8591) 2 mm × 40 mm > 10 μg/day for entire study > 6 months (for 40 wt% and 60 wt% MK- 8591 in PCL, and 50 wt% MK-8591 in EVA) >12 months (for 60 wt% MK-8591 in PCL implants)
8. Biodegradable and nonbiodegradable matrix-based polymer [23■■,37■■] (same polymer and applicator as Nexplanon) Humans (healthy volunteers) (N=16; 12 drug and 4 placebo) Islatravir (ISL) aka EFdA (MK-8591) 2 mm × 40 mm 54 and 62 mg (0.17 mg/day) 12 months+
Bioerodable
9. Ultra-long-acting removable DTG/ PLGA/NMP in 0.3 : 1:2 ratio [25■■] (formulation optimized for mice, not macaques) Rhesus macaque (treatment) Dolutegravir (DTG) 100 mg
10. Ultra-long-acting removable DTG/ PLGA/NMP in 0.3:1:2 ratio [25■■] (formulation optimized for mice) Humanized BLT mouse (prevention) Dolutegravir (DTG) 1 cm 250 mg/kg (5.5–7.0 mg DTG in 50–80 μl) > 5 months (flat shape of concentration: time curve at 140 days)
11. Reservoir-style implant [26■] (extruded tube of a biodegradable polymer, PCL, filled w TAF and castor oil excipient in 2 : 1 ratio) In vitro Tenofovir alafenamide (TAF) 1, 4, and 7cm (length) Wall thickness 100 μm Release rates: 0.28 ±0.06 mg/day (100 μm thickness) Range from 0.15 mg/day (for 200 μm thickness) to 0.91 mg/day (for 45 μm thickness) 1 80 days (in vitro)

Tenofovir prodrugs

Tenofovir is the world’s most widely used antiretroviral, and is currently marketed as two different oral prodrugs. Since its intracellular active metabolite, tenofovir-diphosphate (TFV-diphosphate), has an estimated half-life of 60–100 h, tenofovir prodrugs could easily be given less frequently than once per day while maintaining antiviral effects [8]. The prodrug tenofovir alafenamide (TAF) is 10 times more potent than tenofovir disoproxil fumarate (TDF), and has the added advantage that it is taken up into cells from the plasma as the prodrug, which is converted to the parent drug and then to the active intracellular diphosphate inside the cell [9]. In addition, from the perspective of prevention, TAF has activity against both hepatitis B virus (HBV) and HIV, and could potentially prevent both infections, in addition to treating them in those who have already acquired coinfection [10].

Several approaches have been pursued for the creation of a TAF implant. In the first, investigators loaded pure TAF powder into platinum cured micro-perforated silicone tubing that was coated with polyvinyl alcohol (PVA). The 1.9 × 40 mm implants were inserted subcutaneously into beagle dogs. This implant produced measurable plasma concentrations of tenofovir for more than 6 weeks and delivered tenofovir at an approximately constant rate for 5 to 40 days after implantation [11]. The zero-order, linear kinetics were observed over the first 30 days, after which there was a steeper decline with first-order kinetics because of drug depletion. The implants were well tolerated in the beagles, without significant AEs. Strikingly, intracellular tenofovir-diphosphate concentrations were 11–32 times higher than those associated with effective preexposure prophylaxis in humans in the iPrEX study of 16–48 fmol/106 cells after using TDF [11,12]. One advantage of this implant technology is that its physical characteristics (tube diameter, number, and size of perforations) can be easily modified to alter the release rate of drug. This device has now entered Phase I testing in humans, although no data are yet publically available.

Another approach for innovative delivery of TAF is a silicone-based transdermal matrix-based (drug-in-adhesive) patch with polyisobutylene adhesives, which has been tested on dermatomed human cadaver skin and has been established to yield a permeation flux of 7 μg/cm2/h over 1 week, which extrapolates to a release rate of 8.4 mg of TAF per day. This noninvasive approach offers convenience, simplicity, has no procedural requirements and thus stands to be more user friendly [5■].

The combination of TAF and FTC (shown to be efficacious as PrEP in the DISCOVER trial) [13,14] has been investigated in rhesus macaques, using a refillable nonpolymer nanofluidic implant (nanochannel delivery implant, NDI) consisting of a titanium drug reservoir, silicone nanochannel membrane, and two sealable refillable silicone drug loading ports [15■]. The advantage of this approach is that when drug is exhausted, more drug can be administered transcutaneously into the ports, without the need to remove and reimplant a new device. The NDI achieved concentrations of TFV-diphosphate in PBMCs that reached target 3 days after implantation, and continued to increase from a mean of 71.7 ± 29.7 to 533.3 ± 198.4 fmol/106 PBMCs over 70 days (equivalent to 12 times the iPrEX EC90) [12,15■]. Concentrations of FTC-TP in PBMCs were detected and sustained from day 3 to day 28 (1223.7 ± 416.8 to 1656.7 ± 290.8 fmol/106 PBMCs), but unlike TAF did not attain target protective concentrations (equivalent to an EC90 extrapolated from the iPrEX study of 5000–6000 fmol/106 freshly lysed PBMCs). The NDI was fairly well tolerated, but 1 in 3 animals had swelling/seroma around the device, which resolved in 7 days, one animal had dehiscence over the NDI at 60 days, and 2 animals developed skin ulceration over the implant at day 70.

Entecavir

Developed as a possible way to prevent acquisition of hepatitis B virus infection, entecavir has been given subcutaneously in two different forms. In both cases, subcutaneous delivery in rats was not tolerated. Tested technologies included both a bio-compatible polymer blended with entecavir via hot melt extrusion, and polymer coated tablets, both administered subcutaneously. When a dose of 350 mg/kg was implanted, it did achieve concentrations in the range of those associated with efficacy in rats (44–109 nM). However, in almost all cases, there was skin ulceration with surface tissue adhesion to the implant, and erosion through skin. On histological examination, there was local necrosis overlying the implant, which was attributed to entecavir itself, given that it was also observed in people who received uncoated tablets of entecavir [16].

4’-Ethynyl-2-fluoro-2’-deoxyadenosine (MK8591; islatravir)

4’-Ethynyl-2-fluoro-2’-deoxyadenosine (EFdA; MK8591; islatravir [ISV]) is a highly potent nucleoside reverse transcriptase inhibitor with a novel mechanism of action [17]. ISV is a 4’-substituted NRTI that retains its 3’-hydroxyl group, unlike any other currently approved NRTI. As a consequence, this drug has higher affinity for the active site of the HIV reverse transciptase, contributing to its potency. The incorporation of ISV-monophosphate into the RT active site blocks primer translocation and halts HIV replication without causing chain termination, since the drug contains a 3’-hydroxyl. ISV also has a halogen substitution at the 2-position of the adenine ring that impairs degradation by adenosine deaminase and contributes to the prolonged intracellular half-life of active phosphorylated metabolite ISV-TP, estimated at greater than 72 h [18]. ISV has substantial efficacy in treatment and prevention of HIV in animal models with doses as low as0.1 mg/kg/day. In animals, the drug is safe and well tolerated across a range of doses [19].

ISV has a favorable resistance profile for HIV treatment and prevention. The major resistance mutation associated with the use of this drug has been M184V, which only modestly reduces drug sensitivity in vitro [20]. These suggest the possibility of using ISV in patients already harboring M184V-containing HIV, and of using this drug in combination with other existing antiretroviral NRTIs and other drug classes. ISV is approximately 10-fold more potent against HIV-2 isolates than against HIV-1 in vitro, indicating broad-spectrum activity and possible global utility for this agent [21]. Possible uses for ISV implants, like those for TAF, include preexposure prophylaxis as a single agent, or eventual combination with other long-acting antiretroviral drugs for HIV treatment.

In studies comparing two nondegradable polymer implants in rats, the apparent ISV plasma half-life was nearly 100 days, suggesting the possibility for developing human implants with a dosing interval of 1 year or longer [22]. Phase I clinical studies of ISV are currently underway in human subjects, and preliminary findings in healthy human volunteers showed that a 62 mg implant can achieve a constant, linear (zero-order) release of drug that results in levels of triphosphorylated active metabolite [ISV-TP] in PBMCs of 0.076 pmol/106 over 12 months, which is approximately 7 times the IC50 in vitro for wild-type virus, maintaining intracellular concentrations above the target of 0.05 pmol/106 cells [23■■]. The t1/2 observed in humans of parent ISV was 50–60 h and of active metabolite ISV-TP was 120–177 h. The implants were generally well tolerated, though with local erythema, itching, and induration that was mild and to some extent dose-dependent. Importantly, based on modeling, the projected time at which concentrations fall below the target is not until 16 months after implant was placed. This raises the possibility of implant placement frequency less often than yearly for HIV treatment and prevention [17].

Bioerodable implants

Some problems associated with medical implants could be addressed with newer technologies. For example, bioerodable/biodegradable implants, for instance using polylactic acid or polylactide, have been deployed in drug-eluting stents [24]. In the case of a bioerodadable antiretroviral ‘implant,’ the formulation would be injected, forming a solid gel that would disperse over several months’ time. This has the advantage that the implant system does not require eventual removal. However, if there is the desire to remove the implant for adverse drug effects, surgical removal becomes more difficult, and potentially impossible, several months after injection, because the implant has dispersed.

One example of a bioerodable implant currently in preclinical testing is ultra-long-acting dolutegravir (DTG). Ultra-long-acting DTG delivered via a removable implant system has been tested in a macaque model for HIV treatment and a humanized bone marrow-liver-thymus (BLT) mouse for HIV prevention [25■■]. The technology involves subcutaneous injection of an admixture of DTG, PLGA, and NMP in a ratio of 0.3:1:2. Liquid drug is injected and then solidifies over the first 48 h in vivo into an implant once it reaches the aqueous environment of the body, and then biodegrades slowly, resulting in sustained drug release. This delivers drug at effective concentrations for up to 9 months and can be safely removed should an adverse event or pregnancy occur. Implants were well tolerated in both animal models, with no local necrosis. The sustained drug concentrations resulted in both suppression of viremia in the macaques and prevention from acquiring HIV via a high-dose repeat vaginal challenge.

Another bioerodable approach that has been tested in vitro involves the creation of an extruded tube made of a biodegradable polymer, PCL of various thicknesses, filled with TAF and castor oil excipient in a 2 : 1 ratio [26■,27]. The implant achieved over 180 days daily TAF release rates of 0.28 ± 0.06 mg/day (100 μm thickness), with a range from 0.15 mg/day (for 200 μm thickness) to 0.91 mg/day (for 45 μm thickness). This is above the concentration of 0.15 mg/day that is estimated to be sufficient to achieve target TFV-diphosphate intracellular concentrations for anti-HIV efficacy and protection. Daily release rates can be modified by changing the implant surface area, PCL tube wall thickness (the thinner the more drug release), or PCL crystallinity.

ANTIRETROVIRAL IMPLANTS: CHALLENGES AND OPPORTUNITIES

Theoretical advantages of implant technology for HIV treatment and prevention include the ability to remove the device in the case of side effects or the desire to end therapy, less frequent dosing because of the slow release of drug and longer apparent half-life, a lower drug dose per day because of potency and formulation properties, protection from poor adherence as compared to daily oral drugs, and the possibility of directly observed therapy. In the case of formulations with a prolonged low-level pharmacokinetic ‘tail,’ the device can simply be removed, allowing drug concentrations to decline rapidly. Medical implants are placed where they cannot generally be detected by partners, family members, or colleagues, affording better protection of health privacy. The widespread use of contraceptive implants in low income countries proves that these devices can be implemented cheaply and safely in remote areas.

Although implants have some clear advantages over injectable formulations, they also have some drawbacks (see Table 2). This includes the need for insertion and removal by well trained personnel using sterile technique. Because these devices are removable, it is likely that a novel antiretroviral developed for implant technology would not require a partnered oral formulation or oral lead-in to rule out toxicity or hypersensitivity, as is the case for injectable cabotegravir and rilpivirine [28,29]. Nonetheless, different dose formulations would still need to be developed to allow for dose escalation in clinical dose-finding and safety studies.

In HIV prevention applications, an antiretroviral implant could be developed as a stand-alone. However, partner formulations would need to be identified for HIV treatment. Ideally, a single implant could contain two or more antiretrovirals with similar pharmacokinetic properties, or two separate implants could be inserted simultaneously. As an intermediate approach, a long-lasting implant could be paired with periodic injections of an intramuscular formulation like LA-cabotegravir or LA-rilpivirine, or one or more long-acting, broadly neutralizing monoclonal antibodies [30,31].

CONCLUSION

Implants show great promise for HIV treatment and prevention. Potential advantages over intramuscular injections include the fact that implants are removable in cases of toxicity, produce more consistent and predictable drug release, have pharmacokinetic properties that may not be dependent on the injection site, and likely will not require an oral lead-in phase. In addition, implants can sustain therapeutic drug concentrations and remain in place for years, based on experience with hormonal contraception. However, implants require specialized training for insertion and removal, a minor surgical procedure if they need to be removed, and are more expensive and complex to manufacture than injectables, making transition to a generic marketplace more difficult as compared to injectable formulations. Many of these problems can and will be solved with future innovation, adding to the number of antiretroviral choices available to people living with or at risk of HIV.

KEY POINTS.

  • Long-acting antiretrovirals implants are drug delivery devices that do not depend on user compliance/reliability, and have several advantages over other long-acting antiretroviral formulations incurrent development.

  • An important advantage of implant technology is that sustained-release inert polymer implants can be removed in the case of adverse drug reactions or the desire to discontinue treatment.

  • Another notable advantage of implant technology is avoidance of a prolonged pharmacokinetic ‘tail’ at subtherapeutic concentrations of ARV, which is seen after intramuscular injections, thus lessening the selection pressure for drug resistance.

  • Implants may provide more consistent, predictable drug release than intramuscular formulations.

  • Bioerodable implants offer key advantages over nonbiodegradable implants, chief among them that they do not require removal, but can be removed soon after injection in case of adverse effects.

Financial support and sponsorship

C.F. and E.W. received related support during the preparation of this manuscript from NIH grant NIAID R24 AI118397, Long-Acting/Extended Release Antiretroviral Resource Program (LEAP), www.longactinghiv.org, awarded to Johns Hopkins University. E.W. was also supported in this work by the Johns Hopkins University Center for AIDS Research (CFAR) under award number P30AI094189.

Footnotes

Conflicts of interest

C.F. reports serving as a paid consultant for Cipla Pharmaceuticals, Janssen Pharmaceuticals, Merck Laboratories, Mylan Pharmaceuticals, and ViiV Healthcare, and received research grant support from Gilead Sciences paid to his University. E.W. is an investigator on clinical studies funded by Imquest Biosciences, Inc., GSK/ ViiV, and Navigen Pharmaceuticals, under research contracts managed by Johns Hopkins University.

REFERENCES AND RECOMMENDED READING

Papers of particular interest, published within the annual period of review, have been highlighted as:

■ of special interest

■■ of outstanding interest

  • 1.Barnabas RV, Celum C. Closing the gaps in the HIV care continuum. PLoS Med 2017; 14:e1002443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Landowitz R, Eron JJ, Grinsztein G, et al. Tail-phase safety, tolerability and pharmacokinetics of long-acting injectable cabotegravir in HIV-uninfected individuals: HPTN 077 final results HIV Research for Prevention (HIVR4P), Madrid, Spain; October 21–25, 2018. [Google Scholar]
  • 3.■.Jacobstein R Liftoff: the blossoming of contraceptive implant use in Africa. Glob Health Sci Pract 2018; 6:17–39. [DOI] [PMC free article] [PubMed] [Google Scholar]; Provides useful data and discussion on the skyrocketing uptake of contraceptive implants in Africa, useful for understanding real-world implant social acceptability and demand in another setting than HIV.
  • 4.Family Planning 2020 summary document on Implant Access Program 2019. http://www.familyplanning2020.org/sites/default/files/Our-Work/ppfp/2018%20IAP%202%20pager_VF.pdf. [Accessed 13 August 2019]
  • 5.■.Puri A, Bhattaccharjee SA, Zhang W, et al. Development of a transdermal delivery system for tenofovir alafenamide, a prodrug of tenofovir with potent antiviral activity against HIV and HBV. Pharmaceutics 2019; 11:; pii: E173. [DOI] [PMC free article] [PubMed] [Google Scholar]; Demonstrates feasibility of designing a drug-in-adhesive transdermal patch approach for TAF delivery that achieves clinically relevant permeation flux rates of drug across dermatomed human cadaver skin.
  • 6.Nexplanon® (Etonogestrel) Implant Prescribing Information. https://www.merck.com/product/usa/pi_circulars/n/nexplanon/nexplanon_pi.pdf. [Accessed December 2001]
  • 7.Jadelle® (Levonorgestrel) Implant Prescribing Information. [Accessed December 2016].
  • 8.Chen J, Flexner C, Liberman RG, et al. Biphasic elimination of tenofovir diphosphate and nonlinear pharmacokinetics of zidovudine triphosphate in a microdosing study. J Acquir Immune Defic Syndr 2012; 61:593–599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ruane PJ, DeJesus E, Berger D, 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; 63:449–455. [DOI] [PubMed] [Google Scholar]
  • 10.Childs-Kean LM, Egelund EF, Jourjy J. Tenofovir alafenamide for the treatment of chronic hepatitis B monoinfection. Pharmacotherapy 2018; 38:1051–1057. [DOI] [PubMed] [Google Scholar]
  • 11.Gunawardana M, Remedios-Chan M, Miller CS, et al. Pharmacokinetics of long-acting tenofovir alafenamide (GS-7340) subdermal implant for HIV prophylaxis. Antimicrob Agents Chemother 2015; 59:3913–3919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Anderson PL, Glidden DV, Liu A, et al. Emtricitabine-tenofovir concentrations and preexposure prophylaxis efficacy in men who have sex with men. Sci Transl Med 2012; 4:151ra25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Charles B, Hare JC, Peter R, et al. The phase 3 DISCOVER study: daily F/TAF or F/TDF for HIV pre-exposure prophylaxis. Seattle, WA, USA: CROI; 2019; 2019. [Google Scholar]
  • 14.Spinner CDBJ, Shalit P, et al. DISCOVER study for HIV preexposure prophylaxis (PrEP): F/TAF has a more rapid onset and longer sustained duration of HIV protection compared with F/TDF. Mexico City, Mexico: 10th International Antiviral Society (IAS) Conference on HIV Science; 2019. [Google Scholar]
  • 15.■.Chua CYX, Jain P, Ballerini A, et al. Transcutaneously refillable nanofluidic implant achieves sustained level of tenofovir diphosphate for HIV preexposure prophylaxis. J Control Release 2018; 286:315–325. [DOI] [PMC free article] [PubMed] [Google Scholar]; Preclinical study in rhesus macaques showing that a refillable nonpolymer nanofluidic implant containing TAF/FTC achieves clinically relevant levels of TFV-diphosphate in PBMCs for 83 days, and FTC-TP for 28 days. Ports can be refilled with subcutaneous injection into silicone ports when drug is exhausted, which could potentially be tracked clinically with individualized therapeutic drug monitoring.
  • 16.Henry SJ, Barrett SE, Forster SP, et al. Exploration of long-acting implant formulations of hepatitis B drug entecavir. Eur J Pharm Sci 2019; 136:104958. [DOI] [PubMed] [Google Scholar]
  • 17.Markowitz M, Sarafianos SG. 4’-Ethynyl-2-fluoro-2’-deoxyadenosine, MK-8591: a novel HIV-1 reverse transcriptase translocation inhibitor. Curr Opin HIV AIDS 2018; 13:294–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bernatchez JA, Paul R, Tchesnokov EP, et al. Derivatives of mesoxalic acid block translocation of HIV-1 reverse transcriptase. J Biol Chem 2015; 290:1474–1484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Stoddart CA, Galkina SA, Joshi P, 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; 59:4190–4198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Oliveira M, Brenner BG, Xu H, et al. M184I/V substitutions and E138K/M184I/V double substitutions in HIV reverse transcriptase do not significantly affect the antiviral activity of EFdA. J Antimicrob Chemother 2017; 72:3008–3011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wu VH, Smith RA, Masoum S, 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:; pii: e00744–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Grobler JAFE Barrett SE, et al. Long-acting oral and parenteral dosing of MK-8591 for HIV treatment or prophylaxis. Boston, MA, USA: Conference on Retroviruses and Opportunistic Infections (CROI); 2016. [Google Scholar]
  • 23.■■.Matthews RP, Patel M, Zhu W, et al. First-in-human trial of MK-8591-eluting implants demonstrates concentrations suitable for HIV prophylaxis for at least one year International Antiviral Society (IAS); Mexico City, Mexico: in press. [Google Scholar]; First in human clinical results of an islatravir implant, demonstrating favorable safety and concentrations estimated to be protective over the first year after insertion.
  • 24.Kalra A, Rehman H, Khera S, et al. New-generation coronary stents: current data and future directions. Curr Atheroscler Rep 2017; 19:14. [DOI] [PubMed] [Google Scholar]
  • 25.■■.Kovarova M, Benhabbour SR, Massud I, et al. Ultra-long-acting removable drug delivery system for HIV treatment and prevention. Nat Commun 2018;9:4156. [DOI] [PMC free article] [PubMed] [Google Scholar]; High-interest preclinical study of a bioerodable implant formulation of the established potent ARV Dolutegravir, which can be injected subcutaneously in a liquid form, then immediately solidifies into a solid pellet upon reaching body temperature, and slow-elutes drug over a period of 4 to 5 months. Contains pharmacokinetic in macaques, and a challenge study in humanized BLT mice, demonstrating 80% protection against repeated vaginal challenge with HIVCH040 or HIVTHRO.
  • 26.■.Johnson LM, Krovi SA, Li L, et al. Characterization of a reservoir-style implant for sustained release of tenofovir alafenamide (TAF) for HIV pre-exposure prophylaxis (PrEP). Pharmaceutics 2019; 11:; pii: E315. [DOI] [PMC free article] [PubMed] [Google Scholar]; A study of the fabrication and in-vitro performance of a bioerodable polymer reservoir-style implant of TAF, which achieves sustained release rates of TAF over 180 days that can be tweaked by varying implant parameters such as surface area, PCL tube wall thickness, and PCL crystallinity.
  • 27.Schlesinger E, Johengen D, Luecke E, et al. A tunable, biodegradable, thin-film polymer device as a long-acting implant delivering tenofovir alafenamide fumarate for HIV preexposure prophylaxis. Pharm Res 2016; 33:1649–1656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Margolis DA, Gonzalez-Garcia J, Stellbrink HJ, 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, noninferiority trial. Lancet 2017; 390:1499–1510. [DOI] [PubMed] [Google Scholar]
  • 29.Markowitz M, Frank I, Grant RM, et al. Safety and tolerability of long-acting cabotegravir injections in HIV-uninfected men (ECLAIR): a multicentre, double-blind, randomised, placebo-controlled, phase 2a trial. Lancet HIV 2017; 4:e331–e340. [DOI] [PubMed] [Google Scholar]
  • 30.Caskey M, Klein F, Nussenzweig MC. Broadly neutralizing anti-HIV-1 monoclonal antibodies in the clinic. Nat Med 2019; 25:547–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Caskey M, Klein F, Nussenzweig MC. Broadly neutralizing antibodies for HIV-1 prevention or immunotherapy. N Engl J Med 2016; 375:2019–2021. [DOI] [PubMed] [Google Scholar]
  • 32.Dammerman R, Kim S, Adera M, Schwarz A. Pharmacokinetics and safety of risperidone subcutaneous implants in stable patients with schizophrenia. Clin Pharmacol Drug Dev 2018; 7:298–310. [DOI] [PubMed] [Google Scholar]
  • 33.Dammerman R, Kim S, Adera M, Schwarz A. A phase I, open-label, single dose pharmacokinetic study in stabilized patients with schizophrenia following risperidone implant. Psychopharmacol Bull 2017; 47:36–40. [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 34.Dammerman R, Kim S, Adera M, Schwarz A. A phase-I, 6-month open-label, dose-ranging pharmacokinetic study in stabilized patients with schizophrenia following risperidone implant. Psychopharmacol Bull 2017; 47:29–35. [PubMed] [Google Scholar]
  • 35.Chen J, Walters K, Ashton P. Correlation of in vitro-in vivo release rates for sustained release nevirapine implants in rats. J Contr Rel 2005; 101(1–3): 357–358. [PubMed] [Google Scholar]
  • 36.Partners IPa; 2019. https://www.intarcia.com/pipeline-technology/itca-650.html. [Accessed 12 November 2019]
  • 37.■■.Barrett SE, Teller RS, Forster SP, et al. Extended-duration MK-8591-eluting implant as a candidate for HIV treatment and prevention. Antimicrob AgentsChemother 2018; 62:; pii: e01058–18. [DOI] [PMC free article] [PubMed] [Google Scholar]; An important and seminal study of an Islatravir (ISL) aka EFdA (MK-8591) implant, with PK in vitro and in rats and nonhuman primates, demonstrating that this biodegradable and nonbiodegradable matrix-based implant (same polymer and applicator system as is used in Nexplanon) can achieve more than 180-day extended drug release. Includes model-based projections of plasma PK in humans, and micro-CT and scanning electron microscopy to image implants at various time points and help characterize parameters of implant porosity and drug release.

RESOURCES