CC BY-NC-ND 4.0 · Geburtshilfe Frauenheilkd 2021; 81(07): 789-806
DOI: 10.1055/a-1369-9395
GebFra Science
Review

Development of Hormonal Intravaginal Rings: Technology and Challenges

Die Entwicklung hormonhaltiger Vaginalringe: Technologie und Herausforderungen
Fojan Rafiei
1   Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
,
Hadi Tabesh
1   Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
,
Shayan Farzad
2   Department of Biomedical Engineering, University of Southern California, Los Angeles, California, United States
,
Farah Farzaneh
3   Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
,
Maryam Rezaei
1   Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
,
Fateme Hosseinzade
1   Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
,
Khosrow Mottaghy
4   Institute of Physiology, RWTH Aachen University, Aachen, Germany
› Author Affiliations
 

Abstract

Intravaginal rings (IVRs) are minimally invasive polymeric devices specifically designed to be used for the sustained and prolonged release of various type of drugs such as hormones. One of the benefits of using topical drug delivery systems (e.g., IVRs) is the fact that systemic drug delivery may cause drug resistance due to elevated drug levels. Topical drug delivery also provides higher concentrations of the drug to the target site and has fewer side effects. In addition, when a drug is administered vaginally, the hepatic first-pass effect is avoided, resulting in higher absorption. Contraception and treatments for specific diseases such as endometriosis and hormone deficiencies can be improved by the administration of hormones via an IVR. This article aims to classify and compare various designs of commercially available and non-commercial hormonal IVRs and to analyze their performance. Current challenges affecting the development of IVRs are investigated, and proposed solutions are discussed. A comprehensive search of publications in MEDLINE/PubMed and of commercial product data of IVRs was performed, and the materials, designs, performance, and applications (e.g., contraception, endometriosis, estrogen deficiency and urogenital atrophy) of hormonal IVRs were thoroughly evaluated. Most hormonal IVRs administer female sex hormones, i.e., estrogen and progestogens. In terms of material, IVRs are divided into 3 main groups: silicone, polyurethane, and polyethylene-co-vinyl acetate IVRs. As regards their design, there are 4 major designs for IVRs which strongly affect their performance and the timing and rate of hormone release. Important challenges include reducing the burst release and maintaining the bioavailability of hormones at their site of action over a prolonged period of administration as well as lowering production costs. Hormonal IVRs are a promising method which could be used to facilitate combination therapies by administering multiple drugs in a single IVR while eliminating the side effects of conventional drug administration methods. IVRs could considerably improve womenʼs quality of life all over the world within a short period of time.


#

Zusammenfassung

Vaginalringe (IVRs) sind minimalinvasive polymere Vorrichtungen, die speziell für die langfristige und anhaltende Gabe von spezifischen Medikamenten wie Hormonen entwickelt wurden. Der Vorteil eines örtlichen Wirkstoffabgabesystems (z. B. eines IVR) liegt darin, dass die systemische Wirkstoffabgabe wegen des erhöhten Medikamentenspiegels zu einer Medikamentenresistenz führen kann. Hinzu kommt noch, dass eine örtliche Gabe von Medikamenten zu einer höheren Medikamentenkonzentration im Zielgewebe führt und mit geringeren Nebenwirkungen behaftet ist. Außerdem wird durch die intravaginale Gabe von Medikamenten der First-Pass-Effekt vermieden, was zu einer höheren Medikamentenaufnahme führt. Verhütungsmethoden und Behandlungen von bestimmten Erkrankungen wie Endometriose und Hormondefizite profitieren von dem Einsatz hormoneller IVRs. Ziel dieser Studie war es, verschiedene kommerzielle und nicht kommerzielle hormonelle IVR-Modelle zu klassifizieren, zu vergleichen und ihre Leistungen zu analysieren. Es wurden auch die aktuellen Herausforderungen bei der Entwicklung von IVR-Modelle untersucht und grundlegende Lösungsvorschläge ausführlich besprochen. Zunächst wurde eine umfassende Recherche in MEDLINE/PubMed durchgeführt und die Daten kommerzieller IVR-Produkte wurden gesichtet, um danach die Materialien, Modelle, Leistungen und Anwendungsgebiete (z. B. Verhütung, Endometriose, Östogenmangel und urogenitale Atrophie) von hormonellen IVRs eingehend zu untersuchen. Die meisten hormonellen IVRs werden zur Verabreichung von weiblichen Sexualhormonen, d. h. Östrogenen und Gestagenen, eingesetzt. IVRs können anhand ihrer Grundmaterialien in 3 Hauptgruppen unterteilt werden: Es gibt IVRs aus Silikon, aus Polyurethan und aus Ethylen-Vinylacetat-Copolymere. Was die Ausformung angeht, gibt es im Wesentlichen 4 IVR-Modelle, wobei die jeweilige Form sich stark auf die Leistung sowie den Zeitpunkt und die Geschwindigkeit der Hormonfreisetzung auswirkt. Die wichtigsten Herausforderungen bestehen darin, einen plötzlichen Wirkstoffschub zu verhindern, die Bioverfügbarkeit der Hormone im Zielgebiet über einen längeren Zeitraum aufrechtzuhalten und die Herstellkosten zu senken. Hormonelle IVRs versprechen, aktuelle Herausforderungen mithilfe innovativer Entwicklungen zu bewältigen, indem sie den zielgenauen Transport biomedizinischer Wirkstoffe an den Wirkort und dadurch eine Kombinationstherapie aus mehreren Medikamenten in einem einzigen IVR ermöglichen, ohne die Nebenwirkungen von konventionellen Methoden der Medikamentengabe. Auf längere Sicht könnten IVRs innerhalb kurzer Zeit zu einer erheblichen Verbesserung der Lebensqualität von Frauen weltweit beitragen.


#

Introduction

Intravaginal rings (IVRs) are drug delivery systems (DDS) for the systemic or topical administration of one or more drugs [1]. They are doughnut-shaped polymeric devices that can stay in the vagina between 1 and 12 months. This method of drug delivery overcomes many of the challenges associated with more conventional methods, such as gastrointestinal side effects when drugs are administered orally or the hepatic first-pass effect. Another major advantage of IVRs is their ability to deliver drugs at a constant rate. Using IVRs as DDS can eliminate the need for minor surgical procedures required for other implantable drug delivery methods, making IVRs less invasive and more attractive for patients [2], [3].

The first reports on hormonal vaginal rings were published in 1970. The IVR in question was used to deliver the contraceptive drug medroxyprogesterone acetate, but it was not until the 1980s that the first contraceptive trials with vaginal rings were conducted in the United States [4], [5], [6]. Hormonal IVRs are used to deliver many different types of hormones, particularly steroids, for a number of medical reasons including the treatment of urogenital atrophy and contraception [7], [8], [9]. Several IVRs are in commercial use today. NuvaRing® is a hormonal IVR used for contraception which contains a combination of two steroids [10]. Estring® is another hormonal IVR and is used to treat urogenital atrophy [11]. Annovera® is a silicone IVR used for contraception and containing segesterone acetate and ethinylestradiol [12]. Some evaluated IVRs are not being marketed [13].

Sustained release means that a drug is released over a prolonged period of time, while controlled release indicates a predetermined drug release rate resulting in controlled drug concentrations in blood plasma. Such systems mimic drug infusion pumps which maintain stable drug levels in blood plasma by balancing the kinetics of release and elimination. Hormonal IVRs are beneficial in many ways. Using this type of DDS allows a constant and continuous delivery of hormones. Some local (vaginal) side effects of IVRs have been reported such as a slight increase in vaginal discharge [14]. The use of IVRs makes it possible to administer lower hormone doses, leading to fewer systemic side effects from high levels of hormones in the body [15]. In addition, a combination of different hormones to regulate and improve womenʼs menstrual cycles can be administered using IVRs [16]. However, vaginal rings have some drawbacks such as increased vaginal secretions, the potential for unintended IVR expulsion, differences in drug absorption, and cultural sensitivities [17]. Most women who use IVRs report that they do not feel any sexual discomfort when using it. Studies reported no signs of erosion or bacterial invasion in the vaginal tissue of women who used IVRs over a long period of time [14]. Although loss of the ring has not been specifically reported in recent publications, it is to be expected due to the differences in anatomy and 3D structures of the vaginal cavity in females of different ages and ethnicities.

Various types of hormonal IVRs have been designed to date. Selection of the material used to produce the IVR is critical. The rings are made from different polymers such as silicone, EVA, and PU. The design specifications for IVRs vary depending on the application (e.g., matrix IVR, core IVR, sandwich IVR, etc.). The hormones used in IVRs are usually steroid hormones and can either be natural or synthetic. A combination of two or more hormones is used in many IVRs.

The challenges of designing and optimizing IVRs include burst release, costs, and efficacy. This article looks at these general challenges and categorizes the reported solutions. Eliminating surprises when manufacturing IVRs should result in more efficient rings.

To develop optimized, efficient, and novel future applications for IVRs used in combination therapies, it is important to have a comprehensive knowledge and understanding of all types and performances of vaginal rings. The information on hormonal IVRs was collected from various publications and evaluated and summarized in this paper. The purpose of this paper was to evaluate IVRs in terms of their different design specifications and selected materials to suggest a better approach when designing intravaginal rings.


#

Characteristics of Hormonal IVRs

Hormone classification

For many years, hormones were divided into three groups: peptide and protein-based hormones, acid-based hormones and steroid hormones [18]. More recent discoveries have shown that not all types of hormones fit neatly into these categories. Prostaglandins, for example, are made of small fatty acids [19]. Hormones can also be categorized according to their chemical nature, their mechanism of action, or the effects they trigger. As regards their chemical nature, hormones can be divided into two groups, namely, steroid and non-steroid hormones. Non-steroid hormones consist of five different groups: amines, peptides, proteins, glycoproteins, and eicosanoids ([Fig. 1]) [20].

Zoom Image
Fig. 1 Classification of hormones according to their chemical nature.

Another way of classifying hormones is to categorize them as natural or synthesized. Natural hormones are produced in the human body while synthesized hormones are produced by synthesis and their chemical structure is not identical to that of natural hormones. Nowadays, natural hormones can also be produced by synthesis.

Modified amino acids in the body make up amine hormones that include thyroid hormones. Modification of amino acids happens when the carboxyl group of amino acids is removed while the amine group remains intact [20]. Thyroid hormones have two major effects on the body. The first effect is on cellular differentiation and development, while the second effect affects the metabolic pathways [18]. Glycoprotein hormones such as follicle-stimulating hormone (FSH), luteinizing hormone (LH), thyroid-stimulating hormone (TSH), and human chorionic gonadotropin (CG) hormone control and regulate numerous reproductive and metabolic functions in the human body. Stimulation from the hypothalamus affects gonadotropin release more than negative feedback from the ovaries. Glycoprotein hormones have various therapeutic potentials due to their extensive regulation roles [21], [22]. Protein and peptide hormones are both made from amino acid residues. Protein hormones include insulin, glucagon, and somatotropins while oxytocin and vasopressin are considered peptide hormones [20]. Despite the common belief that androgens are purely male sex hormones, androgens are also produced by the ovaries and adrenal glands in women. Dehydroepiandrosterone (DHEA) is one of the most common adrenal steroids in the human body [23].

Some steroid hormones are produced by the human adrenal cortex in response to specific stimuli. Cortisol is a hormone that is secreted in response to stress as is aldosterone, another stress hormone. These hormones play an essential role in regulating the water balance and blood pressure levels [18], [24]. The other group of steroid hormones are the sex hormones, produced in the testes in males and the ovaries in females [20]. The sexual differentiation of males prior to birth and maturation during puberty is prompted by androgens. Functioning of the male genitals in adult men is also maintained by these hormones. The role of androgens in other organs such as the central nervous system, the immune system, and muscle tissue is less recognized [25], [26].

Progestogens and estrogen play an important role in controlling the secretion of gonadotropins in the body. The most important female sex hormones are estradiol and progesterone. Estrone and estriol are less potent estrogens which become more prominent in pregnancy [27].

The hormones most commonly used in IVRs are female sex hormones. The capacity of the vaginal epithelium to absorb steroids is one of the reasons why IVRs are so efficient [28], [29]. Both natural and synthesized hormones are used in IVRs. Progering® is a marketed IVR that delivers progesterone, while NuvaRing contains a synthesized type of progestogen called etonogestrel [30].

Progestogens, levonorgestrel, estradiol acetate, estradiol, segesterone acetate (Nestorone®), ethinylestradiol, etonogestrel, megestrol, norgestrienone, and norethindrone acetate are female sex hormones which are used alone or in combination in various commercially available and non-commercial IVRs.


#

Selection of material

Selecting the proper material for IVRs is one of the most important and critical steps in the modification of the drug release mechanism. Because of how IVRs are applied and the length of time they remain in the body, the characteristics of the IVR material which need to be considered when designing IVRs for the controlled release of drugs are the solubility, diffusion coefficient, initial drug loading and stability of the polymer in addition to the polymerʼs biocompatibility [31].

Certain properties and physical processes which occur when manufacturing IVRs are limiting factors which influence the rate of drug release. Flexibility and transparency are important physical attributes that need to be considered when designing intravaginal rings [32]. Polymer materials are selected to manufacture IVRs because of their flexibility; their use depends on the grade and composition of the polymer [33].

Polymers are a mix of amorphous and crystalline structures, and this affects their level of transparency. With transparency one of the factors determining the choice of materials [34], polymers are a perfect option as the level of transparency can be adjusted. The chains of more transparent polymers are tightly matched [35]. Transparency is an esthetic criterion which can enhance patient compliance [36]. The benefits of using polymeric materials for drug delivery systems are ease of manufacture, flexibility, biocompatibility, transparency, and low cost. Elastomers and thermoplastics are good candidate materials for IVRs. The mechanical properties of IVRs are evaluated based on their function and the duration of drug release. IVRs need to be flexible enough for insertion into the vaginal cavity, rigid enough to stay in place, and soft enough to avoid abrasion to the vaginal epithelium [37], [38], as well as having low vaginal expulsion rates [39], [40]. Polymer structures play a critical role in engineering the controlled release of drugs [41].

The only polymers currently used to manufacture commercially available hormonal IVRs are polyethylene-co-vinyl acetate (EVA, used in NuvaRing), polydimethylsiloxane (silicone used in Estring and Femring®) [41] and polyurethane (PU used in Ornibel®) [31], [42], [43], [44], [45], [46], [47].

Silicone

Silicones are a class of synthetic polymers with a backbone made of repeating silicon-oxygen bonds; the main repeating unit is known as a siloxane. Silicon atoms bond to organic groups, usually methyl groups. The most common silicone is polydimethylsiloxane (PDMS). Other groups such as phenyl, vinyl, and trifluoropropyl may be used as methyl substituents. Silicones are used as fluids, emulsions, resins or elastomers, depending on the concurrent presence of organic groups attached to the inorganic backbone [48].

Because of their flexibility and biocompatibility, silicone elastomers are widely used in medical devices and drug delivery systems and in the cosmetic and food industries [43], [44]. The main characteristics of silicones are chemical stability, low surface energy, and hydrophobicity. Because of their biocompatibility and low toxicity, silicones are widely used in the manufacture of medical products that come into direct contact with the human body [50].


#

PU

Polyurethanes (PUs) were first developed by Bayer in 1937. They are a class of condensed polymers which contain urethanes in their molecular backbone [51], [52], [53]. Polyurethanes are synthesized by a polymerization reaction between isocyanates and polyols in the presence of a catalyst or ultraviolet light activation [54]. Polyurethanes have segmented polymeric characteristics, i.e., hard and soft parts. The hard part is composed of isocyanate components and the soft part of polyol. The mechanical strength of the polymer relies on the hard part and its flexibility depends on the weight percentage of the soft part [53]. Because of its segmented polymeric structure, range of physical properties, and good biocompatibility, PU is used in many biomedical devices including catheters, pacemakers, wound dressings, and drug implants [55].

In some studies, PU was the base material used in the manufacture of vaginal rings, and the sustained drug release through its polymeric chain was measured [31], [41], [56]. Ornibel is a commercially available, polyurethane-based intravaginal ring used for contraception.


#

EVA

Elastomeric polymers such as ethylene-vinyl acetate copolymers (EVA) are a good material for IVRs, due to their rate-controlling properties. EVA is a transparent copolymer made of ethylene and vinyl acetate monomers. The vinyl acetate content plays a key role in determining the mechanical properties, ease of manufacture, and drug release rates from the copolymer [1], [57]. The rigidity of the polymer increases as the level of the vinyl acetate decreases. Reduction of the content also leads to an increase in the relative amounts of amorphous structures and reduces the degree of crystallinity, resulting in a rubbery and more permeable polymer which allows drugs to be released more rapidly from the substrate. Accordingly, the release rate of drug increases with EVA [58], [59].

In addition to these properties, EVA is biocompatible, non-toxic, does not stimulate inflammatory reactions, and can be easily processed. More importantly, the solubility and diffusion coefficient of each drug through the EVA polymeric chain can be tailored by changing the amount of vinyl acetate [60].

Various commercial medical devices make use of the drug delivery properties of EVA. Well-known EVA-based devices include Ocusert® (an ophthalmologic DDS) to treat glaucoma [61], Implanon®/Nexplanon® which are long-acting implants for the continuous release of etonogestrel [63], [64], Vitrasert® (an eye implant containing antiviral agents), and ganciclovir to treat cytomegalovirus infection [65].

Nuvaring is a commercially available contraceptive IVR made from EVA [66]. In some studies, EVA has been used to manufacture IVR prototypes for the sustained release of various active pharmaceutical ingredients (APIs), for example, UC781 (a type of nonnucleoside reverse transcriptase inhibitor used to prevent the transmission of HIV) [67], MIV-150 (a nonnucleoside reverse transcriptase inhibitor), zinc acetate (ZA), carrageenan (CG) and levonorgestrel (LNG) [68]. In some cases, such as UC781, the release rate from EVA-based IVRs was found to be higher than that of silicone IVRs [67].

[Table 1] compares some of physicochemical and biological properties of the three polymers. It shows that silicone, PU and EVA have almost similar physicochemical and biological properties. However, the molecular weight of PU is lower than that of silicone and EVA. In terms of recycling, EVA and PU are more environmentally friendly than silicone.

Table 1 Comparison of important physicochemical and biological properties of polymers generally used for IVR manufacture.

Property

Material

Silicone

PU

EVA

Biocompatibility

✓ [69], [70]

✓ [54], [71]

✓ [57]

Biostability

✓ [49]

✓ [71]

✓ [41]

Reshaping capabilities

✓ [69], [70]

✓ [71]

✓ [72]

Crystallinity

✓ [73]

✓ [74]

✓ [57]

Low cost

✓ [75]

✓ [54]

✓ [70]

Transparency

✓ [76]

✓ [54], [74]

✓ [57]

Low weight

✗ [75]

✓ [54]

✗ [77]

Hydrophilic nature

✗ [78]

✗ [71]

✗ [57], [79]

Recycling

✗ [70]

✓ [54], [71]

✓ [54], [71]


#
#

Design

Delivering steroid hormones via the female reproductive tract bypasses the first-pass hepatic metabolism and provides constant steroid levels, both of which increase the bioavailability of the steroid hormones [28]. The most common DDS that provide sustained and controlled release are osmotic pressure, reservoir (using a rate-controlling membrane [RCM]), and matrix systems [81]. IVRs are designed either in the form of a matrix (homogeneous dispersion), a reservoir or a hybrid (a combination of matrix and reservoir). IVRs can also be used as a casing for vaginal tablets or rods, and are then referred to as insert IVRs. [Fig. 2] shows a comprehensive classification of these four types of IVR design which are commercially available or have been proposed in the literature.

Zoom Image
Fig. 2 Intravaginal ring designs.

The cross-sections of the various types of IVRs shown in [Fig. 2] are displayed in [Fig. 3], which differentiates between the types of drug distribution through the polymeric substrates.

Zoom Image
Fig. 3a Single-drug matrix IVR, b multi-drug segmented matrix IVR, c single-drug core IVR, d single-drug incomplete core IVR, e multi-drug continuous core continuous RCM, f multi-drug segmented core continuous RCM, g multi-drug segmented core segmented RCM, h sandwich IVR, i tablet/pod/rod insert IVR, j core-matrix (hybrid) IVR.

Matrix (homogeneous dispersion) IVR

Insoluble

A single injection of an elastomer mix containing drug molecules that are dispersed homogeneously in the matrix of the polymer results in a matrix IVR. The solubility of the polymer is lower than the loaded drug in insoluble matrix IVRs [1]. The entire surface area of matrix IVRs is exposed to vaginal tissue/fluid. In the first stage, burst release occurs due to the presence of immobilized drug particles separated from the crystal lattice at the surface of the intravaginal ring. This results in the creation of a concentration gradient that drives the release process thermodynamically. In the second stage, the drug molecules closest to the surface of the ring are diffused through the polymer and released into the medium (i.e., the vaginal fluid and/or interstitial fluid of the vaginal tissue). As the drug release continues, the unmedicated surface is separated from the inner drug-loaded segment of the matrix ring by the creation of a drug exhaustion zone. As the thickness of the unmedicated zone increases, the surface area of the inward-moving depletion border decreases. Consequently, the pathway from where the remaining drug is diffused increases as the amount of the released drug reduces over time and the drug near the surface of the ring becomes depleted. In vitro, the release from a matrix IVR under sink conditions can be explained by the Higuchi equation ([Eq. 1]) [2], [82], [83].

Q=sqrt[D_p(2A-C_p)C_pt]Zoom Image

Where Q denotes cumulative release per unit area, Dp signifies the drug diffusion coefficient of the polymer, Cp represents drug solubility in the polymer, A designates drug loading per unit volume, and t is time.

Insoluble matrix IVRs can be designed as either multi or single-drug systems. Multi-drug insoluble matrix IVRs can be segmented or continuous. Segmented IVRs are designed for multiple drugs using multiple polymers, while in continuous multi-drug IVRs, different drugs are homogeneously dispersed using one type of polymer. Segmented multi-drug IVRs, shown in [Fig. 3 b], provide additional advantages, including the ability to control the release rate of each segment individually, compared to continuous multi-drug IVRs [1], [84].


#

Bio-soluble

Silicone elastomers, EVA, and PU are biostable polymers which are used to create insoluble homogeneous dispersion matrix IVRs. However, scientists have proposed using certain bio-soluble biocompatible polymers to manufacture IVRs. In a study by Vartiainen et al., matrix rings manufactured from bio-soluble styrene-butadiene block copolymers were tested for their capacity to deliver 17β-estradiol to treat postmenopausal symptoms [85]. Bio-soluble polymers can control the release of drugs by means of degradation mechanisms instead of diffusion. This enables the delivery of both hydrophilic and hydrophobic APIs, including those with large molecular weights, to vaginal tissue. In another study by Han et al., non-hormonal intravaginal rings were manufactured from bio-soluble acacia gum and sodium methacrylate to deliver a combination of antiretroviral HIV microbicides for 28 days [84], [86]. This type of IVR can be manufactured as a multi or single-drug device, as shown in [Fig. 2] [1].


#
#

Reservoir IVR

Core

During the manufacture of core IVRs, a drug is placed in a central zone and surrounded by a drug-free polymeric rate-controlling membrane (RCM). The drug carrier at the core of a reservoir IVR can be either a polymer (the same as the RCM or a different polymer) or another type of biomaterial, e.g., paraffin. Core type IVRs can be designed and manufactured as either single or multi-drug delivery systems. Multi-drug reservoir IVRs can potentially be designed in one of three ways, i.e., as continuous core continuous RCM; segmented core continuous RCM; or segmented core segmented RCM. The core part of reservoir IVRs can vary in length. Depending on the application of the IVR and the amount of the drug required for treatment, the full core can be divided into smaller core lengths ([Fig. 3 c, d, and f]) [1]. The molecules of the drug have to first detach themselves from the core crystal lattice, then diffuse into the drug-free RCM and then disseminate into the medium surrounding the IVR. The drug is constantly released until the drugʼs concentration at the core is depleted. One advantage of core IVRs is that release of drugs occurs in zero order compared to matrix IVRs. Aside from that, the release characteristics of this type of IVR can be easily modified. Modifications can be achieved by changing the thickness of the RCM (h) or by varying the length of the core (L). A reduction in RCM thickness leads to an enhanced release rate, due to the shorter diffusion pathway [2], [87].

Q = (D_p C_p t) ∕ hZoom Image

The ratio of core to RCM diameter (b/a) affects the cumulative release rate and is represented by Crankʼs equation [88].

Q = (2pi D_p C_p L t) ∕ ln(b/a)Zoom Image

Where L denotes core length, a represents the cross-sectional diameter of the core, and b signifies the cross-sectional diameter of the RCM.

As equation 3 shows, when the core length decreases, the rate of drug release also decreases. This type of IVR can be manufactured either by reaction injection molding or by using an extrusion process. A burst release may be observed in the first few days, especially in condensation-cured silicone IVRs, due to the presence of solid drugs in the RCM [2], [89].


#

Sandwich (shell)

In sandwich IVRs a layer containing the drug is located below the surface of the IVR. This layer can be located between a nonmedicated polymer surface and a central core that may or may not contain a second drug ([Fig. 3 h]). Since the drug layer is located a fraction of a millimeter below the surface, this particular IVR is suitable for delivering drugs with poor polymer diffusion characteristics. The manufacture of shell IVRs is less costly due to the lower drug concentrations in the drug layers compared with matrix-type IVRs where the drug is homogeneously dispersed throughout the whole IVR. As shown in [Fig. 4], a sandwich IVR can minimize side effects due to the constant drug release compared to other types such as matrix and core IVRs [2], [90]. These types of reservoir IVRs can also be manufactured as both multi or single-drug delivery systems. Multi-drug sandwich IVRs can be designed as continuous core continuous RCM, segmented core continuous RCM, and segmented core segmented RCM [1].

Zoom Image
Fig. 4 Cumulative release profile of a matrix (homogeneous dispersion) IVRs, b core reservoir and pod IVRs, and c sandwich reservoir IVRs.

#
#

Insert IVR

Tablet and rod

These types of IVRs are manufactured as nonmedicated polymeric bodies with cavities in which multiple drug-loaded vaginal rods or tablets can be inserted. Rods can be manufactured using several different methods. For example, a dispersed mixture of a particular type of polymer and drug can be injected into a PVC tubing using a plastic syringe. The PVC tubing is then removed and the polymeric rod can be cut into suitable sizes for insertion into the body of the ring [91]. These types of insert IVRs can be used as single or multi-drug delivery systems by the insertion of different tablets or drug-loaded rods into a single IVR.


#

Pod

Single or multi-drug release is achievable using IVRs with several small drug-containing sectors, called pods, which can be loaded with the same or different drugs [2]. Although matrix IVR designs have been developed for clinical trials, pod IVRs can provide sustained drug release over longer periods as indicated by the drug release profile shown in [Fig. 4 b] [92].

Pods are made from polymers containing homogeneously dispersed drugs coated with a layer of polymer to create a drug pod. These pods, which can have identical or different geometries, are then incorporated in a nonmedicated continuous or segmented IVR. The release rate of pod IVRs is controlled by their nonmedicated polymeric membrane, the characteristics of the sectors, and the number of pods in each IVR [1], [93], resulting in an independent delivery of drugs in pseudo-zero order from each pod. Pod design also eliminates the initial burst release, which is one of the challenges associated with other IVR designs [93]. The release profile of pod IVRs is similar to that of core IVRs [94]. The safety of pod IVRs has been investigated in both rabbits and humans [95], and pod IVRs have been shown to be a harmless and efficient drug delivery system [96], [97]. Pod IVRs can be manufactured for the delivery of single or multiple drugs.


#
#

Hybrid IVR

Using an IVR which is a combination of conventional IVR types, i.e., matrix, reservoir, or insert, is a novel approach suggested by current research. It could be considered a hybrid IVR. It has been proposed that core-matrix hybrid IVRs could be manufactured using a core embedded in a hot-melt extruded polymer containing the active agent [1], [68]. Another possible design for a hybrid IVR could be an insert-matrix IVR consisting of a medicated polymeric body with cavities for the insertion of vaginal rods, tablets, or pods ([Fig. 3 j]).

The drug release profiles of the major IVR types (matrix, core, and sandwich) are shown in [Fig. 4].

Some studies have reported on the in vitro cumulative release profiles of matrix and reservoir [12], [68]. Another study has discussed the release profile of a reservoir-type IVR releasing different amounts of drug [98].

Commercially available and non-commercial rings are listed in [Tables 2] and [3].

Table 2 List of commercially available hormonal IVRs.

IVR Name

Company

Hormone

Release rate

Application period

IVR material

IVR type

Application

Ref.

ANNOVERA®

Therapeutics
MD

Nestorone (segesterone acetate) and ethinylestradiol

segesterone acetate: 150 µg

ethinylestradiol: 13 µg

1 year

silicone elastomer

pod

contraception

[99], [100], [101]

Estring

Pfizer Inc., USA

Estradiol

7.5 µg/day

90 days

silicone elastomer

core reservoir

urogenital atrophy

[4], [84]

Femring

Warner Chilcott/
Actavis, UK

estradiol acetate

0.5 µg/day

3 months

silicone elastomer

core reservoir

urogenital atrophy, estrogen replacement therapy (ERT), contraception

[30], [84], [102], [103]

Fertiring®

Silesia Laboratories

progesterone

variable

3 months

silicone elastomer

insoluble matrix

contraception

[80], [84], [104]

NuvaRing

NV Organon Co in the Netherlands/Merck & Co., USA

etonogestrel and ethinylestradiol

8 µg/day

21 days

EVA

core reservoir

contraception

[84], [105], [106]

Ornibel

Exeltis Healthcare

etonogestrel and ethinylestradiol

8 µg/day

21 days

PU core and EVA external membrane

core reservoir

contraception

[105]

Progering

Silesia Laboratories

progesterone

10 mg/day

90 days

silicone elastomer

insoluble matrix

contraception

[80], [84], [103]

Table 3 List of non-commercial hormonal IVRs.

Hormones

IVR materials

IVR type

Application

Release rate

Intended duration in situ

Ref.

Levonorgestrel

silicone elastomer

core reservoir

contraception

20 µg/day

90 days

[107]

Nestorone

elastomer LSR (liquid silicone rubber)

core reservoir

contraception

50, 75, or 100 µg/day

6 months

[108]

Nestorone and ethinylestradiol

silicone elastomer

rod insert

contraception

10, ~ 13.5 and 7.5 µg/day

1 year

[109], [110], [111]


#
#
#

Hormonal Ring Applications

Contraception

Contraceptive vaginal rings have been the focus of many gynecological studies for more than 40 years [28], [29].

Steroids, whether alone or in combination, diffuse at a constant rate through the ring and are directly absorbed through the vaginal epithelium into the systemic circulation. Progestogens are not normally used alone in such contraception methods as skin patches, rings, pills, etc. A combination of hormones, for example a combination of estrogen and progestin [112], etonogestrel/ethinylestradiol (ENG/EE) [28], segesterone acetate/ethinylestradiol (SA/EE) rings, LNG/E2, norethindrone acetate (NETAc)/EE IVRs [28], and norelgestromin/ethinylestradiol skin patches [113] and levonorgestrel/ethinylestradiol pills [114] are preferably used in contraceptive DDS. Since progesterone is one of the most important hormones in the menstrual cycle, it has significant uses in contraception. The cyclic release of luteinizing hormone is inhibited by the increased concentration of progesterone in nonpregnant women, with production of the follicle-stimulating hormone inhibited by higher levels of progesterone [115]. There are two methods by which progestins prevent pregnancy:

  1. by preventing sperm from reaching the egg by changing the cervical mucus,

  2. by preventing implantation of the fertilized oocyte through changes to the uterine lining [116].

A combination of etonogestrel and ethinylestradiol are used in contraceptive vaginal rings. Contraceptive IVRs containing etonogestrel (a progestin) and ethinylestradiol (an estrogen) function by preventing ovulation [105].

Contraceptive IVRs have several advantages compared with other conventional contraceptive methods: convenience of use, fewer premenstrual symptoms, lighter and more regular menstrual cycles, as well as reducing the risk of certain cancers such as breast cancer because of the lower hormone doses [118].

Three rings containing only progestogens have been evaluated. These rings contain progesterone, levonorgestrel (LNG), and the progestin Nestorone (NES). Progestogen-only rings are specifically designed for continuous use compared to combination drug IVRs which are designed for cyclic use (3 weeks in/1 week out) [28].

NuvaRing, a commercially available contraceptive IVR, contains a combination of etonogestrel and ethinylestradiol [16], [119], [120]. These APIs are loaded in an EVA core with an outer diameter of 54 mm and a cross-section of 4 mm [121]. Using NuvaRing for the recommended 3-week period completely inhibits ovulation with an efficacy of 98% [112], [122].

Progering is another contraceptive ring, manufactured by Silesia SA Laboratories, Chile. A silicone elastomer is used in the manufacture of this ring, and progesterone is homogeneously dispersed inside the matrix IVR. The cross-sectional diameter of Progering is 8.4 mm and the overall diameter is 58 mm. Progesterone is released at an average rate of 10 mg per day over three months [115], [123]. Other commercially available contraceptive IVRs are listed in [Table 2].

Research is being conducted into a contraceptive IVR containing Nestorone and ethinylestradiol. The drugs are loaded into two rods, with one rod containing only Nestorone while the other contains both drugs. This ring can be used for 1 year, which makes it an efficient contraceptive method [110], [124]. Some of the other non-commercial contraceptive IVRs which are currently being developed are presented in [Table 3].


#

Endometriosis

Endometriosis is an estrogen-dependent medical condition caused by the ectopic growth of endometrial tissue in women of reproductive age. It occurs when tissue similar to the lining of the uterus or endometrium forms outside the uterine cavity [125], [126]. Endometriosis is usually found in the pelvic cavity and can attach to any of the female reproductive organs such as the uterus, fallopian tubes, ovaries, uterosacral ligaments, peritoneum, or any of the spaces between the bladder, uterus, bowel, vagina, and rectum [15], [125], [128]. The diagnosis of endometriosis in primary care is mainly clinical. There is currently no cure for endometriosis, so medical care has focused on pain reduction. The pituitary gland releases gonadotropins, hormones that stimulate the production of gonadal steroids such as estrogen which aggravate the symptoms of endometriosis and uterine fibroids [15].

Elagolix is a gonadotropin-releasing hormone (GnRH) antagonist that reduces gonadotropin levels, which in turn suppresses estradiol. This decreases dysmenorrhea, dyspareunia, and endometriosis-related pain [127]. Initial treatment consists of the administration of common agents used for primary dysmenorrhea, such as nonsteroidal anti-inflammatory drugs (NSAIDs), estrogen/progestin combination contraceptives, or progestin-only contraceptives [131]. There is some evidence that these agents are helpful and have few adverse effects. NSAIDs are often the first-line treatment for endometriosis, followed by hormone therapy [132]. Hormonal treatments that decrease estrogen and progesterone production are used to treat endometriosis [127]. The inhibition of aromatase in premenopausal women reduces estrogen production, resulting in counter-regulation by the pituitary. This eventually leads to an increase in gonadotropin levels and stimulates follicular development, which may lead to the formation of ovarian cysts. This can be a drawback when treating premenopausal women [131]. Vaginally administered ethinylestradiol and etonogestrel can be used either for contraception or to treat dysmenorrhea resulting from endometriosis [110].

IVRs are an ideal method to treat women suffering from endometriosis. The use of IVRs instead of other conventional therapies for endometriosis-associated pain such as transdermal patches was observed to be more effective as they were better at reducing dysmenorrhea. IVRs were noticeably more accepted by patients because of their convenience of use, which resulted in higher user satisfaction [133]. There are currently no IVRs to treat endometriosis on the market; however, research is being conducted into the use of hormonal IVRs to treat this disease. An IVR containing a combination of the aromatase inhibitor anastrozole and progestin has been developed for the treatment of recurrent endometriosis in women of reproductive age [127], [131]. In another study, an ethinylestradiol and etonogestrel-loaded IVR, which inhibits ovulation, was observed to be effective to treat women with endometriosis, dysmenorrhea, and polycystic ovarian syndrome [110].


#

Estrogen deficiency

A decline in the levels of the primary sex hormone in women (estrogen) is referred to as estrogen deficiency. Lower estrogen levels in the body cause changes to the breasts, genitals, urinary tract, and even the skin [134], [135].

Oral estrogens are not absorbed effectively, and about seventy percent is metabolized during the first pass through the liver [2], [136]. In addition to the gastrointestinal side effects of oral estrogen, a further challenge of administering estrogen orally is bolus estrogen doses which result in undesirably high levels of estrogen after each dose and the necessity of administering recurrent doses. Transdermal drug delivery systems such as skin patches are the most commonly used estrogen replacement therapies and reduce the risks associated with high doses of estrogen such as thromboembolism. However, studies have reported that excessive estrogen exposure is higher when using transdermal drug delivery systems compared with IVRs or low-dose oral contraceptives [133]. Side effects of high estrogen doses include breast and endometrial cancer, heart attacks, blood clots, and stroke [137], [138]. Some of the methods used to ensure constant estrogen levels in the body are aggressive and require surgical insertion and removal. More importantly, these methods are not able to provide suitable doses of estrogen over a long period [139].

Vaginal estrogen therapy has many advantages such as a direct vaginal impact, the high bioavailability of the administered drugs, the option to use low doses of drugs compared to oral and parenteral administration, higher acceptance by patients, and fewer side effects such as endometrial stimulation, hyperplasia, breast tenderness, and adenocarcinoma [140], [141]. Among the estrogens, 17 β-estradiol (E2), is often used to treat estrogen deficiency. This specific type of estrogen compensates for a deficiency of endogenous estrogens caused by ovarian failure. In comparison with other natural estrogens, E2 has proven to be more efficient for estrogen replacement therapy (ERT) and hormone replacement therapy (HT). E2 has fewer side effects, specifically prevents blood clots and decreases the risk of venous thrombosis. This type of natural estrogen is broadly used to treat estrogen deficiency-associated disorders such as urogenital atrophy, and menopausal and postmenopausal symptoms [139].

ERT was long considered the cornerstone of therapy for postmenopausal women with osteoporosis [142]. Studies in vitro and in vivo have supported the hypothesis that estrogen works by slowing bone resorption by preventing the production of pro-osteoclastogenic cytokines mediated by osteoblast lineage cells. This impedes the formation of osteoclasts and also increases bone mineral density [143], [144], [145]. In one study, women who underwent estrogen replacement therapy for longer than 1 year after menopause had an 80% lower incidence risk of Alzheimerʼs disease compared to women who did not have the therapy [146].

Vaginal creams, tablets, and rings deliver drugs via the vagina, and so far, three creams, two rings, and one vaginal tablet have been approved by FDA for the treatment of urogenital atrophy [147], [148]. Estrace [149], Premarin [150], and Estragyn [151] are commercially available vaginal creams; Vagifem [152] is the only commercially available vaginal tablet. Estring [153] and Femring [154] are two commercially available vaginal rings to treat urogenital atrophy.

Many patients believe that vaginal creams and tablets are hard to use and sometimes forget to apply the cream or tablet, which needs to be administered at specific times. Other disadvantages of using creams or tablets are their low and uneven absorption capacity. Therefore, a long-acting DDS is ideal for releasing hormones such as estradiol, estriol, and conjugated estrogens [155], [156], [157], [158], [159]. Estring and Femring vaginal rings, which offer sustained and controlled release of hormones, provide such conditions.

The risks of systemic estrogen or estrogen plus progestogen administration include stroke, venous thromboembolism, and invasive breast cancer. Low-dose vaginal estrogens have been approved by the FDA. They bypass the hepatic first-pass effect and there is no evidence that they cause the above-mentioned side effects, but they carry the same warnings about health risks (e.g., breast and endometrial cancer, cardiovascular disorder and probable dementia) as estrogens administered systemically [160]. Estradiol levels which are higher than postmenopausal levels lead to endometrial proliferation, which does not occur when vaginal estrogens are used. However, if there is a systemic absorption of estrogen it is suggested that progestogens should be used simultaneously [161].

Urogenital atrophy

Menopause usually occurs in women between the ages of 49 and 52, when the egg is no longer released from ovaries [142]. Estrogen levels play an essential role in maintaining the mucopolysaccharide and collagen content of the mucosa and in preserving the thickness and moisture of urogenital tract tissue. Changes in the structure and function of the genitourinary system after menopause are the result of decreased estrogen levels. The reduction of estrogen levels leads to physiological, histological, and anatomical changes to the genitourinary system, also known as urogenital atrophy [135], [162]. Effects of estrogen deficiency include thinning of the vaginal epithelium, decreased vaginal elasticity and blood flow, and decreased secretion by the Bartholin glands, resulting in damage to the vaginal mucosa [163]. Symptoms of urogenital atrophy include genital symptoms such as dryness, burning, and vaginal irritation; sexual symptoms such as painful intercourse and postpartum hemorrhage; and urinary symptoms including nocturia, recurrent urinary tract infection, urinary incontinence, urinary urgency, etc. In total, these symptoms are referred to as the genitourinary syndrome of menopause [135], [147], [164], [165], [166], [167].

HT treats urogenital atrophy by increasing estrogen levels in the body. There are three ways to administer the drug: oral, transdermal, and intravaginal [168]. FDA-approved estrogen-loaded IVRs are suitable to treat postmenopausal urogenital atrophy and lower urinary tract symptoms. These IVRs are usually loaded with low dosages of estradiol and may not need attendant progestogens [155], [158], [168]. However, symptoms of urogenital atrophy can also occur in women with low estrogen levels for other reasons, for example, due to breastfeeding or drugs that lower estrogen levels [19]. The treatment of urogenital atrophy is highly dependent on the symptoms of the disease. Initially, non-pharmacological treatments can be used; for example, sexual activity can help maintain vaginal health during menopause [169]. Vaginal estrogen therapy can lead to higher levels of estrogen in serum; however, they will not exceed normal postmenopausal ranges. Moreover, high systemic levels of estrogen can have side effects such as venous thromboembolism and stroke [160].

Estring is a commercially available 17 β-estradiol-loaded IVR developed to treat urogenital atrophy. The IVR is designed to release a low dose of estrogen continuously for 3 months [2].

Vaginal estradiol therapy can increase the risk of recurrence in patients diagnosed with breast cancer. The use of estradiol-containing vaginal rings and tablets results in a significant increase in estradiol serum levels, indicating systemic absorption in the first few weeks of HT [170].

The schematic diagram of hormonal IVR applications is demonstrated in [Fig. 5].

Zoom Image
Fig. 5 Diagram showing applications for IVRs.

#
#
#

Current Challenges and Possible Solutions

Reduction of burst release

One of the most important challenges complicating the medical use of intravaginal rings is burst release due to Fickian or non-Fickian drug diffusion. In addition to the negative economic effect of higher doses, burst release can have unwelcome side effects [171].

A number of studies used membranes made of different polymers as coatings for different types of drug delivery devices [17]. One example of this is Nuvaring, which has a membrane of ethylene-vinyl acetate copolymer (EVA) surrounding an EVA core (with a different vinyl acetate content) impregnated with etonogestrel and ethinylestradiol [14]. A high burst release rate of 20 – 40 percent was observed during in vitro tests of IVRs manufactured from EVA. Cellulose membranes are employed to prevent the initial burst release in IVRs. The release rates of EVA rings can be efficiently controlled using membranes [172].

The release rate can be adjusted to specific required values by modifying the membrane thickness. Membranes have specific permeabilities which control drug release in a DDS. As the membrane becomes thicker, the release rate of the drugs decreases. In summary, the goal is to delay the diffusion of API through the polymeric device to reduce the amount delivered [173].

Another method for a controlled DDS is to manufacture a system that releases the loaded drug at a prearranged time or in pulses. Such a system can also be manufactured in response to environmental changes [174].


#

Long-term drug delivery

The drug delivery times of currently available contraceptive IVRs range from three weeks to one year [175], [176].

The primary argument for a longer delivery duration is to make the product more affordable. Microbicide IVRs are therefore designed for one to three monthsʼ use [177]. The impact of the duration of IVR use on user acceptance, device performance, and biocompatibility has not yet been comprehensively reported and is worth further evaluation.

A long-term polymeric DDS can be created by many methods, including osmotic pumps [178], matrices with controllable swelling [179], diffusion [180], erosion [181], different drug-loading profiles [182], [183], [184], and the use of multilayered membranes as matrices [174], [185]. As regards diffusion, drug release increases when the drug has a higher diffusion coefficient [186]. A diffusion barrier such as an empty polymeric barrier can be used to create a long-term DDS [187]. Swelling of the polymer results in an increase in the length of the diffusion pathway. The drug release rate decreases due to a reduction in the drug concentration gradient [53]. Drug delivery systems designed for long-term use should have low erosion and degradation rates. To achieve this goal, water-repellent surfaces can be used to manufacture such systems [55].


#

Sustained bioavailability

Drug bioavailability varies as it depends on factors such as the drugʼs solubility in the elastomer and vaginal fluid, the drugʼs diffusion coefficient in the elastomer, the volume of vaginal fluid, the drugʼs partition coefficient between the IVR and the vaginal fluid and tissue, the rate of diffusion and drug removal through vaginal tissue, and the rate of anterior to posterior advection of the vaginal fluid [188].

One method of achieving sustained bioavailability is to design drug delivery systems that respond to environmental changes. These systems change their drug release rate according to different stimuli such as light, temperature, specific molecules, mechanical forces, etc. to which they are exposed. Drug delivery systems can also be designed to respond to environmental changes. These are all methods which can be used to manufacture controlled drug delivery systems with reduced burst release [174].


#

Maximizing efficacy

Increasing the bioavailability of a drug can increase its efficacy. However, if the drug is lipid-soluble, as most hormones are, increasing bioavailability can be difficult. The Population Council is collaborating with HRA Pharma (Paris, France) and the NICHD to develop a 3-month contraceptive IVR which releases a progesterone receptor modulator. The aim is to provide an estrogen-free contraceptive that does not require daily oral intake. Ulipristal acetate, a derivative of 19-norprogesterone, binds specifically to the human progesterone receptor [8], [189]. One study reported inhibition of ovulation in 68% of the cycles studied and concluded that further testing with higher release rates of ulipristal acetate was required to achieve ovulation suppression in a higher percentage of cycles [189].

Using microbicides that contain one or more APIs is another way to increase the efficacy of IVRs. In some studies, drug levels have been shown to correlate best in tissue with microbicide efficacy [190], [191], [192], [193].

Choosing a suitable polymer for IVRs based on the polymerʼs solubility and stability and the required API release rate is another way to maximize the efficacy of IVRs [193].


#

Cost reduction

Price is one of the parameters at the commercialization stage. The price of the rings is affected by the choice of materials, method of manufacture, and type of loaded drug. Using different low-cost techniques can help reduce the costs of manufacturing drug delivery systems [194], [195]. Using low-cost materials such as bio-based polymers is another way to manufacture more economical drug delivery systems [194] ([Table 4]).

Table 4 Major challenges in development of hormonal IVRs and proposed solutions.

Challenge

Possible solutions

Reduction of burst release

  • Additional membranes

  • Modification of membrane thickness

  • Drug released at a prearranged time

Long-term drug delivery

  • Osmotically driven pumps

  • Matrices with controllable swelling

  • Diffusion rates

  • Erosion rates (using water-repellent surface)

  • Non-uniform drug-loading profiles

  • Multilayered matrices

Sustained bioavailability

  • Changes in the drug release rate when faced with different stimuli

Maximizing efficacy

  • Using selective hormone receptors

  • Using microbicides

  • Using the appropriate polymer for the required API

Cost reduction

  • Low-cost techniques

  • Low-cost materials such as bio-based polymers


#
#

Future Direction

Intravaginal rings are promising methods for the prolonged delivery of hormones for various purposes. IVRs are currently mainly used for contraception; however, they can also be used to treat diseases such as endometriosis and urogenital atrophy and for estrogen therapy. IVRs will soon also be used in combination therapies, e.g., for HIV prevention and contraception, making IVRs a highly efficient treatment for women alongside implants. In addition to hormones, other drugs can also be delivered systemically through the vaginal tract. Although there are many commercially available IVRs on the market, there are still some challenges which must be overcome for IVRs to become more efficient. One major challenge is to make IVRs that are able to deliver different types of drugs for a long period of time. Other challenges include manufacturing IVRs that are low-cost, as this will make them accessible for larger female populations. Solving these challenges will make IVRs the most efficient drug delivery system, not only specifically for female diseases but to deliver drugs that need to be systemically absorbed.


#

Conclusion

Hormonal intravaginal rings are highly efficient systems for delivering hormones. The advantages of IVRs are their efficacy in delivering drugs, their ability to bypass the hepatic first-pass effect, prolonged drug release, ease of administration, and a reduction in the number of required appointments with a physician. However, women will still need to see a physician to obtain a prescription and guidance on how to use IVRs. The sustained release of hormones from IVRs and the possibility of using multiple hormones and drugs at the same time can be highly efficient for many patients. Silicone elastomer is one of the most commonly used polymers for the manufacture of different drug delivery systems as it is highly biocompatible. Therefore, silicone is probably the most efficient polymer for IVRs. Various IVR manufacturing designs such as pod, sandwich, and core are used for different therapeutic purposes to maximize the efficiency of the IVR.

Hormonal IVRs are among the most efficient and effective methods to treat a wide range of diseases. These novel drug delivery systems can enhance womenʼs lives in a number of ways and improve their quality of life.


#

Informed Consent

Informed consent was not required for this study.


#

Funding

External funding was not required for this investigation.


#
#

Conflict of Interest

The authors declare that they have no conflict of interest.

  • References

  • 1 Malcolm RK, Boyd PJ, McCoy CF. et al. Microbicide vaginal rings: Technological challenges and clinical development. Advanced Drug Delivery Reviews 2016; 103: 33-56
  • 2 Malcolm RK. The intravaginal ring. Drug Pharm Sci 2003; 126: 775-790
  • 3 Hussain A, Ahsan F. The vagina as a route for systemic drug delivery. J Control Release 2005; 103: 301-313
  • 4 Murphy DJ, Boyd P, McCoy CF. et al. Controlling levonorgestrel binding and release in a multi-purpose prevention technology vaginal ring device. J Control Release 2016; 226: 138-147
  • 5 Duncan GW. Medicated devices and methods. In: Google Patents; 1970
  • 6 Schopflin G. Drug excipient of silicone rubber. Google Patents; 1977
  • 7 Guthrie KM, Rosen RK, Vargas SE. et al. User evaluations offer promise for pod-intravaginal ring as a drug delivery platform: A mixed methods study of acceptability and use experiences. PloS One 2018; 13: e0197269
  • 8 Jensen J, Edelman A, Chen B. et al. Continuous dosing of a novel contraceptive vaginal ring releasing Nestorone® and estradiol: pharmacokinetics from a dose-finding study. Contraception 2018; 97: 422-427
  • 9 Keller M, Ray L, Atrio JM. et al. Early termination of a phase 1 trial of tenofovir disoproxil fumarate vaginal ring. Contraception 2018; 1: 20
  • 10 Nave R, Jalkanen T, Talling C. et al. The Effect of Drug Content Reduction on the In Vitro and In Vivo Properties of Levonorgestrel-Releasing Intravaginal Rings. J Pharm Sci 2018; 107: 1020-1027
  • 11 Fetherston SM, Boyd P, McCoy CF. et al. A silicone elastomer vaginal ring for HIV prevention containing two microbicides with different mechanisms of action. Eur J Pharm Sci 2013; 48: 406-415
  • 12 Boyd P, Variano B, Spence P. et al. In vitro release testing methods for drug-releasing vaginal rings. J Control Release 2019; 313: 54-69
  • 13 Thurman AR, Schwartz JL, Brache V. et al. Randomized, placebo controlled phase I trial of safety, pharmacokinetics, pharmacodynamics and acceptability of tenofovir and tenofovir plus levonorgestrel vaginal rings in women. PloS One 2018; 13: e0199778
  • 14 Lete I, Cuesta MC, Marín JM. et al. Vaginal health in contraceptive vaginal ring users–A review. Eur J Contracept Reprod Health Care 2013; 18: 234-241
  • 15 Buggio L, Lazzari C, Monti E. et al. “Per vaginam” topical use of hormonal drugs in women with symptomatic deep endometriosis: a narrative literature review. Arch Gynecol Obstet 2017; 296: 435-444
  • 16 Wieder DR, Pattimakiel L. Examining the efficacy, safety, and patient acceptability of the combined contraceptive vaginal ring (NuvaRing®). Int J Womens Health 2010; 2: 401
  • 17 Helbling IM, Ibarra JC, Luna JA. The optimization of an intravaginal ring releasing progesterone using a mathematical model. Pharm Res 2014; 31: 795-808
  • 18 Norman AW, Litwack G. Hormones. Oxford: Academic Press; 1997
  • 19 Gandhi J, Chen A, Dagur G. et al. Genitourinary syndrome of menopause: an overview of clinical manifestations, pathophysiology, etiology, evaluation, and management. Am J Obstet Gynecol 2016; 215: 704-711
  • 20 Hamid AA, Issa MB, Nizar NNA. Hormones. In: Ali E, Nizar NNA. eds. Preparation and Processing of Religious and Cultural Foods. 1st ed.. Sawston, United Kingdom, Cambridge, United Kingdom: Elsevier; 2018: 253-277
  • 21 Manoharan S. Engineering the N-Glycosylation Pathway in Pichia Pastoris for the Expression of Glycoprotein Hormones. Electronic Theses and Dissertations of Indian Institute of Science. Bangalore, India: Indian Institute of Science; 2018 Accessed March 12 2020 at: https://etd.iisc.ac.in/handle/2005/3017
  • 22 Cahoreau C, Klett D, Combarnous Y. Structure–function relationships of glycoprotein hormones and their subunitsʼ ancestors. Front Endocrinol (Lausanne) 2015; 6: 26
  • 23 Arlt W. Androgen therapy in women. Eur J Endocrinol 2006; 154: 1-11
  • 24 Lee DY, Kim E, Choi MH. Technical and clinical aspects of cortisol as a biochemical marker of chronic stress. BMB Rep 2015; 48: 209-216
  • 25 Tenover JL. Testosterone replacement therapy in older adult men. Int J Androl 1999; 22: 300-306
  • 26 Vanderschueren D, Vandenput L, Boonen S. et al. Androgens and bone. Endocr Rev 2004; 25: 389-425
  • 27 Norman AW, Henry HL. Hormones. Oxford: Elsevier Science; 2014
  • 28 Brache V, Payán LJ, Faundes A. Current status of contraceptive vaginal rings. Contraception 2013; 87: 264-272
  • 29 Dziuk P, Cook B. Passage of steroids through silicone rubber. Endocrinology 1966; 78: 208-211
  • 30 Murphy DJ, Desjardins D, Boyd P. et al. Impact of ring size and drug loading on the pharmacokinetics of a combination dapivirine-darunavir vaginal ring in cynomolgus macaques. Int J Pharm 2018; 550: 300-308
  • 31 Verstraete G, Vandenbussche L, Kasmi S. et al. Thermoplastic polyurethane-based intravaginal rings for prophylaxis and treatment of (recurrent) bacterial vaginosis. Int J Pharm 2017; 529: 218-226
  • 32 McCoy CF, Millar BG, Murphy DJ. et al. Mechanical testing methods for drug-releasing vaginal rings. Int J Pharm 2019; 559: 182-191
  • 33 Koutsamanis I, Eder S, Beretta M. et al. Formulation and processability screening for the rational design of ethylene-vinyl acetate based intra-vaginal rings. Int J Pharm 2019; 564: 90-97
  • 34 Malcolm RK, Woolfson AD, Toner CF. et al. Long-term, controlled release of the HIV microbicide TMC120 from silicone elastomer vaginal rings. J Antimicrob Chemother 2005; 56: 954-956
  • 35 McCoy CF, Murphy DJ, Boyd P. et al. Packing polymorphism of dapivirine and its impact on the performance of a dapivirine-releasing silicone elastomer vaginal ring. J Pharm Sci 2017; 106: 2015-2025
  • 36 托马斯, 施维塔, et al. 尤噶昂卡. Intravaginal ring for the delivery of unique combinations of antimicrobial compositions. Google Patents; 2016
  • 37 Mishell jr. DR, Colodny SZ, Swanson LA. The effect of an oral contraceptive on tests of thyroid function. Fertil Steril 1969; 20: 335
  • 38 Bounds W, Szarewski A, Lowe D. et al. Preliminary report of unexpected local reactions to a progestogen-releasing contraceptive vaginal ring. Eur J Obstet Gynecol Reprod Biol 1993; 48: 123-125
  • 39 Demetrio C, Ridout M, Campbell M. et al. Coping with extra Poisson variability in the analysis of factors influencing vaginal ring expulsions. Stat Med 1994; 13: 873-876
  • 40 Campbell MJ, Machin D, DʼArcangues C. Coping with extra Poisson variability in the analysis of factors influencing vaginal ring expulsions. Stat Med 1991; 10: 241-254
  • 41 Gupta KM, Pearce SM, Poursaid AE. et al. Polyurethane intravaginal ring for controlled delivery of dapivirine, a nonnucleoside reverse transcriptase inhibitor of HIV-1. J Pharm Sci 2008; 97: 4228-4239
  • 42 Boyd P, Fetherston SM, McCoy CF. et al. Matrix and reservoir-type multipurpose vaginal rings for controlled release of dapivirine and levonorgestrel. Int J Pharm 2016; 511: 619-629
  • 43 Derby N, Aravantinou M, Kenney J. et al. An intravaginal ring that releases three antiviral agents and a contraceptive blocks SHIV-RT infection, reduces HSV-2 shedding, and suppresses hormonal cycling in rhesus macaques. Drug Deliv Transl Res 2017; 7: 840-858
  • 44 Externbrink A, Eggenreich K, Eder S. et al. Development and evaluation of accelerated drug release testing methods for a matrix-type intravaginal ring. Eur J Pharm Biopharm 2017; 110: 1-12
  • 45 Mashak A, Rahimi A. Silicone polymers in controlled drug delivery systems: a review. Iran Polym J 2009; 18: 279-295
  • 46 Shabir Q, Webb K, Nadarassan D. et al. Quantification and reduction of the residual chemical reactivity of passivated biodegradable porous silicon for drug delivery applications. Silicon 2018; 10: 349-359
  • 47 Kumeria T, McInnes SJ, Maher S. et al. Porous silicon for drug delivery applications and theranostics: recent advances, critical review and perspectives. Expert Opin Drug Deliv 2017; 14: 1407-1422
  • 48 Colas A, Curtis J. Silicone biomaterials: history and chemistry. Biomater Sci-UK 2004; 2: 80-85
  • 49 Vondráček P, Doležel B. Biostability of medical elastomers: a review. Biomaterials 1984; 5: 209-214
  • 50 Colas A, Aguadisch L. Silicones in pharmaceutical applications. Chimie Nouvelle 1997; 15: 1779
  • 51 Bayer O, Siefken W, Rinke H. et al. A process for the production of polyurethanes and polyureas. German Patent DRP 1937; 728981
  • 52 Aksoy A. Synthesis and surface modification studies of biomedical polyurethanes to improve long term biocompatibility [Doctoral dissertation]. Ankara, Turkey: Graduate School of Natural and Applied Sciences of Middle East Technical University; 2008
  • 53 Lowinger M, Barrett S, Zhang F. et al. Sustained release drug delivery applications of polyurethanes. Pharmaceutics 2018; 10: 55
  • 54 Akindoyo J. et al. Polyurethane types, synthesis and applications–a review. Rsc Advances 2016; 6.115: 114453-114482
  • 55 Fu Y, Kao WJ. Drug release kinetics and transport mechanisms of non-degradable and degradable polymeric delivery systems. Expert Opin Drug Deliv 2010; 7: 429-444
  • 56 Clark JT, Clark MR, Shelke NB. et al. Engineering a segmented dual-reservoir polyurethane intravaginal ring for simultaneous prevention of HIV transmission and unwanted pregnancy. PloS One 2014; 9: e88509
  • 57 Schneider C, Langer R, Loveday D. et al. Applications of ethylene vinyl acetate copolymers (EVA) in drug delivery systems. J Control Release 2017; 262: 284-295
  • 58 Tallury P, Alimohammadi N, Kalachandra S. Poly (ethylene-co-vinyl acetate) copolymer matrix for delivery of chlorhexidine and acyclovir drugs for use in the oral environment: effect of drug combination, copolymer composition and coating on the drug release rate. Dent Mater 2007; 23: 404-409
  • 59 Brogly M, Nardin M, Schultz J. Effect of vinylacetate content on crystallinity and second‐order transitions in ethylene–vinylacetate copolymers. J Appl Polym Sci 1997; 64: 1903-1912
  • 60 Van Laarhoven J, Kruft M, Vromans H. In vitro release properties of etonogestrel and ethinyl estradiol from a contraceptive vaginal ring. Int J Pharm 2002; 232: 163-173
  • 61 Kuno N, Fujii S. Recent advances in ocular drug delivery systems. Polymers 2011; 3: 193-221
  • 62 Pharriss BB, Erickson R, Bashaw J. et al. Progestasert: a uterine therapeutic system for long-term contraception: 1. Philosophy and clinical efficacy. Fertil Steril 1974; 25: 915-921
  • 63 Hohmann H, Creinin MD. The contraceptive implant. Clin Obstet Gynecol 2007; 50: 907-917
  • 64 Mommers E, Blum G-F, Gent TG. et al. Nexplanon, a radiopaque etonogestrel implant in combination with a next-generation applicator: 3-year results of a noncomparative multicenter trial. Am J Obstet Gynecol 2012; 207: 388.e1-e6
  • 65 Bourges J, Bloquel C, Thomas A. et al. Intraocular implants for extended drug delivery: therapeutic applications. Adv Drug Deliv Rev 2006; 58: 1182-1202
  • 66 Novak A, De la Loge C, Abetz L. et al. The combined contraceptive vaginal ring, NuvaRing®: an international study of user acceptability. Contraception 2003; 67: 187-194
  • 67 McConville C, Major I, Friend DR. et al. Development of a UC781 releasing polyethylene vinyl acetate vaginal ring. Drug Deliv Transl Res 2012; 2: 489-497
  • 68 Ugaonkar SR, Wesenberg A, Wilk J. et al. A novel intravaginal ring to prevent HIV-1, HSV-2, HPV, and unintended pregnancy. J Control Release 2015; 213: 57-68
  • 69 Mc Cullagh SD, Malcolm RK, Woolfson AD. et al. Release kinetics of oleyl alcohol from a self-lubricating silicone biomaterial. J Mater Chem 2004; 14: 1093-1098
  • 70 Malcolm RK, Fetherston SM, McCoy CF. et al. Vaginal rings for delivery of HIV microbicides. Int J Womens Health 2012; 4: 595
  • 71 Kim S, Liu S. Smart and biostable polyurethanes for long-term implants. ACS Biomater Sci Eng 2018; 4: 1479-1490
  • 72 Qi X, Yang W, Yu L. et al. Design of ethylene-vinyl acetate copolymer fiber with two-way shape memory effect. Polymers 2019; 11: 1599
  • 73 Ohlberg SM, Alexander LE, Warrick E. Crystallinity and orientation in silicone rubber. I. X‐ray studies. J Polym Sci 1958; 27: 1-17
  • 74 Gomez CM, Gutierrez D, Asensio M. et al. Transparent thermoplastic polyurethanes based on aliphatic diisocyanates and polycarbonate diol. J Elastom Plast 2017; 49: 77-95
  • 75 Rosato DV, Rosato DV, Rosato MV. Plastic Product Material and Process Selection Handbook. Oxford: Elsevier; 2004
  • 76 Vaicekauskaite J, Mazurek P, Vudayagiri S. et al. Mapping the mechanical and electrical properties of commercial silicone elastomer formulations for stretchable transducers. J Mater Chem C 2020; 8: 1273-1279
  • 77 Shastri PV. Toxicology of polymers for implant contraceptives for women. Contraception 2002; 65: 9-13
  • 78 Rajendra V, Chen Y, Brook MA. Structured hydrophilic domains on silicone elastomers. Polym Chem 2010; 1: 312-320
  • 79 Stanković M, Frijlink HW, Hinrichs WL. Polymeric formulations for drug release prepared by hot melt extrusion: application and characterization. Drug Discov Today 2015; 20: 812-823
  • 80 Friend DR. Intravaginal rings: controlled release systems for contraception and prevention of transmission of sexually transmitted infections. Drug Deliv Transl Res 2011; 1: 185-193
  • 81 Kamaly N, Yameen B, Wu J. et al. Degradable controlled-release polymers and polymeric nanoparticles: mechanisms of controlling drug release. Chem Rev 2016; 116: 2602-2663
  • 82 Higuchi T. Mechanism of sustained‐action medication. Theoretical analysis of rate of release of solid drugs dispersed in solid matrices. J Pharm Sci 1963; 52: 1145-1149
  • 83 Roseman T, Higuchi W. Release of medroxyprogesterone acetate from a silicone polymer. J Pharm Sci 1970; 59: 353-357
  • 84 Malcolm RK, Edwards K-L, Kiser P. et al. Advances in microbicide vaginal rings. Antiviral Res 2010; 88: S30-S39
  • 85 Vartiainen J, Wahlström T, Nilsson C-G. Effects and acceptability of a new 17β-oestradiol-releasing vaginal ring in the treatment of postmenopausal complaints. Maturitas 1993; 17: 129-137
  • 86 Han YA, Singh M, Saxena BB. Development of vaginal rings for sustained release of nonhormonal contraceptives and anti-HIV agents. Contraception 2007; 76: 132-138
  • 87 Chien Y. Nasal Drug Delivery and Delivery Systems. In: Chien YW. Novel Drug Delivery Systems. Drugs and the pharmaceutical Sciences. 2nd ed.. New York: Marcel Dekker, Inc; 1992: 50
  • 88 Crank J. The Mathematics of Diffusion. London: Oxford University Press; 1979
  • 89 Russell JA, Malcolm RK, Campbell K. et al. High-performance liquid chromatographic determination of 17β-estradiol and 17β-estradiol-3-acetate solubilities and diffusion coefficients in silicone elastomeric intravaginal rings. J Chromatogr B Biomed Sci Appl 2000; 744: 157-163
  • 90 Jackanicz TM. Levonorgestrel and estradiol release from an improved contraceptive vaginal ring. Contraception 1981; 24: 323-339
  • 91 Morrow RJ, Woolfson AD, Donnelly L. et al. Sustained release of proteins from a modified vaginal ring device. Eur J Pharm Biopharm 2011; 77: 3-10
  • 92 Spence P, Garg AB, Woodsong C. et al. Recent work on vaginal rings containing antiviral agents for HIV prevention. Curr Opin HIV AIDS 2015; 10: 264-270
  • 93 Baum MM, Butkyavichene I, Gilman J. et al. An intravaginal ring for the simultaneous delivery of multiple drugs. J Pharm Sci 2012; 101: 2833-2843
  • 94 Baum MM, Butkyavichene I, Churchman SA. et al. An intravaginal ring for the sustained delivery of tenofovir disoproxil fumarate. Int J Pharm 2015; 495: 579-587
  • 95 Ashton P, Brown J, Pearson P. et al. Intravitreal ganciclovir pharmacokinetics in rabbits and man. J Ocul Pharmacol 1992; 8: 343-347
  • 96 Anand R, Nightingale SD, Fish RH. et al. Control of cytomegalovirus retinitis using sustained release of intraocular ganciclovir. Arch Ophthalmol 1993; 111: 223-227
  • 97 Musch DC, Martin DF, Gordon JF. et al. Treatment of cytomegalovirus retinitis with a sustained-release ganciclovir implant. N Engl J Med 1997; 337: 83-90
  • 98 Woolfson AD, Malcolm RK, Morrow RJ. et al. Intravaginal ring delivery of the reverse transcriptase inhibitor TMC120 as an HIV microbicide. Int J Pharm 2006; 325: 82-89
  • 99 [Anonym] Annovera: annual, comfortable, controllable birth control. Accessed December 15, 2020 at: https://www.annovera.com/
  • 100 [Anonym] Annovera – FDA. Issued: 08/2018. Accessed May 3, 2021 at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/209627s000lbl.pdf
  • 101 McBride JW, Boyd P, Dias N. et al. Vaginal rings with exposed cores for sustained delivery of the HIV CCR5 inhibitor 5P12-RANTES. J Control Release 2019; 298: 1-11
  • 102 FDA. Prescribing Information. Femring® (estradiol acetate vaginal ring). Accessed May 3, 2021 at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/021367s009lbl.pdf
  • 103 Thurman AR, Clark MR, Hurlburt JA. et al. Intravaginal rings as delivery systems for microbicides and multipurpose prevention technologies. Int J Womens Health 2013; 5: 695
  • 104 Schwarze J-E, Villa S, Manzur A. et al. Progesterone-releasing vaginal ring for luteal phase support after superovulation and intrauterine insemination cycles, a pilot study. JBRA Assist Reprod 2013; DOI: 10.5935/1518-0557.20130072.
  • 105 Algorta J, Diaz M, de Benito R. et al. Pharmacokinetic bioequivalence, safety and acceptability of Ornibel®, a new polymer composition contraceptive vaginal ring (etonogestrel/ethinylestradiol 11.00/3.474 mg) compared with Nuvaring®(etonogestrel/ethinylestradiol 11.7/2.7 mg). Eur J Contracept Reprod Health Care 2017; 22: 429-438
  • 106 Roumen FJ, op ten Berg MM, Hoomans EH. The combined contraceptive vaginal ring (NuvaRing®): first experience in daily clinical practice in The Netherlands. Eur J Contracept Reprod Health Care 2006; 11: 14-22
  • 107 Koetsawang S, Gao J, Krishna U. et al. Microdose intravaginal levonorgestrel contraception: A multicentre clinical trial: I. Contraceptive efficacy and side effects. Contraception 1990; 41: 105-124
  • 108 Brache V, Alvarez-Sanchez F, Faundes A. et al. Progestin-only contraceptive rings. Steroids 2000; 65: 687-691
  • 109 Massai R, Quinteros E, Reyes MV. et al. Extended use of a progesterone-releasing vaginal ring in nursing women: a phase II clinical trial. Contraception 2005; 72: 352-357
  • 110 Kerns J, Darney P. Vaginal ring contraception. Contraception 2011; 83: 107-115
  • 111 Sivin I, Mishell jr. DR, Alvarez F. et al. Contraceptive vaginal rings releasing Nestorone® and ethinylestradiol: a 1-year dose-finding trial. Contraception 2005; 71: 122-129
  • 112 Bjarnadóttir RI. Update on contraceptive vaginal rings. Reviews in Gynaecological Practice 2003; 3: 156-159
  • 113 Galzote RM, Rafie S, Teal R. et al. Transdermal delivery of combined hormonal contraception: a review of the current literature. Int J Womens Health 2017; 9: 315
  • 114 Stewart M, Black K. Choosing a combined oral contraceptive pill. Aust Prescr 2015; 38: 6
  • 115 Nath A, Sitruk-Ware R. Progesterone vaginal ring for contraceptive use during lactation. Contraception 2010; 82: 428-434
  • 116 Rivera R, Yacobson I, Grimes D. The mechanism of action of hormonal contraceptives and intrauterine contraceptive devices. Am J Obstet Gynecol 1999; 181: 1263-1269
  • 117 Ballagh SA. Vaginal ring hormone delivery systems in contraception and menopause. Clin Obstet Gynecol 2001; 44: 106-113
  • 118 Wagner M-S, Black A. The Combined Contraceptive Vaginal Ring: an Update. Current Obstetrics and Gynecology Reports 2016; 5: 1-12
  • 119 Gruber CJ. The combined contraceptive vaginal ring (NuvaRing): evaluation of the clinical and pharmacological evidence. Womens Health (Lond) 2006; 2: 351-356
  • 120 Roumen FJ. Review of the combined contraceptive vaginal ring, NuvaRing® . Ther Clin Risk Manag 2008; 4: 441
  • 121 Mulders TM, Dieben TO, Bennink HJC. Ovarian function with a novel combined contraceptive vaginal ring. Hum Reprod 2002; 17: 2594-2599
  • 122 Fan GS, Ren M, Di W. et al. Efficacy and safety of the contraceptive vaginal ring (NuvaRing) compared with a combined oral contraceptive in Chinese women: a 1-year randomised trial. Eur J Contracept Reprod Health Care 2016; 21: 303-309
  • 123 Reproductive Health Supplies Coalition. Progesterone vaginal ring. Brussels, Belgium: Reproductive Health Supplies Coalition; Accessed May 3 2021 at: https://www.rhsupplies.org/fileadmin/uploads/rhsc/Working_Groups/New_Underused_RH_Technologies_Caucus/Documents/Technical_Briefs/rhsc-brief-progesterone_A4.pdf
  • 124 Johansson ED, Sitruk-Ware R. New delivery systems in contraception: vaginal rings. Am J Obstet Gynecol 2004; 190: S54-S59
  • 125 Friend DR. Drug delivery for the treatment of endometriosis and uterine fibroids. Drug Deliv Transl Res 2017; 7: 829-839
  • 126 Parasar P, Ozcan P, Terry KL. Endometriosis: epidemiology, diagnosis and clinical management. Curr Obstet Gynecol Rep 2017; 6: 34-41
  • 127 Pluchino N, Freschi L, Wenger J-M. et al. Innovations in classical hormonal targets for endometriosis. Expert Rev Clin Pharmacol 2016; 9: 317-327
  • 128 Razzi S, Luisi S, Calonaci F. et al. Efficacy of vaginal danazol treatment in women with recurrent deeply infiltrating endometriosis. Fertil Steril 2007; 88: 789-794
  • 129 Ryan IP, Schriock ED, Taylor RN. Isolation, characterization, and comparison of human endometrial and endometriosis cells in vitro. J Clin Endocrinol Metab 1994; 78: 642-649
  • 130 ACOG Committee on Practice Bulletins–Gynecology. ACOG Practice Bulletin No. 51. Chronic pelvic pain. Obstet Gynecol 2004; 103: 589-605
  • 131 Reinecke I, Schultze-Mosgau MH, Nave R. et al. Model‐based dose selection for intravaginal ring formulations releasing anastrozole and levonorgestrel intended for the treatment of endometriosis symptoms. J Clin Pharmacol 2017; 57: 640-651
  • 132 Dunselman G, Vermeulen N, Becker C. et al. ESHRE guideline: management of women with endometriosis. Hum Reprod 2014; 29: 400-412
  • 133 Vercellini P, Barbara G, Somigliana E. et al. Comparison of contraceptive ring and patch for the treatment of symptomatic endometriosis. Fertil Steril 2010; 93: 2150-2161
  • 134 Sartori MGF, Feldner jr. P, Jarmy-Di Bella Z. et al. Sexual steroids in urogynecology. Climacteric 2011; 14: 5-14
  • 135 Palacios S, Nappi R, Bruyniks N. et al. The European Vulvovaginal Epidemiological Survey (EVES): prevalence, symptoms and impact of vulvovaginal atrophy of menopause. Climacteric 2018; 21: 286-291
  • 136 Van Keep P, Utian W, Vermeulen A. Potency and hepato-cellular Effects of Oestrogens after oral, percutaneous, and subcutaneous Administration. In: Utian WH, van Keep PA, Vermeulen A. eds. The controversial Climacteric. Dordrecht: Springer; 1982: 103-125
  • 137 Antisell J, Poon T, Borey A, Briddell J, Palesch S. Hormone Delivery System: The Contraceptive Ring. Ithaca, New York: Cornell University Library; 2006. Accessed May 3 2021 at: https://ecommons.cornell.edu/handle/1813/3060
  • 138 Kuhl H. Pharmacology of estrogens and progestogens: influence of different routes of administration. Climacteric 2005; 8: 3-63
  • 139 Nabahi S. Intravaginal drug delivery device. Google Patents; 1998
  • 140 Cicinelli E. Intravaginal oestrogen and progestin administration: advantages and disadvantages. Best Pract Res Clin Obstet Gynaecol 2008; 22: 391-405
  • 141 Casper F, Petri E. Local treatment of urogenital atrophy with an estradiol-releasing vaginal ring: a comparative and a placebo-controlled multicenter study. Vaginal Ring Study Group. Int Urogynecol J Pelvic Floor Dysfunct 1999; 10: 171-176
  • 142 Hill DA, Crider M, Hill SR. Hormone therapy and other treatments for symptoms of menopause. Am Fam Physician 2016; 94: 884-889
  • 143 Jilka RL, Takahashi K, Munshi M. et al. Loss of estrogen upregulates osteoblastogenesis in the murine bone marrow. Evidence for autonomy from factors released during bone resorption. J Clin Invest 1998; 101: 1942-1950
  • 144 Khosla S, Oursler MJ, Monroe DG. Estrogen and the skeleton. Trends Endocrinol Metab 2012; 23: 576-581
  • 145 Almeida M, Laurent MR, Dubois V. et al. Estrogens and androgens in skeletal physiology and pathophysiology. Physiol Rev 2017; 97: 135-187
  • 146 Paganini-Hill A, Henderson VW. Estrogen deficiency and risk of Alzheimerʼs disease in women. Am J Epidemiol 1994; 140: 256-261
  • 147 Lindahl SH. Reviewing the options for local estrogen treatment of vaginal atrophy. Int J Womens Health 2014; 6: 307
  • 148 Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause 2013; 20: 888-902 quiz 903–904
  • 149 Warner Chilcott Laboratories. Estrace (estradiol vaginal cream, USP, 0.01 %) [prescribing information]. Rockaway, NJ: Warner Chilcott Laboratories; 2011
  • 150 Food and Drug Administration. NDA 020216/S-067. Supplement Approval. Accessed May 3, 2021 at: https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2011/020216s067ltr.pdf
  • 151 Rybacki JJ. DOSAGE AND ADMINISTRATION. Published at 6/21/2017 on RxList website. Accessed October 29, 2020 at: https://www.rxlist.com/estragyn-drug.htm
  • 152 Neidecker MV. Patterns of Medication Use and Related Health Care Service Utilization Associated with Vaginal Estrogen Therapy in Medicaid-Enrolled Women [Diss.]. The Ohio State University; 2009
  • 153 [Anonym] Estring. New York: Pharmacia & Upjohn Co; 2008
  • 154 [Anonym] Femring. Rockaway, NJ: Warner Chilcott Inc; 2005
  • 155 Smith P, Heimer G, Lindskog M. et al. Oestradiol-releasing vaginal ring for treatment of postmenopausal urogenital atrophy. Maturitas 1993; 16: 145-154
  • 156 Barentsen R, van de Weijer PH, Schram JH. Continuous low dose estradiol released from a vaginal ring versus estriol vaginal cream for urogenital atrophy. Eur J Obstet Gynecol Reprod Biol 1997; 71: 73-80
  • 157 Holmgren P-Å, Lindskog M, Von Schoultz B. Vaginal rings for continuous low-dose release of oestradiol in the treatment of urogenital atrophy. Maturitas 1989; 11: 55-63
  • 158 Schmidt G, Andersson S-B, Nordle Ö. et al. Release of 17-beta-oestradiol from a vaginal ring in postmenopausal women: pharmacokinetic evaluation. Gynecol Obstet Invest 1994; 38: 253-260
  • 159 Suckling J, Lethaby A, Kennedy R. Local oestrogen for vaginal atrophy in postmenopausal women. Cochrane Database Syst Rev 2006; (04) CD001500
  • 160 Crandall CJ, Hovey KM, Andrews CA. et al. Breast cancer, endometrial cancer, and cardiovascular events in participants who used vaginal estrogen in the Womenʼs Health Initiative Observational Study. Menopause 2018; 25: 11-20
  • 161 Krause M, Wheeler 2nd TL, Snyder TE. et al. Local Effects of Vaginally Administered Estrogen Therapy: A Review. J Pelvic Med Surg 2009; 15: 105-114
  • 162 Castelo-Branco C, Cancelo MJ, Villero J. et al. Management of post-menopausal vaginal atrophy and atrophic vaginitis. Maturitas 2005; 52: 46-52
  • 163 Naumova I, Castelo-Branco C. Current treatment options for postmenopausal vaginal atrophy. Int J Womens Health 2018; 10: 387
  • 164 Calleja-Agius J, Brincat M. Urogenital atrophy. Climacteric 2009; 12: 279-285
  • 165 Bell RJ, Rizvi F, Islam RM. et al. A systematic review of intravaginal testosterone for the treatment of vulvovaginal atrophy. Menopause 2018; 25: 704-709
  • 166 Robinson D, Cardozo LD. The role of estrogens in female lower urinary tract dysfunction. Urology 2003; 62: 45-51
  • 167 Pruthi S, Simon JA, Early AP. Current overview of the management of urogenital atrophy in women with breast cancer. Breast J 2011; 17: 403-408
  • 168 Willhite LA, OʼConnell MB. Urogenital atrophy: prevention and treatment. Pharmacotherapy 2001; 21: 464-480
  • 169 Leiblum S, Bachmann G, Kemmann E. et al. Vaginal atrophy in the postmenopausal woman: the importance of sexual activity and hormones. JAMA 1983; 249: 2195-2198
  • 170 Moegele M, Buchholz S, Seitz S. et al. Vaginal Estrogen Therapy for Patients with Breast Cancer. Geburtshilfe Frauenheilkd 2013; 73: 1017-1022
  • 171 Huang X, Brazel CS. On the importance and mechanisms of burst release in matrix-controlled drug delivery systems. J Control Release 2001; 73: 121-136
  • 172 Helbling IM, Ibarra JC, Luna JA. The use of cellulose membrane to eliminate burst release from intravaginal rings. AAPS J 2016; 18: 960-971
  • 173 Tønnesen HH, Karlsen J. Alginate in drug delivery systems. Drug Dev Ind Pharm 2002; 28: 621-630
  • 174 Sershen S, West J. Implantable, polymeric systems for modulated drug delivery. Adv Drug Deliv Rev 2002; 54: 1225-1235
  • 175 Tiedeken M, Westhoff CL, Cohen A. et al. Bone turnover markers in women participating in a dose-finding trial of a contraceptive vaginal ring releasing Nestorone and estradiol. Contraception 2019; 99: 329-334
  • 176 Stifani BM, Plagianos M, Vieira CS. et al. Factors associated with nonadherence to instructions for using the Nestorone®/ethinyl estradiol contraceptive vaginal ring. Contraception 2018; 97: 415-421
  • 177 Nel A, Bekker L-G, Bukusi E. et al. Safety, acceptability and adherence of dapivirine vaginal ring in a microbicide clinical trial conducted in multiple countries in Sub-Saharan Africa. PloS One 2016; 11: e0147743
  • 178 Sefton MV. Implantable pumps. Medical Applications of Controlled Release 1984; 1: 129-158
  • 179 Conte U, Maggi L. A flexible technology for the linear, pulsatile and delayed release of drugs, allowing for easy accommodation of difficult in vitro targets. J Control Release 2000; 64: 263-268
  • 180 Lee E, Kim S, Kim S. et al. Drug release from hydrogel devices with ratecontrolling barriers. J Membr Sci 1980; 7: 293-303
  • 181 Yang L, Fassihi R. Modulation of diclofenac release from a totally soluble controlled release drug delivery system. J Control Release 1997; 44: 135-140
  • 182 Hildgen P, McMullen J. A new gradient matrix: formulation and characterization. J Control Release 1995; 34: 263-271
  • 183 Lu S, Anseth KS. Photopolymerization of multilaminated poly (HEMA) hydrogels for controlled release. J Control Release 1999; 57: 291-300
  • 184 Lu S, Ramirez WF, Anseth KS. Photopolymerized, multilaminated matrix devices with optimized nonuniform initial concentration profiles to control drug release. J Pharm Sci 2000; 89: 45-51
  • 185 Qiu Y, Chidambaram N, Flood K. Design and evaluation of layered diffusional matrices for zero-order sustained-release. J Control Release 1998; 51: 123-130
  • 186 Narasimhan B, Peppas NA. Molecular analysis of drug delivery systems controlled by dissolution of the polymer carrier. J Pharm Sci 1997; 86: 297-304
  • 187 Park M, Park CG, Lee SH. et al. Polymeric tube-shaped devices with controlled geometry for programmed drug delivery. Macromol Res 2012; 20: 960-967
  • 188 Johnson TJ. Antiretroviral eluting intravaginal Rings to prevent the sexual Transmission of HIV. Salt Lake City, Utah: The University of Utah; 2012
  • 189 Jensen JT. Vaginal ring delivery of selective progesterone receptor modulators for contraception. Contraception 2013; 87: 314-318
  • 190 Singer R, Mawson P, Derby N. et al. An intravaginal ring that releases the NNRTI MIV-150 reduces SHIV transmission in macaques. Sci Transl Med 2012; 4: 150ra123
  • 191 Kenney J, Aravantinou M, Singer R. et al. An antiretroviral/zinc combination gel provides 24 hours of complete protection against vaginal SHIV infection in macaques. PloS One 2011; 6: e15835
  • 192 Dobard C, Sharma S, Martin A. et al. Durable protection from vaginal simian-human immunodeficiency virus infection in macaques by tenofovir gel and its relationship to drug levels in tissue. J Virol 2012; 86: 718-725
  • 193 Derby N, Zydowsky T, Robbiani M. In search of the optimal delivery method for anti-HIV microbicides: are intravaginal rings the way forward?. Expert Rev Anti Infect Ther 2013; 11: 5-8
  • 194 Gao S, Tang G, Hua D. et al. Stimuli-responsive bio-based polymeric systems and their applications. J Mater Chem B 2019; 7: 709-729
  • 195 Jiang J, Xie J, Ma B. et al. Mussel-inspired protein-mediated surface functionalization of electrospun nanofibers for pH-responsive drug delivery. Acta Biomater 2014; 10: 1324-1332

Correspondence

Dr.-Ing. Hadi Tabesh
Assist. Professor of Chemical/Biomedical Engineering
Department of Life Science Engineering
Faculty of New Sciences and Technologies
University of Tehran
North Kargar street
Tehran
Iran   

Publication History

Received: 08 November 2020

Accepted after revision: 20 January 2021

Article published online:
13 July 2021

© 2021. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commecial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

  • References

  • 1 Malcolm RK, Boyd PJ, McCoy CF. et al. Microbicide vaginal rings: Technological challenges and clinical development. Advanced Drug Delivery Reviews 2016; 103: 33-56
  • 2 Malcolm RK. The intravaginal ring. Drug Pharm Sci 2003; 126: 775-790
  • 3 Hussain A, Ahsan F. The vagina as a route for systemic drug delivery. J Control Release 2005; 103: 301-313
  • 4 Murphy DJ, Boyd P, McCoy CF. et al. Controlling levonorgestrel binding and release in a multi-purpose prevention technology vaginal ring device. J Control Release 2016; 226: 138-147
  • 5 Duncan GW. Medicated devices and methods. In: Google Patents; 1970
  • 6 Schopflin G. Drug excipient of silicone rubber. Google Patents; 1977
  • 7 Guthrie KM, Rosen RK, Vargas SE. et al. User evaluations offer promise for pod-intravaginal ring as a drug delivery platform: A mixed methods study of acceptability and use experiences. PloS One 2018; 13: e0197269
  • 8 Jensen J, Edelman A, Chen B. et al. Continuous dosing of a novel contraceptive vaginal ring releasing Nestorone® and estradiol: pharmacokinetics from a dose-finding study. Contraception 2018; 97: 422-427
  • 9 Keller M, Ray L, Atrio JM. et al. Early termination of a phase 1 trial of tenofovir disoproxil fumarate vaginal ring. Contraception 2018; 1: 20
  • 10 Nave R, Jalkanen T, Talling C. et al. The Effect of Drug Content Reduction on the In Vitro and In Vivo Properties of Levonorgestrel-Releasing Intravaginal Rings. J Pharm Sci 2018; 107: 1020-1027
  • 11 Fetherston SM, Boyd P, McCoy CF. et al. A silicone elastomer vaginal ring for HIV prevention containing two microbicides with different mechanisms of action. Eur J Pharm Sci 2013; 48: 406-415
  • 12 Boyd P, Variano B, Spence P. et al. In vitro release testing methods for drug-releasing vaginal rings. J Control Release 2019; 313: 54-69
  • 13 Thurman AR, Schwartz JL, Brache V. et al. Randomized, placebo controlled phase I trial of safety, pharmacokinetics, pharmacodynamics and acceptability of tenofovir and tenofovir plus levonorgestrel vaginal rings in women. PloS One 2018; 13: e0199778
  • 14 Lete I, Cuesta MC, Marín JM. et al. Vaginal health in contraceptive vaginal ring users–A review. Eur J Contracept Reprod Health Care 2013; 18: 234-241
  • 15 Buggio L, Lazzari C, Monti E. et al. “Per vaginam” topical use of hormonal drugs in women with symptomatic deep endometriosis: a narrative literature review. Arch Gynecol Obstet 2017; 296: 435-444
  • 16 Wieder DR, Pattimakiel L. Examining the efficacy, safety, and patient acceptability of the combined contraceptive vaginal ring (NuvaRing®). Int J Womens Health 2010; 2: 401
  • 17 Helbling IM, Ibarra JC, Luna JA. The optimization of an intravaginal ring releasing progesterone using a mathematical model. Pharm Res 2014; 31: 795-808
  • 18 Norman AW, Litwack G. Hormones. Oxford: Academic Press; 1997
  • 19 Gandhi J, Chen A, Dagur G. et al. Genitourinary syndrome of menopause: an overview of clinical manifestations, pathophysiology, etiology, evaluation, and management. Am J Obstet Gynecol 2016; 215: 704-711
  • 20 Hamid AA, Issa MB, Nizar NNA. Hormones. In: Ali E, Nizar NNA. eds. Preparation and Processing of Religious and Cultural Foods. 1st ed.. Sawston, United Kingdom, Cambridge, United Kingdom: Elsevier; 2018: 253-277
  • 21 Manoharan S. Engineering the N-Glycosylation Pathway in Pichia Pastoris for the Expression of Glycoprotein Hormones. Electronic Theses and Dissertations of Indian Institute of Science. Bangalore, India: Indian Institute of Science; 2018 Accessed March 12 2020 at: https://etd.iisc.ac.in/handle/2005/3017
  • 22 Cahoreau C, Klett D, Combarnous Y. Structure–function relationships of glycoprotein hormones and their subunitsʼ ancestors. Front Endocrinol (Lausanne) 2015; 6: 26
  • 23 Arlt W. Androgen therapy in women. Eur J Endocrinol 2006; 154: 1-11
  • 24 Lee DY, Kim E, Choi MH. Technical and clinical aspects of cortisol as a biochemical marker of chronic stress. BMB Rep 2015; 48: 209-216
  • 25 Tenover JL. Testosterone replacement therapy in older adult men. Int J Androl 1999; 22: 300-306
  • 26 Vanderschueren D, Vandenput L, Boonen S. et al. Androgens and bone. Endocr Rev 2004; 25: 389-425
  • 27 Norman AW, Henry HL. Hormones. Oxford: Elsevier Science; 2014
  • 28 Brache V, Payán LJ, Faundes A. Current status of contraceptive vaginal rings. Contraception 2013; 87: 264-272
  • 29 Dziuk P, Cook B. Passage of steroids through silicone rubber. Endocrinology 1966; 78: 208-211
  • 30 Murphy DJ, Desjardins D, Boyd P. et al. Impact of ring size and drug loading on the pharmacokinetics of a combination dapivirine-darunavir vaginal ring in cynomolgus macaques. Int J Pharm 2018; 550: 300-308
  • 31 Verstraete G, Vandenbussche L, Kasmi S. et al. Thermoplastic polyurethane-based intravaginal rings for prophylaxis and treatment of (recurrent) bacterial vaginosis. Int J Pharm 2017; 529: 218-226
  • 32 McCoy CF, Millar BG, Murphy DJ. et al. Mechanical testing methods for drug-releasing vaginal rings. Int J Pharm 2019; 559: 182-191
  • 33 Koutsamanis I, Eder S, Beretta M. et al. Formulation and processability screening for the rational design of ethylene-vinyl acetate based intra-vaginal rings. Int J Pharm 2019; 564: 90-97
  • 34 Malcolm RK, Woolfson AD, Toner CF. et al. Long-term, controlled release of the HIV microbicide TMC120 from silicone elastomer vaginal rings. J Antimicrob Chemother 2005; 56: 954-956
  • 35 McCoy CF, Murphy DJ, Boyd P. et al. Packing polymorphism of dapivirine and its impact on the performance of a dapivirine-releasing silicone elastomer vaginal ring. J Pharm Sci 2017; 106: 2015-2025
  • 36 托马斯, 施维塔, et al. 尤噶昂卡. Intravaginal ring for the delivery of unique combinations of antimicrobial compositions. Google Patents; 2016
  • 37 Mishell jr. DR, Colodny SZ, Swanson LA. The effect of an oral contraceptive on tests of thyroid function. Fertil Steril 1969; 20: 335
  • 38 Bounds W, Szarewski A, Lowe D. et al. Preliminary report of unexpected local reactions to a progestogen-releasing contraceptive vaginal ring. Eur J Obstet Gynecol Reprod Biol 1993; 48: 123-125
  • 39 Demetrio C, Ridout M, Campbell M. et al. Coping with extra Poisson variability in the analysis of factors influencing vaginal ring expulsions. Stat Med 1994; 13: 873-876
  • 40 Campbell MJ, Machin D, DʼArcangues C. Coping with extra Poisson variability in the analysis of factors influencing vaginal ring expulsions. Stat Med 1991; 10: 241-254
  • 41 Gupta KM, Pearce SM, Poursaid AE. et al. Polyurethane intravaginal ring for controlled delivery of dapivirine, a nonnucleoside reverse transcriptase inhibitor of HIV-1. J Pharm Sci 2008; 97: 4228-4239
  • 42 Boyd P, Fetherston SM, McCoy CF. et al. Matrix and reservoir-type multipurpose vaginal rings for controlled release of dapivirine and levonorgestrel. Int J Pharm 2016; 511: 619-629
  • 43 Derby N, Aravantinou M, Kenney J. et al. An intravaginal ring that releases three antiviral agents and a contraceptive blocks SHIV-RT infection, reduces HSV-2 shedding, and suppresses hormonal cycling in rhesus macaques. Drug Deliv Transl Res 2017; 7: 840-858
  • 44 Externbrink A, Eggenreich K, Eder S. et al. Development and evaluation of accelerated drug release testing methods for a matrix-type intravaginal ring. Eur J Pharm Biopharm 2017; 110: 1-12
  • 45 Mashak A, Rahimi A. Silicone polymers in controlled drug delivery systems: a review. Iran Polym J 2009; 18: 279-295
  • 46 Shabir Q, Webb K, Nadarassan D. et al. Quantification and reduction of the residual chemical reactivity of passivated biodegradable porous silicon for drug delivery applications. Silicon 2018; 10: 349-359
  • 47 Kumeria T, McInnes SJ, Maher S. et al. Porous silicon for drug delivery applications and theranostics: recent advances, critical review and perspectives. Expert Opin Drug Deliv 2017; 14: 1407-1422
  • 48 Colas A, Curtis J. Silicone biomaterials: history and chemistry. Biomater Sci-UK 2004; 2: 80-85
  • 49 Vondráček P, Doležel B. Biostability of medical elastomers: a review. Biomaterials 1984; 5: 209-214
  • 50 Colas A, Aguadisch L. Silicones in pharmaceutical applications. Chimie Nouvelle 1997; 15: 1779
  • 51 Bayer O, Siefken W, Rinke H. et al. A process for the production of polyurethanes and polyureas. German Patent DRP 1937; 728981
  • 52 Aksoy A. Synthesis and surface modification studies of biomedical polyurethanes to improve long term biocompatibility [Doctoral dissertation]. Ankara, Turkey: Graduate School of Natural and Applied Sciences of Middle East Technical University; 2008
  • 53 Lowinger M, Barrett S, Zhang F. et al. Sustained release drug delivery applications of polyurethanes. Pharmaceutics 2018; 10: 55
  • 54 Akindoyo J. et al. Polyurethane types, synthesis and applications–a review. Rsc Advances 2016; 6.115: 114453-114482
  • 55 Fu Y, Kao WJ. Drug release kinetics and transport mechanisms of non-degradable and degradable polymeric delivery systems. Expert Opin Drug Deliv 2010; 7: 429-444
  • 56 Clark JT, Clark MR, Shelke NB. et al. Engineering a segmented dual-reservoir polyurethane intravaginal ring for simultaneous prevention of HIV transmission and unwanted pregnancy. PloS One 2014; 9: e88509
  • 57 Schneider C, Langer R, Loveday D. et al. Applications of ethylene vinyl acetate copolymers (EVA) in drug delivery systems. J Control Release 2017; 262: 284-295
  • 58 Tallury P, Alimohammadi N, Kalachandra S. Poly (ethylene-co-vinyl acetate) copolymer matrix for delivery of chlorhexidine and acyclovir drugs for use in the oral environment: effect of drug combination, copolymer composition and coating on the drug release rate. Dent Mater 2007; 23: 404-409
  • 59 Brogly M, Nardin M, Schultz J. Effect of vinylacetate content on crystallinity and second‐order transitions in ethylene–vinylacetate copolymers. J Appl Polym Sci 1997; 64: 1903-1912
  • 60 Van Laarhoven J, Kruft M, Vromans H. In vitro release properties of etonogestrel and ethinyl estradiol from a contraceptive vaginal ring. Int J Pharm 2002; 232: 163-173
  • 61 Kuno N, Fujii S. Recent advances in ocular drug delivery systems. Polymers 2011; 3: 193-221
  • 62 Pharriss BB, Erickson R, Bashaw J. et al. Progestasert: a uterine therapeutic system for long-term contraception: 1. Philosophy and clinical efficacy. Fertil Steril 1974; 25: 915-921
  • 63 Hohmann H, Creinin MD. The contraceptive implant. Clin Obstet Gynecol 2007; 50: 907-917
  • 64 Mommers E, Blum G-F, Gent TG. et al. Nexplanon, a radiopaque etonogestrel implant in combination with a next-generation applicator: 3-year results of a noncomparative multicenter trial. Am J Obstet Gynecol 2012; 207: 388.e1-e6
  • 65 Bourges J, Bloquel C, Thomas A. et al. Intraocular implants for extended drug delivery: therapeutic applications. Adv Drug Deliv Rev 2006; 58: 1182-1202
  • 66 Novak A, De la Loge C, Abetz L. et al. The combined contraceptive vaginal ring, NuvaRing®: an international study of user acceptability. Contraception 2003; 67: 187-194
  • 67 McConville C, Major I, Friend DR. et al. Development of a UC781 releasing polyethylene vinyl acetate vaginal ring. Drug Deliv Transl Res 2012; 2: 489-497
  • 68 Ugaonkar SR, Wesenberg A, Wilk J. et al. A novel intravaginal ring to prevent HIV-1, HSV-2, HPV, and unintended pregnancy. J Control Release 2015; 213: 57-68
  • 69 Mc Cullagh SD, Malcolm RK, Woolfson AD. et al. Release kinetics of oleyl alcohol from a self-lubricating silicone biomaterial. J Mater Chem 2004; 14: 1093-1098
  • 70 Malcolm RK, Fetherston SM, McCoy CF. et al. Vaginal rings for delivery of HIV microbicides. Int J Womens Health 2012; 4: 595
  • 71 Kim S, Liu S. Smart and biostable polyurethanes for long-term implants. ACS Biomater Sci Eng 2018; 4: 1479-1490
  • 72 Qi X, Yang W, Yu L. et al. Design of ethylene-vinyl acetate copolymer fiber with two-way shape memory effect. Polymers 2019; 11: 1599
  • 73 Ohlberg SM, Alexander LE, Warrick E. Crystallinity and orientation in silicone rubber. I. X‐ray studies. J Polym Sci 1958; 27: 1-17
  • 74 Gomez CM, Gutierrez D, Asensio M. et al. Transparent thermoplastic polyurethanes based on aliphatic diisocyanates and polycarbonate diol. J Elastom Plast 2017; 49: 77-95
  • 75 Rosato DV, Rosato DV, Rosato MV. Plastic Product Material and Process Selection Handbook. Oxford: Elsevier; 2004
  • 76 Vaicekauskaite J, Mazurek P, Vudayagiri S. et al. Mapping the mechanical and electrical properties of commercial silicone elastomer formulations for stretchable transducers. J Mater Chem C 2020; 8: 1273-1279
  • 77 Shastri PV. Toxicology of polymers for implant contraceptives for women. Contraception 2002; 65: 9-13
  • 78 Rajendra V, Chen Y, Brook MA. Structured hydrophilic domains on silicone elastomers. Polym Chem 2010; 1: 312-320
  • 79 Stanković M, Frijlink HW, Hinrichs WL. Polymeric formulations for drug release prepared by hot melt extrusion: application and characterization. Drug Discov Today 2015; 20: 812-823
  • 80 Friend DR. Intravaginal rings: controlled release systems for contraception and prevention of transmission of sexually transmitted infections. Drug Deliv Transl Res 2011; 1: 185-193
  • 81 Kamaly N, Yameen B, Wu J. et al. Degradable controlled-release polymers and polymeric nanoparticles: mechanisms of controlling drug release. Chem Rev 2016; 116: 2602-2663
  • 82 Higuchi T. Mechanism of sustained‐action medication. Theoretical analysis of rate of release of solid drugs dispersed in solid matrices. J Pharm Sci 1963; 52: 1145-1149
  • 83 Roseman T, Higuchi W. Release of medroxyprogesterone acetate from a silicone polymer. J Pharm Sci 1970; 59: 353-357
  • 84 Malcolm RK, Edwards K-L, Kiser P. et al. Advances in microbicide vaginal rings. Antiviral Res 2010; 88: S30-S39
  • 85 Vartiainen J, Wahlström T, Nilsson C-G. Effects and acceptability of a new 17β-oestradiol-releasing vaginal ring in the treatment of postmenopausal complaints. Maturitas 1993; 17: 129-137
  • 86 Han YA, Singh M, Saxena BB. Development of vaginal rings for sustained release of nonhormonal contraceptives and anti-HIV agents. Contraception 2007; 76: 132-138
  • 87 Chien Y. Nasal Drug Delivery and Delivery Systems. In: Chien YW. Novel Drug Delivery Systems. Drugs and the pharmaceutical Sciences. 2nd ed.. New York: Marcel Dekker, Inc; 1992: 50
  • 88 Crank J. The Mathematics of Diffusion. London: Oxford University Press; 1979
  • 89 Russell JA, Malcolm RK, Campbell K. et al. High-performance liquid chromatographic determination of 17β-estradiol and 17β-estradiol-3-acetate solubilities and diffusion coefficients in silicone elastomeric intravaginal rings. J Chromatogr B Biomed Sci Appl 2000; 744: 157-163
  • 90 Jackanicz TM. Levonorgestrel and estradiol release from an improved contraceptive vaginal ring. Contraception 1981; 24: 323-339
  • 91 Morrow RJ, Woolfson AD, Donnelly L. et al. Sustained release of proteins from a modified vaginal ring device. Eur J Pharm Biopharm 2011; 77: 3-10
  • 92 Spence P, Garg AB, Woodsong C. et al. Recent work on vaginal rings containing antiviral agents for HIV prevention. Curr Opin HIV AIDS 2015; 10: 264-270
  • 93 Baum MM, Butkyavichene I, Gilman J. et al. An intravaginal ring for the simultaneous delivery of multiple drugs. J Pharm Sci 2012; 101: 2833-2843
  • 94 Baum MM, Butkyavichene I, Churchman SA. et al. An intravaginal ring for the sustained delivery of tenofovir disoproxil fumarate. Int J Pharm 2015; 495: 579-587
  • 95 Ashton P, Brown J, Pearson P. et al. Intravitreal ganciclovir pharmacokinetics in rabbits and man. J Ocul Pharmacol 1992; 8: 343-347
  • 96 Anand R, Nightingale SD, Fish RH. et al. Control of cytomegalovirus retinitis using sustained release of intraocular ganciclovir. Arch Ophthalmol 1993; 111: 223-227
  • 97 Musch DC, Martin DF, Gordon JF. et al. Treatment of cytomegalovirus retinitis with a sustained-release ganciclovir implant. N Engl J Med 1997; 337: 83-90
  • 98 Woolfson AD, Malcolm RK, Morrow RJ. et al. Intravaginal ring delivery of the reverse transcriptase inhibitor TMC120 as an HIV microbicide. Int J Pharm 2006; 325: 82-89
  • 99 [Anonym] Annovera: annual, comfortable, controllable birth control. Accessed December 15, 2020 at: https://www.annovera.com/
  • 100 [Anonym] Annovera – FDA. Issued: 08/2018. Accessed May 3, 2021 at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/209627s000lbl.pdf
  • 101 McBride JW, Boyd P, Dias N. et al. Vaginal rings with exposed cores for sustained delivery of the HIV CCR5 inhibitor 5P12-RANTES. J Control Release 2019; 298: 1-11
  • 102 FDA. Prescribing Information. Femring® (estradiol acetate vaginal ring). Accessed May 3, 2021 at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/021367s009lbl.pdf
  • 103 Thurman AR, Clark MR, Hurlburt JA. et al. Intravaginal rings as delivery systems for microbicides and multipurpose prevention technologies. Int J Womens Health 2013; 5: 695
  • 104 Schwarze J-E, Villa S, Manzur A. et al. Progesterone-releasing vaginal ring for luteal phase support after superovulation and intrauterine insemination cycles, a pilot study. JBRA Assist Reprod 2013; DOI: 10.5935/1518-0557.20130072.
  • 105 Algorta J, Diaz M, de Benito R. et al. Pharmacokinetic bioequivalence, safety and acceptability of Ornibel®, a new polymer composition contraceptive vaginal ring (etonogestrel/ethinylestradiol 11.00/3.474 mg) compared with Nuvaring®(etonogestrel/ethinylestradiol 11.7/2.7 mg). Eur J Contracept Reprod Health Care 2017; 22: 429-438
  • 106 Roumen FJ, op ten Berg MM, Hoomans EH. The combined contraceptive vaginal ring (NuvaRing®): first experience in daily clinical practice in The Netherlands. Eur J Contracept Reprod Health Care 2006; 11: 14-22
  • 107 Koetsawang S, Gao J, Krishna U. et al. Microdose intravaginal levonorgestrel contraception: A multicentre clinical trial: I. Contraceptive efficacy and side effects. Contraception 1990; 41: 105-124
  • 108 Brache V, Alvarez-Sanchez F, Faundes A. et al. Progestin-only contraceptive rings. Steroids 2000; 65: 687-691
  • 109 Massai R, Quinteros E, Reyes MV. et al. Extended use of a progesterone-releasing vaginal ring in nursing women: a phase II clinical trial. Contraception 2005; 72: 352-357
  • 110 Kerns J, Darney P. Vaginal ring contraception. Contraception 2011; 83: 107-115
  • 111 Sivin I, Mishell jr. DR, Alvarez F. et al. Contraceptive vaginal rings releasing Nestorone® and ethinylestradiol: a 1-year dose-finding trial. Contraception 2005; 71: 122-129
  • 112 Bjarnadóttir RI. Update on contraceptive vaginal rings. Reviews in Gynaecological Practice 2003; 3: 156-159
  • 113 Galzote RM, Rafie S, Teal R. et al. Transdermal delivery of combined hormonal contraception: a review of the current literature. Int J Womens Health 2017; 9: 315
  • 114 Stewart M, Black K. Choosing a combined oral contraceptive pill. Aust Prescr 2015; 38: 6
  • 115 Nath A, Sitruk-Ware R. Progesterone vaginal ring for contraceptive use during lactation. Contraception 2010; 82: 428-434
  • 116 Rivera R, Yacobson I, Grimes D. The mechanism of action of hormonal contraceptives and intrauterine contraceptive devices. Am J Obstet Gynecol 1999; 181: 1263-1269
  • 117 Ballagh SA. Vaginal ring hormone delivery systems in contraception and menopause. Clin Obstet Gynecol 2001; 44: 106-113
  • 118 Wagner M-S, Black A. The Combined Contraceptive Vaginal Ring: an Update. Current Obstetrics and Gynecology Reports 2016; 5: 1-12
  • 119 Gruber CJ. The combined contraceptive vaginal ring (NuvaRing): evaluation of the clinical and pharmacological evidence. Womens Health (Lond) 2006; 2: 351-356
  • 120 Roumen FJ. Review of the combined contraceptive vaginal ring, NuvaRing® . Ther Clin Risk Manag 2008; 4: 441
  • 121 Mulders TM, Dieben TO, Bennink HJC. Ovarian function with a novel combined contraceptive vaginal ring. Hum Reprod 2002; 17: 2594-2599
  • 122 Fan GS, Ren M, Di W. et al. Efficacy and safety of the contraceptive vaginal ring (NuvaRing) compared with a combined oral contraceptive in Chinese women: a 1-year randomised trial. Eur J Contracept Reprod Health Care 2016; 21: 303-309
  • 123 Reproductive Health Supplies Coalition. Progesterone vaginal ring. Brussels, Belgium: Reproductive Health Supplies Coalition; Accessed May 3 2021 at: https://www.rhsupplies.org/fileadmin/uploads/rhsc/Working_Groups/New_Underused_RH_Technologies_Caucus/Documents/Technical_Briefs/rhsc-brief-progesterone_A4.pdf
  • 124 Johansson ED, Sitruk-Ware R. New delivery systems in contraception: vaginal rings. Am J Obstet Gynecol 2004; 190: S54-S59
  • 125 Friend DR. Drug delivery for the treatment of endometriosis and uterine fibroids. Drug Deliv Transl Res 2017; 7: 829-839
  • 126 Parasar P, Ozcan P, Terry KL. Endometriosis: epidemiology, diagnosis and clinical management. Curr Obstet Gynecol Rep 2017; 6: 34-41
  • 127 Pluchino N, Freschi L, Wenger J-M. et al. Innovations in classical hormonal targets for endometriosis. Expert Rev Clin Pharmacol 2016; 9: 317-327
  • 128 Razzi S, Luisi S, Calonaci F. et al. Efficacy of vaginal danazol treatment in women with recurrent deeply infiltrating endometriosis. Fertil Steril 2007; 88: 789-794
  • 129 Ryan IP, Schriock ED, Taylor RN. Isolation, characterization, and comparison of human endometrial and endometriosis cells in vitro. J Clin Endocrinol Metab 1994; 78: 642-649
  • 130 ACOG Committee on Practice Bulletins–Gynecology. ACOG Practice Bulletin No. 51. Chronic pelvic pain. Obstet Gynecol 2004; 103: 589-605
  • 131 Reinecke I, Schultze-Mosgau MH, Nave R. et al. Model‐based dose selection for intravaginal ring formulations releasing anastrozole and levonorgestrel intended for the treatment of endometriosis symptoms. J Clin Pharmacol 2017; 57: 640-651
  • 132 Dunselman G, Vermeulen N, Becker C. et al. ESHRE guideline: management of women with endometriosis. Hum Reprod 2014; 29: 400-412
  • 133 Vercellini P, Barbara G, Somigliana E. et al. Comparison of contraceptive ring and patch for the treatment of symptomatic endometriosis. Fertil Steril 2010; 93: 2150-2161
  • 134 Sartori MGF, Feldner jr. P, Jarmy-Di Bella Z. et al. Sexual steroids in urogynecology. Climacteric 2011; 14: 5-14
  • 135 Palacios S, Nappi R, Bruyniks N. et al. The European Vulvovaginal Epidemiological Survey (EVES): prevalence, symptoms and impact of vulvovaginal atrophy of menopause. Climacteric 2018; 21: 286-291
  • 136 Van Keep P, Utian W, Vermeulen A. Potency and hepato-cellular Effects of Oestrogens after oral, percutaneous, and subcutaneous Administration. In: Utian WH, van Keep PA, Vermeulen A. eds. The controversial Climacteric. Dordrecht: Springer; 1982: 103-125
  • 137 Antisell J, Poon T, Borey A, Briddell J, Palesch S. Hormone Delivery System: The Contraceptive Ring. Ithaca, New York: Cornell University Library; 2006. Accessed May 3 2021 at: https://ecommons.cornell.edu/handle/1813/3060
  • 138 Kuhl H. Pharmacology of estrogens and progestogens: influence of different routes of administration. Climacteric 2005; 8: 3-63
  • 139 Nabahi S. Intravaginal drug delivery device. Google Patents; 1998
  • 140 Cicinelli E. Intravaginal oestrogen and progestin administration: advantages and disadvantages. Best Pract Res Clin Obstet Gynaecol 2008; 22: 391-405
  • 141 Casper F, Petri E. Local treatment of urogenital atrophy with an estradiol-releasing vaginal ring: a comparative and a placebo-controlled multicenter study. Vaginal Ring Study Group. Int Urogynecol J Pelvic Floor Dysfunct 1999; 10: 171-176
  • 142 Hill DA, Crider M, Hill SR. Hormone therapy and other treatments for symptoms of menopause. Am Fam Physician 2016; 94: 884-889
  • 143 Jilka RL, Takahashi K, Munshi M. et al. Loss of estrogen upregulates osteoblastogenesis in the murine bone marrow. Evidence for autonomy from factors released during bone resorption. J Clin Invest 1998; 101: 1942-1950
  • 144 Khosla S, Oursler MJ, Monroe DG. Estrogen and the skeleton. Trends Endocrinol Metab 2012; 23: 576-581
  • 145 Almeida M, Laurent MR, Dubois V. et al. Estrogens and androgens in skeletal physiology and pathophysiology. Physiol Rev 2017; 97: 135-187
  • 146 Paganini-Hill A, Henderson VW. Estrogen deficiency and risk of Alzheimerʼs disease in women. Am J Epidemiol 1994; 140: 256-261
  • 147 Lindahl SH. Reviewing the options for local estrogen treatment of vaginal atrophy. Int J Womens Health 2014; 6: 307
  • 148 Management of symptomatic vulvovaginal atrophy: 2013 position statement of The North American Menopause Society. Menopause 2013; 20: 888-902 quiz 903–904
  • 149 Warner Chilcott Laboratories. Estrace (estradiol vaginal cream, USP, 0.01 %) [prescribing information]. Rockaway, NJ: Warner Chilcott Laboratories; 2011
  • 150 Food and Drug Administration. NDA 020216/S-067. Supplement Approval. Accessed May 3, 2021 at: https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2011/020216s067ltr.pdf
  • 151 Rybacki JJ. DOSAGE AND ADMINISTRATION. Published at 6/21/2017 on RxList website. Accessed October 29, 2020 at: https://www.rxlist.com/estragyn-drug.htm
  • 152 Neidecker MV. Patterns of Medication Use and Related Health Care Service Utilization Associated with Vaginal Estrogen Therapy in Medicaid-Enrolled Women [Diss.]. The Ohio State University; 2009
  • 153 [Anonym] Estring. New York: Pharmacia & Upjohn Co; 2008
  • 154 [Anonym] Femring. Rockaway, NJ: Warner Chilcott Inc; 2005
  • 155 Smith P, Heimer G, Lindskog M. et al. Oestradiol-releasing vaginal ring for treatment of postmenopausal urogenital atrophy. Maturitas 1993; 16: 145-154
  • 156 Barentsen R, van de Weijer PH, Schram JH. Continuous low dose estradiol released from a vaginal ring versus estriol vaginal cream for urogenital atrophy. Eur J Obstet Gynecol Reprod Biol 1997; 71: 73-80
  • 157 Holmgren P-Å, Lindskog M, Von Schoultz B. Vaginal rings for continuous low-dose release of oestradiol in the treatment of urogenital atrophy. Maturitas 1989; 11: 55-63
  • 158 Schmidt G, Andersson S-B, Nordle Ö. et al. Release of 17-beta-oestradiol from a vaginal ring in postmenopausal women: pharmacokinetic evaluation. Gynecol Obstet Invest 1994; 38: 253-260
  • 159 Suckling J, Lethaby A, Kennedy R. Local oestrogen for vaginal atrophy in postmenopausal women. Cochrane Database Syst Rev 2006; (04) CD001500
  • 160 Crandall CJ, Hovey KM, Andrews CA. et al. Breast cancer, endometrial cancer, and cardiovascular events in participants who used vaginal estrogen in the Womenʼs Health Initiative Observational Study. Menopause 2018; 25: 11-20
  • 161 Krause M, Wheeler 2nd TL, Snyder TE. et al. Local Effects of Vaginally Administered Estrogen Therapy: A Review. J Pelvic Med Surg 2009; 15: 105-114
  • 162 Castelo-Branco C, Cancelo MJ, Villero J. et al. Management of post-menopausal vaginal atrophy and atrophic vaginitis. Maturitas 2005; 52: 46-52
  • 163 Naumova I, Castelo-Branco C. Current treatment options for postmenopausal vaginal atrophy. Int J Womens Health 2018; 10: 387
  • 164 Calleja-Agius J, Brincat M. Urogenital atrophy. Climacteric 2009; 12: 279-285
  • 165 Bell RJ, Rizvi F, Islam RM. et al. A systematic review of intravaginal testosterone for the treatment of vulvovaginal atrophy. Menopause 2018; 25: 704-709
  • 166 Robinson D, Cardozo LD. The role of estrogens in female lower urinary tract dysfunction. Urology 2003; 62: 45-51
  • 167 Pruthi S, Simon JA, Early AP. Current overview of the management of urogenital atrophy in women with breast cancer. Breast J 2011; 17: 403-408
  • 168 Willhite LA, OʼConnell MB. Urogenital atrophy: prevention and treatment. Pharmacotherapy 2001; 21: 464-480
  • 169 Leiblum S, Bachmann G, Kemmann E. et al. Vaginal atrophy in the postmenopausal woman: the importance of sexual activity and hormones. JAMA 1983; 249: 2195-2198
  • 170 Moegele M, Buchholz S, Seitz S. et al. Vaginal Estrogen Therapy for Patients with Breast Cancer. Geburtshilfe Frauenheilkd 2013; 73: 1017-1022
  • 171 Huang X, Brazel CS. On the importance and mechanisms of burst release in matrix-controlled drug delivery systems. J Control Release 2001; 73: 121-136
  • 172 Helbling IM, Ibarra JC, Luna JA. The use of cellulose membrane to eliminate burst release from intravaginal rings. AAPS J 2016; 18: 960-971
  • 173 Tønnesen HH, Karlsen J. Alginate in drug delivery systems. Drug Dev Ind Pharm 2002; 28: 621-630
  • 174 Sershen S, West J. Implantable, polymeric systems for modulated drug delivery. Adv Drug Deliv Rev 2002; 54: 1225-1235
  • 175 Tiedeken M, Westhoff CL, Cohen A. et al. Bone turnover markers in women participating in a dose-finding trial of a contraceptive vaginal ring releasing Nestorone and estradiol. Contraception 2019; 99: 329-334
  • 176 Stifani BM, Plagianos M, Vieira CS. et al. Factors associated with nonadherence to instructions for using the Nestorone®/ethinyl estradiol contraceptive vaginal ring. Contraception 2018; 97: 415-421
  • 177 Nel A, Bekker L-G, Bukusi E. et al. Safety, acceptability and adherence of dapivirine vaginal ring in a microbicide clinical trial conducted in multiple countries in Sub-Saharan Africa. PloS One 2016; 11: e0147743
  • 178 Sefton MV. Implantable pumps. Medical Applications of Controlled Release 1984; 1: 129-158
  • 179 Conte U, Maggi L. A flexible technology for the linear, pulsatile and delayed release of drugs, allowing for easy accommodation of difficult in vitro targets. J Control Release 2000; 64: 263-268
  • 180 Lee E, Kim S, Kim S. et al. Drug release from hydrogel devices with ratecontrolling barriers. J Membr Sci 1980; 7: 293-303
  • 181 Yang L, Fassihi R. Modulation of diclofenac release from a totally soluble controlled release drug delivery system. J Control Release 1997; 44: 135-140
  • 182 Hildgen P, McMullen J. A new gradient matrix: formulation and characterization. J Control Release 1995; 34: 263-271
  • 183 Lu S, Anseth KS. Photopolymerization of multilaminated poly (HEMA) hydrogels for controlled release. J Control Release 1999; 57: 291-300
  • 184 Lu S, Ramirez WF, Anseth KS. Photopolymerized, multilaminated matrix devices with optimized nonuniform initial concentration profiles to control drug release. J Pharm Sci 2000; 89: 45-51
  • 185 Qiu Y, Chidambaram N, Flood K. Design and evaluation of layered diffusional matrices for zero-order sustained-release. J Control Release 1998; 51: 123-130
  • 186 Narasimhan B, Peppas NA. Molecular analysis of drug delivery systems controlled by dissolution of the polymer carrier. J Pharm Sci 1997; 86: 297-304
  • 187 Park M, Park CG, Lee SH. et al. Polymeric tube-shaped devices with controlled geometry for programmed drug delivery. Macromol Res 2012; 20: 960-967
  • 188 Johnson TJ. Antiretroviral eluting intravaginal Rings to prevent the sexual Transmission of HIV. Salt Lake City, Utah: The University of Utah; 2012
  • 189 Jensen JT. Vaginal ring delivery of selective progesterone receptor modulators for contraception. Contraception 2013; 87: 314-318
  • 190 Singer R, Mawson P, Derby N. et al. An intravaginal ring that releases the NNRTI MIV-150 reduces SHIV transmission in macaques. Sci Transl Med 2012; 4: 150ra123
  • 191 Kenney J, Aravantinou M, Singer R. et al. An antiretroviral/zinc combination gel provides 24 hours of complete protection against vaginal SHIV infection in macaques. PloS One 2011; 6: e15835
  • 192 Dobard C, Sharma S, Martin A. et al. Durable protection from vaginal simian-human immunodeficiency virus infection in macaques by tenofovir gel and its relationship to drug levels in tissue. J Virol 2012; 86: 718-725
  • 193 Derby N, Zydowsky T, Robbiani M. In search of the optimal delivery method for anti-HIV microbicides: are intravaginal rings the way forward?. Expert Rev Anti Infect Ther 2013; 11: 5-8
  • 194 Gao S, Tang G, Hua D. et al. Stimuli-responsive bio-based polymeric systems and their applications. J Mater Chem B 2019; 7: 709-729
  • 195 Jiang J, Xie J, Ma B. et al. Mussel-inspired protein-mediated surface functionalization of electrospun nanofibers for pH-responsive drug delivery. Acta Biomater 2014; 10: 1324-1332

Zoom Image
Fig. 1 Classification of hormones according to their chemical nature.
Zoom Image
Fig. 2 Intravaginal ring designs.
Zoom Image
Fig. 3a Single-drug matrix IVR, b multi-drug segmented matrix IVR, c single-drug core IVR, d single-drug incomplete core IVR, e multi-drug continuous core continuous RCM, f multi-drug segmented core continuous RCM, g multi-drug segmented core segmented RCM, h sandwich IVR, i tablet/pod/rod insert IVR, j core-matrix (hybrid) IVR.
Q=sqrt[D_p(2A-C_p)C_pt]Zoom Image
Q = (D_p C_p t) ∕ hZoom Image
Q = (2pi D_p C_p L t) ∕ ln(b/a)Zoom Image
Zoom Image
Fig. 4 Cumulative release profile of a matrix (homogeneous dispersion) IVRs, b core reservoir and pod IVRs, and c sandwich reservoir IVRs.
Zoom Image
Fig. 5 Diagram showing applications for IVRs.