1. Introduction
Long-acting (LA) drug delivery systems (DDSs) harness polymer properties to achieve spatiotemporal control over release and drug biodistribution. This allows the LA dosage regimens to extend from days to months. Such DDSs significantly reduce the burden of chronic disease. Treatment success parallel medication adherence. The development of LA medicines are traced back to the 1930s with the discovery of hydrophobic drug release from implants [
8]. However, the discovery of low molecular weight (MW) dye diffusion through silicone tubing laid the groundwork for rate-controlling LA DDS polymers [
9]. Biopharmaceutical research activities directed their efforts to convert biocompatible silicone tubing into versatile drug-delivery materials. These efforts included improved uses of atropine, histamines, anesthetics, steroids, antimalarial, and antischistosomal agents [
9]. Since the 1960s, the work of Drs. Folkman, Langer, Higuchi, Roseman, Peppas, Heller, Ringsdorf, and Speiser pioneered the LA DDSs advancements seen today. Widespread research resulted in the development of LA DDS which extended the dosage intervals for a broad range of drug regimens. This resulted in improved treatment outcomes for contraception and for treatments of psychosis, diabetes, osteoporosis, and ocular diseases.
A key example of a treatable chronic disease that would benefit from LA DDSs is human immune deficiency virus type one (HIV-1) infection. Here, prior studies demonstrated that non-adherence to therapeutic regimens was a significant limiting factor in achieving successful treatment outcomes. One notable is the development of viral resistance. Another is simply remembering to take the medicines on daily prescribed requirements. Indeed, patients have repeatedly expressed treatment preferences for infrequent dosing regimens as without a viable cure or preventive vaccine, daily oral antiretroviral therapy (ART) is a principal means for HIV-1 treatment and prevention. Over the past two decades, considerable efforts have been focused on the development of LA ARTs that includes a specific focus on controlled-release LA formulations. Such formulations include solid implants, vaginal rings (VRs), and surfactant-stabilized aqueous nanocrystal suspensions. In these depot-forming DDSs, the drug is either encapsulated in biodegradable polymers for implantation in subcutaneous (SC) spaces, in reproductive organs, or formulated as drug nanocrystals for either intramuscular (IM) or SC injections. Extensive tests have culminated in the approval of surfactant-stabilized aqueous nanocrystal LA cabotegravir and rilpivirine (CAB and RPV LA) suspensions by the US Food and Drug Administration (FDA). CAB and RPV LA offers the convenience of monthly or bimonthly dosing for HIV-1 treatment [
11,
12,
13].
Multiple randomized clinical trials have demonstrated comparable efficacy between CAB and RPV LA and a standard daily oral regimen [
12,
14,
15]. Based on the data that 91% of prior clinical trial participants preferred LA ART over daily oral medicines, CAB LA proved more effective in preventing HIV-1 infection when compared to daily oral Truvada [
16]. This propelled its US FDA approval in December 2021 [
13]. The remarkable efficacy of CAB LA was linked to improved treatment satisfaction and therapeutic adherence. However, the limitations associated with CAB LA include the need for frequent clinical visits, injection site reactions, variable pharmacokinetics (PK) profiles, prolonged terminal phase tailings, high costs, and co-morbid conditions.
Alternative approaches that include
in situ forming implants (ISFIs), solid implants (SIs), and prodrug nanocrystals are being developed to further improve the PK profiles and other listed limitations associated with CAB LA. Notably, surfactant-coated stearoylated CAB prodrug nanocrystals achieved sustained therapeutic drug levels for up to a year in preclinical models [
17]. Other studies demonstrated that poly(lactic acid-co-glycolic acid) (PLGA) based ISFI could potentially extend CAB dosing intervals to every 6-months [
18]. Other studies have also shown that solid implants containing highly potent ARVs, such as tenofovir alafenamide (TAF) and islatravir (ISL) can simplify ART dosages [2,19-21]. However, in November 2021, Merck announced that participants who received once-weekly ISL plus MK-8507 experienced a decline in CD4 T cells. Further review found that CD4 counts also fell in people taking once-daily ISL plus doravirine for HIV-1 treatment, while those taking ISL alone for pre-exposure prophylaxis (PrEP) experienced a decline in total lymphocyte counts. As a result, studies of ISL for PrEP have been discontinued. These findings underscore the importance of establishing safety profiles of new chemical entities prior to combining them with other therapies. Polymeric TAF implant technologies have demonstrated a zero-order release kinetics of the pharmacologically active tenofovir diphosphate (TFV-DP) metabolite in preclinical models of PK studies [
3]. These include non-degradable materials that provide sustained drug delivery. The use of self-administered microbicides can significantly reduce the risk of HIV-1 infection. Vaginal rings (VRs) that consist of non-biodegradable elastomeric polymers are being developed as self-administrable PrEP treatment targeting at-risk populations [
22]. Several VRs of TAF and dapivirine (DPV), a non-nucleoside reverse transcriptase inhibitor (NNRTI), have demonstrated that such formulations could extend ART dosing intervals [
22,
23,
24]. In two separate phase 3 clinical trials, known as the RING and the ASPIRE studies, DPV VRs demonstrated their effectiveness in reducing HIV-1 transmission. Compared to a placebo ring, the DPV ring reduced transmission rates by 31% in the RING study and 27% in the ASPIRE study [
25,
26]. These results suggest that DPV ring could supplement existing HIV prevention practices. Microneedles (MNs), also referred to as microarray patches (MAPs), are another class of LA DDSs that use drug-diffusible, rate-controlling, non-biodegradable, or slowly biodegradable polymer membranes to extend the apparent half-lives of drugs [
10]. Due to the non-invasive and self-administrable nature, the goal of MNs is to expand LA ART options and to appeal to a wide variety of users that include pediatric populations. Examples include CAB and RPV-loaded MNs that are currently being explored as alternative treatment options to daily oral therapy as well as injectables [
4,
27]. However, further work is required to demonstrate improved or comparable PK and efficacy profiles to the existing CAB and RPV LA injectable formulations.
This review summarizes the physiochemical and biological parameters of the commonly used polymers used for LA DDSs in treatment and prevention regimens for HIV-1 infection. LA ART formulations either in pre-clinical or clinical development are discussed. For each of these delivery systems, polymer compositions and drug release kinetics are discussed. Additionally, an expert opinion section highlights LA ART DDS design considerations which could potentially encourage widespread acceptance and utilization among the key target populations.
2. LA DDSs
The emergence of polymer-based DDSs can be traced back to the 1930s when the implantation of pellets with hydrophobic substances were identified for their ability to facilitate sustained drug release [
28]. Examples include pellets with estradiol for prostate cancer treatment and testosterone pellets for the treatment of hypogonadism [
29]. Later, clinical use of depot-forming formulations loaded with hydrophobic drugs in either water-based or oily mediums, such as procaine penicillin G in water and fluphenazine decanoate in sesame oil, became popular [
30,
31]. Since then, more drug delivery approaches and models have been developed to improve our understanding of the materials and drug release mechanisms. In the 1960s, T. Higuchi presented his renowned "Higuchi model" to describe the drug release kinetics from various sustained release matrix systems [
32]. The model suggested that the release of the solid drugs dispersed in a matrix varies with the square root of time. At the same time, Folkman discovered that silicone rubber could act as a drug reservoir, allowing for constant drug release after implantation. This breakthrough led to the conceptualization of the rate-controlling membranes or reservoir-based implants [
33]. Notable examples of this concept in action are Ocuserts®, which are designed to deliver ocular drugs at predictable and controlled rates [
34].
The challenges associated with non-biodegradable implants, such as the necessity of surgical removal after product life ends and adverse implant site reactions, spurred the development of biodegradable implants. The development and clinical utilization of biodegradable polymers like polylactic acid (PLA), polyglycolide (PGA), and PLGA dates back to the 1970s [
35]. At the early stage of development, clinical applications of these biodegradable polymers were restricted to surgical sutures [
35]. Later on, PLA and PLGA microparticles and pellet depot systems for delivering contraceptive drugs and luteinizing hormone-releasing hormone (LHRH) analogs were developed [
36]. Other notable examples of commercial products where the biodegradable copolymers were utilized include Lupron Depot and Zoladex® implants for treatment of prostate cancer, breast cancer, and endometriosis-like disease [
36]. Furthermore, a polymer based VR was patented by Upjohn Company for sustained drug release [
37]. These developments marked significant milestones in the evolution of polymer-based LA DDSs. Additionally, extensive research was undertaken to develop silicone-based VRs for contraception. In-depth research, primarily sponsored by the World Health Organization (WHO), paved the way for the clinical approval of multiple contraceptive implants in the early 1990s.
The development of LA ARTs began in the early 2000s, with an initial focus on VRs and implants. Later, attention shifted towards nanocrystal formulations[
38]. Clinical trials for the nanocrystal aqueous suspensions started around 2008 (
Figure 1), including phase II trials for HIV-1 prevention (PrEP) with LA RPV [
39]. However, the development of this formulation as a single agent for PrEP faced challenges linked to the high prevalence of NNRTI mutations [
39,
40]. After a decade of research and several clinical trials, the first LA complete therapy for HIV-1 treatment was approved in 2020 [
41].
3. Drug Release Kinetics from LA DDSs
In polymer-based DDSs, drug release refers to a transfer process in which drug molecules are released from the inner core or matrix to the outer surface of the delivery system and eventually into the surrounding environment or tissue [
42]. The rate of drug release from polymer-based DDSs can be modulated by choosing an appropriate polymer with a suitable DDS design. The terminology ‘long-acting’ is the ability to extend the duration of action of a therapeutic agent for a longer period of time. Most often, the terms long-acting, controlled-release, sustained-release, and extended-release are used synonymously [
43]. According to the United States Pharmacopeia (USP), the term ‘extended-release’ or ‘long-acting’ or ‘sustained-release’ is defined as 'a deliberate modification to protract the release rate of an active pharmaceutical ingredient (API) in comparison to an immediate release dosage form' [
44]. In this review, the terminology ‘long-acting’ ‘sustained-’ or ’controlled- release’ are used to designate formulations that can extend the apparent half-lives of drugs. The release of drugs from a delivery system can follow one or more mechanisms, which can be correlated with a number of existing release kinetics models. The commonly used models are zero-order kinetic, first-order-order kinetic, Higuchi, Korsmeyer-Peppas, Peppas-Sahlin, and Hixson-Crowell. The zero-order kinetic model considers that there is no drug concentration influence on drug release rates. The zero-order kinetic models are reflected by the mathematical equation -1, where C
0 and C
t represent the amount of drug at the start and the amount released at time ‘t’.
For first-order kinetics, the rate of drug release is proportional to the concentration of the remaining drug in the delivery system and represented by Eq-2, where dc/dt is the rate of drug release, k1 is the first-order rate constant, and ‘C’ is the concentration of the remaining drug at time, t.
The Korsmeyer-Peppas model (Eq-3) reflects dissolution-mediated drug release. ‘Mt’ and ‘M∞’ is the amount of eluted drug, ‘t’ is the recorded time ‘∞’. The ratio of ‘Mt/M∞’ denoted the fraction of drug release at the time, ‘t’. Kk is the Korsmeyer rate constant.
The Peppas–Sahlin equation (Eq-4) links too the diffusion and relaxation-mediated drug release. The diffusion exponent is represented by ‘m’, and kinetic constants are represented by K1 and K2.
The Higuchi model (Eq-5) is based on diffusion-mediated drug release. Here ‘Q’ indicates the amount of drug released per unit area at time t and KH is the Higuchi constant.
The Hixson-Crowell model (Eq-6) applies to uniform-size drug particles, where the rate of drug release is controlled by drug dissolution. This is governed by the surface area or diameter of the drug-encased particles. The rate of drug dissolution is based on the cube root of the drug mass. The M0 and Mt are the mass of the drug at the initial and at the recorded time ‘t’. KH-C symbolizes the Hixson-Crowell release constant.
An ideal LA DD should exhibit a zero-order drug release kinetics where a constant amount of drug is released per unit time. However, maintaining zero-order kinetics is very challenging, and it majorly depends on the physicochemical properties of the drug and excipients [
45,
46]. Diffusion, osmotic pumping, swelling, degradation or erosion-induced release are the commonly reported mechanisms for polymer-based DDSs.
Non-degradable polymer-based DDSs can be either reservoir or matrix form. In the reservoir form of DDSs, the release rate is governed by the thickness of the polymer membrane and permeability of the drug through the polymer membrane. Whereas, in matrix form of DDSs, Fickian diffusion remains the underlying release mechanism [
47]. Diffusion refers to the random movement of the drug molecules from the higher to the lower concentration region in the matrix. The rate of diffusion in LA DDSs can be described by Fick’s law [
47]. According to this law, the drug release rate depends on the concentration gradient (
) and diffusibility (D) of the drug through the polymer matrix (Equation-7). Particularly for slab-like matrix, Equation-7 can be simply transformed to Equation-8.
……… ….Eq-7;
D is the diffusion coefficient or diffusivity, C is the concentration.
… …………Eq-8;
Mt is the sum of drug released at time t, M0 is the total of the drug-loaded mass, D is the diffusion coefficient, and h is the thickness of the slab-like matrix.
According to Eq-8, the release rate is directly proportional to the drug's diffusibility (D) through the slab and inversely proportional to the thickness (h) of the polymer [
47]. Although, Eq-8 can be transformed to other various forms based on the geometry of the DDSs, the parameters, such as the drug’s diffusibility and slab thickness, play their role in a similar way. For a particular DDS, the release kinetics can be zero-order when ‘h’ and ‘D’ remain constant over time (Eq-8) [
47]. The diffusibility is influenced by the size of the drug molecule relative to the pore size of the matrix. Furthermore, matrix pore size and density have been governed by the properties of the polymer used to fabricate the matrix, such as nature of the monomers and the molar composition used to synthesize the polymer [
48]. Matrix systems lack a rate-controlling membrane, so the diffusion rate is affected by non-constant drug concentration gradient and diffusion distance. Additionally, the diffusion distance is reliant on the polymer's swelling [
48].
In degradable polymer-based DDSs, drug release is majorly controlled by the rate of polymer degradation or erosion and osmotic pumping of the drugs. The chemical degradation of the polymer is influenced by its hydrophilicity. Hydrophilic polymers can absorb water, resulting in an increase in their pore size. This allows them to initiate drug release [
49]. Over time, the polymer undergoes degradation, which increases the number and size of pores, eventually leading to continuous release of drug. Unlike degradation, erosion is an alternative method of the drug release process where polymeric chain segments are dissolved by keeping their chemical structure intact [
48]. Erosion processes can happen either on the surface or bulk, or a combination of both places on the DDS. Surface erosion gradually reduces the size of DDS from the outward to inwards [
50,
51]. In bulk erosion, water permeates the entire bulk of the polymer matrix, leading to a uniform degradation with no significant change in their initial size [
51]. In addition, bulk erosion may produce faster and unpredictable drug release kinetics, making it less favorable for LA DDSs [
52]. Osmotic pumping is another method of drug release, where osmotic pressure drives the influx of water into the non-swelling segment of DDS, resulting in drug release [
53].
Interestingly, the prevalent drug release pattern from LA DDSs is the triphasic rather than monophasic drug release pattern [
45]. The first phase of drug release from an LA DDS demonstrates an initial burst release due to the rapid release of drug molecules located near the surface of the DDS or the surrounding tissue [
54]. The extent of this burst release is influenced by factors such as the design and morphology of the DDS, polymer properties, fabrication process, storage conditions, and homogeneity of the drug-matrix concentration [
54]. While an initial burst release may be desired for rapid drug action, it can significantly reduce the drug depot concentration, consequently impacting the DDS longevity. Strategies such as hydrophobic polymer coating on the outer surface of DDS can curtail this challenge [
54]. The second phase of drug release involves a slow-release period, where the drug diffuses through the polymer matrix. This phase concurrently occurs with polymer degradation in biodegradable systems [
45]. The third phase may exhibit faster release as bulk erosion of the matrix starts. Drug release can also follow a biphasic trend, transitioning from the initial burst release to the zero-order kinetics [
55]. Overall, the kinetics of drug release are complex and influenced by multiple factors. Understanding and optimizing the release profile of DDSs are crucial for achieving desired therapeutic outcomes and ensuring the efficacy of DDSs.
Molecular weight (MW) and the molar ratio of the monomeric units of a polymer play a crucial role in determining its physicochemical properties, including solubility, crystallinity, glass transition temperature (T
g), and mechanical strength. Polymers with less elasticity result in non-deformable matrices and smaller pore formation, leading to a slower drug release [
56]. Moreover, the degradation of the copolymer is influenced by the nature and molar ratio of monomeric units [
57]. For example, a higher molar ratio of hydrophilic monomer, glycolic acid (GA) in PLGA composition leads to its faster biodegradation and subsequent drug release [
58].
Polymer crystallinity refers to the proportion of crystalline and amorphorus region within the polymer structure [
59]. The ratio of different monomeric units in a copolymer affects the polymer's crystallinity and T
g. Most polymers are semi-crystalline in nature, in which the amorphous domains separate the crystalline domains. The water permeability of the polymer is primarily controlled by the percentage of crystallinity. An increased level of polymer crystallinity decreases the overall water permeability, resulting in a slower rate of polymer biodegradation and a decrease in the drug release rate [
60]. In high MW polymers, the influence of crystallinity on drug release depends on the presence of monomer crystallinity [
61]. A notable example is the biodegradability of PLGA, where the ratio of its monomers, lactic acid and GA, determines the degree of crystallinity, subsequent biodegradation and drug release profile. PLLA, composed of L-lactic acid, is highly crystalline, while PDLA, composed of D-lactic acid, is completely amorphous. Like PLLA, PGA is also a highly crystalline polymer. The degree of crystallinity and biodegradability of PLGA copolymer depends on the molar composition of lactic acid and GA comonomers in the copolymer [
62]. Furthermore, the ratio of the amorphous and crystalline regions also influences the polymer's T
g, which in turn affects its mechanical strength and water permeability. For example, a polymer with T
g equal or close to physiological temperature can transform into a rubbery state, facilitating water diffusion and promoting drug release [
48].
6. Conclusion and Future Perspective of la Drug Delivery Systems
The prospects for polymer-based LA formulations are bright, given our rapidly expanding knowledge of polymer properties and drug release mechanisms. While current LA drugs for HIV offer improvement over traditional oral HIV medications, there are user and treatment related gaps that could be addressed by the emerging LA drug delivery technologies such as prodrug approaches and implants. Ideally, the future LA ART medicines should possess three main characteristics.
First, they should be easy to manufacture, scalable, and user-friendly, with a dosing schedule extended at least beyond two months to synchronize dosing with the existing routine patient laboratory test clinic visits. A desirable target product profile is therefore a six-month ART dosage regimen. However, achieving a six-month dosage regimen with the exixting parent drugs is challenging due to their inherent features that include rapid drug metabolism upon absorption from the implant/injection site depot. The constraints on the allowable volume for injections or the size of implantable products, especially for SC or IM delivery, further intensify this challenge. Sometimes, the required drug amount surpasses these permissible limits. A potential solution lies in choosing drugs that are highly potent and have a high resistance to mutations. Prodrug approaches could also be used to achieve this goal by improving intracellular drug delivery and absorption, distribution, metabolism, and excretion (ADME) properties of parent drugs to extend their half-lives. When identifying the optimal drug for LA formulation development, the co-existing medical conditions in patients should also be considered. Given that a significant number of HIV patients (~ 7.5%) are co-infected with hepatitis B or tuberculosis, LA drugs that can concurrently target multiple viruses are especially valuable [
279].
The second desired feature of LA formulations is to minimize post-market complications. This includes curtailing prolonged PK tail in the terminal phase of drug release, inhibiting the emergence of drug-resistant mutations, and ensuring that the treatment can be reversed, especially for newer drugs with no established safety profiles, if adverse reactions occur. For example, the FDA-approved CAB LA exhibits a long PK tail after the last injection [
280]. The CDC recommends that those at risk of HIV continue with oral CAB or PrEP for at least a year after discontinuing CAB LA. Yet, making the shift from LA drugs to oral PrEP isn't always straightforward, as many prefer LA treatments over daily oral ART. There are also concerns about how easily LA treatments can be reversed in the event of complications. These have been circumvented through oral lead-ins prior to administering LA injectable agents. Solid LA implantable devices might offer an alterantive reversal solution, but the need for surgical removal post-lifespan dilutes this advantage.
The third desired feature of LA formulations is their widespread adoption and utilization by the end users. Factors like cost, storage stability, transportation requirements, and specific storage conditions can significantly influence the acceptance and use of LA medicines. Given that HIV is most common in resource-limited settings with limited financial capacity for medication, inadequate cold storage infrastructure, and a scarcity of medical professionals, an LA medicine that is affordable, stable at room temperature, and self-administrable would greatly boost its adoption and application.
Currently, no LA technology, whether in clinical or preclinical development, embodies all three of these ideal features, suggesting that a "one size fits all" approach to LA therapy is inappropriate. Given this, there's a push to tailor LA medicines to the unique needs of specific sub-groups. Understanding the interactions between the polymers, drugs, and their release mechanisms will enable more precise tuning of drug release rates from these formulations. Additionally, to ensure the clinical success of LA ARVs in such resource-limited settings, it is imperative to involve all stakeholders in the early phase of formulation development. These stakeholders include end-users, healthcare professionals, business developers, and policymakers. Collaboration, careful planning, and implementation consideration are essential to prioritize and design the most promising ARVs and LA DDSs that facilitate effective implementation in the most HIV-burdened settings. Moreover, the development of LA ARVs may present regulatory challenges, such as establishing optimal dosing regimens to address the need for combination antiretroviral therapy, and managing missed doses and potential drug-drug interactions in patients with co-morbidities. Some of these challenges are particularly important for PrEP dosing schedules, as there is no precise biomarker to gauge outcomes. In such situations, conducting thorough in-vivo PK studies and evaluating PrEP effectiveness in preclinical animal models becomes imperative. Nonetheless, selecting an appropriate animal model to determine PrEP efficacy remains a significant challenge in preclinical evaluation, given that none of the existing models can fully replicate the human transmission and immunogenicity of HIV infection. Additionally, since LA ARVs can persist in the body for extended periods, ensuring the long-term safety of both the active ingredients and excipients used in LA ARVs in preclinical in-vivo models is of significance. While the excipients used in LA ARVs are generally recognized as safe (GRAS), extensive GLP toxicology studies in relevant models are needed to broaden the usage in special populations such as pediatric patients. The requirement for extensive, extended safety study timelines introduces further challenges, including increased overall product development costs and time. In this context, the use of physiologically based pharmacokinetic (PBPK) modeling can be a valuable tool for predicting drug PK. This approach can also streamline formulation screening and expedite the development of LA products.
Overall, advancements in polymer chemistry and insights into their drug release mechanisms have paved the way for promising strategies to extend HIV-1 treatment dosage regimens beyond two months. Nevertheless, there are developmental and translational hurdles that must be overcome for these products to successfully transition to clinical application.