142
Views
63
CrossRef citations to date
0
Altmetric
Review

Intravaginal rings as delivery systems for microbicides and multipurpose prevention technologies

, , &
Pages 695-708 | Published online: 21 Oct 2013

Abstract

There is a renewed interest in delivering pharmaceutical products via intravaginal rings (IVRs). IVRs are flexible torus-shaped drug delivery systems that can be easily inserted and removed by the woman and that provide both sustained and controlled drug release, lasting for several weeks to several months. In terms of women’s health care products, it has been established that IVRs effectively deliver contraceptive steroids and steroids for the treatment of postmenopausal vaginal atrophy. A novel application for IVRs is the delivery of antiretroviral drugs for the prevention of human immunodeficiency virus (HIV) genital infection. Microbicides are antiviral drugs delivered topically for HIV prevention. Recent reviews of microbicide IVRs have focused on technologies in development and optimizing ring design. IVRs have several advantages, including the ability to deliver sustained drug doses for long periods of time while bypassing first pass metabolism in the gut. IVRs are discreet, woman-controlled, and do not require a trained provider for placement or fitting. Previous data support that women and their male sexual partners find IVRs highly acceptable. Multipurpose prevention technology (MPT) products provide protection against unintended/mistimed pregnancy and reproductive tract infections, including HIV. Several MPT IVRs are currently in development. Early clinical testing of new microbicide and MPT IVRs will require a focus on safety, pharmacokinetics and pharmacodynamics. Specifically, IVRs will have to deliver tissue concentrations of drugs that are pharmacodynamically active, do not cause mucosal alterations or inflammation, and do not change the resident microbiota. The emergence of resistance to antiretrovirals will need to be investigated. IVRs should not disrupt intercourse or have high rates of expulsion. Herein, we reviewed the microbicide and MPT IVRs currently in development, with a focus on the clinical aspects of IVR assessment and the challenges facing microbicide and MPT IVR product development, clinical testing, and implementation. The information in this review was drawn from PubMed searches and a recent microbicide/MPT product development workshop organized by CONRAD.

Introduction

Globally, 34 million people are currently infected with human immunodeficiency virus (HIV), and incident infections are more common in women than men, especially in sub-Saharan Africa.Citation1 Recent data from the Microbicide Trials Network’s Vaginal and Oral Interventions to Control the Epidemic (VOICE) study revealed HIV incidence at the clinical sites as high as 9%, especially in young, single women.Citation2 The prevalence of HIV in South Africa (SA) has increased from 10.6% in 2008 to 12.3% in 2012, according to data presented recently at the 6th SA AIDS Conference in Durban.Citation3 Condom use “at last sex” has fallen in all age groups, most dramatically, from 85.2% to 67.4% among men aged 15 to 24 and from 66.5% to 51% in women of the same age.Citation3 This highlights that the need to develop and implement preventive strategies continues to be one of the highest public health research priorities.

Microbicides are topical products designed to prevent HIV acquisition when applied to the cervicovaginal or rectal mucosa. The first successful microbicide was tenofovir (TFV) 1% gel. Pericoital use of TFV 1% vaginal gel was demonstrated to be safe and partially effective at reducing HIV-1 and herpes simplex virus-2 (HSV-2) incidence in the Centre for the AIDS Programme of Research in South Africa (CAPRISA) 004 Phase IIb study.Citation4 In addition to showing effectiveness in this landmark study, TFV has shown efficacy in preventing infection in macaque and humanized mouse models and in several clinical trials, after oral administration.Citation5Citation8 TFV gel was shown to provide 39%–54% protection from HIV acquisition, depending on adherence and the time of use.Citation4 Further analysis of efficacy among drug compliers suggests levels of protection between 74% and 90%.Citation9,Citation10 Adherence and protocol compliance have plagued microbicide trials, and new, less user-dependent dosage forms and delivery systems are needed if significant effectiveness is to be attained. Intravaginal rings (IVRs) are intended to provide continuous, discreet protection without the need to disrupt the sex act. There is hope that they represent a more acceptable form of microbicide delivery, thereby increasing adherence and anti-HIV effectiveness.

Almost half of all pregnancies worldwide, estimated to be over 100 million annually, are unintended.Citation11Citation13 Despite the existence of a variety of effective contraceptives available, discontinuation or nonuse remains high, primarily due to cost, side effects, inconvenient dosing schedules, poor access to prescription products, and/or poor acceptance of the method by the male partner, resulting in the unacceptably high rate of unintended or mistimed pregnancies.Citation14 Statistics clearly show an unmet need for highly effective contraception, especially in less developed countries, where 99% of worldwide maternal deaths occur.Citation12 Not surprisingly, these countries, especially the ones of sub-Saharan Africa and south Asia, are also at the core of the acquired immunodeficiency syndrome (AIDS) epidemic.

Poverty, malnutrition, lack of education, and gender inequality fuel both unplanned pregnancies and HIV transmission. There are a significant number of women, especially in less developed countries, needing protection against sexually transmitted infections (STIs), in particular HIV/AIDS, and long-term, highly effective contraceptive methods to prevent unplanned or mistimed pregnancies and provide optimal birth spacing and family size. Highly effective contraceptives (eg, sterilization, intrauterine devices, and hormonal contraceptives) typically provide little or no protection against STIs, while the barrier methods (eg, condoms) have unacceptably high contraceptive failure rates with typical use. Therefore, there is an urgent need to develop multipurpose prevention technologies (MPTs) providing both contraception and microbicidal activity, that are safe, highly effective, acceptable, and low-cost, especially ones that are suitable for use in less developed countries.

In the past, the main strategies for developing combined HIV prevention and contraceptive technologies were the use of physical barriers.Citation15 The physical barriers comprise male and female condoms, diaphragms, and cervical caps. The typical contraceptive use failure rates for these methods range from 15%–30%.Citation16 Physical barriers are heavily dependent on user adherence and partner cooperation and, in most cases, need to be applied immediately before intercourse. Physical barriers for contraception, while having few side effects, offer none of the health benefits typically found with hormonal contraceptives, such as reduction in menstrual blood loss.Citation17 In terms of HIV prevention, a randomized controlled trial of the diaphragm showed no further reduction of the rates of HIV or STI male-to-female transmission over the condom.Citation18

Recently, there has been a renewed interest in developing MPT IVRs. IVRs are flexible torus-shaped drug delivery systems that can be easily inserted and removed by the woman and provide both sustained and controlled drug release, lasting for several weeks to several months. IVRs, which deliver contraceptive steroids and steroids for the treatment of postmenopausal vaginal atrophy, have already been approved and are available worldwide. Antiretroviral (ARV) medications can also be released from IVRs, at concentrations that are expected to prevent the acquisition of HIV-1.Citation19

Herein, we review the history and advantages of IVRs, microbicides, and the MPT IVRs currently in development, focusing on the clinical aspects of IVR evaluation and the challenges facing microbicide and MPT IVR product development, clinical testing, and implementation.

History

The first report of vaginally administered drugs was published in 1918 by Macht, who demonstrated that several drugs, including morphine, atropine and potassium iodide, could be vaginally absorbed.Citation20 The field of vaginal drug delivery has since flourished to include a diversity of applications, with numerous marketed products currently available for contraception (eg, IVRs: NuvaRing® [etonogestrel/ethinyl estradiol] [Merck & Co, Inc, Whitehouse Station, NJ, USA] and Progering® [progesterone] [Silesia Laboatorios, Santiago, Chile]; film: Vaginal Contraceptive Film® [nonoxynol-9] [VCF; Apothecus Pharmaceutical Corp, Oyster Bay, NY,USA]; and the sponge: Today® sponge [nonoxynol-9] [Mayer Laboratories Inc, Berkeley, CA, USA]), for hormone replacement therapy (IVRs: Femring® [estradiol acetate] [Warner Chilcott LLC, Rockaway, NJ, USA], Estring® [estradiol] [Pfizer, Inc, New York, NY, USA], and Fertiring® [progesterone] [Silesia Laboatorios]; creams: Premarin® [conjugated estrogens] [Pfizer, Inc, New York, NY, USA], Estrace® [estradiol] [Shire plc, Dublin, Ireland], Estrasorb™ [estradiol] [Medicis Pharmaceuticals Corp, Scottsdale, AZ, USA], and Ogen® [estropipate] [Pfizer, Inc]; and tablets: Premarin® and Vagifem® [estradiol] [Novo Nordisk, Bagsværd, Denmark]), for antimicrobial treatment (gel: Metrogel® [metronidazole] [Galderma Laboratories, Fort Worth, TX, USA]; creams: Cleocin® [clindamycin] [Pfizer, Inc], Clindesse® [Ther-Rx Corp, Chesterfield, MO, USA] or ClindaMax® [clindamycin phosphate] [Nycomed, Zurich, Switzerland], Gyne-Lotrimin® [Schering-Plough HealthCare Products, Inc, Memphis, TN, USA] or Mycelex® [Bayer HealthCare Pharmaceuticals Corp, Montville, NJ, USA] or Femcare® [clotrimazole] [Schering-Plough HealthCare Products, Inc, Memphis, TN, USA], Gynezol® [Parke-Med, SA] or Femstat® [butoconazole] [Bayer HealthCare Pharmaceuticals Corp], Monistat® [micon-azole nitrate] [Insight Pharmaceutical LLC, Trevose, PA, USA], Vagistat-1® [tioconazole] [Bristol-Myers Squibb, New York, NY, USA], and Terazol® [terconazole] [Janssen Pharmaceuticals, Inc, Titusville, NJ, USA]; suppositories: Cleocin, Terazol, and Monistat; tablets: Gyne-Lotrimin or Mycelex or Femcare), cervical ripening (vaginal insert: Cervidil® [dinoprostone] [Forest Laboratories, Inc, New York, NY, USA] and gel: Prepidil® [dinoprostone] [Pfizer, Inc]), and pregnancy termination (suppositories: Prostin E2® [dinoprostone] [Pfizer, Inc] and tablets: Cytotec® [misoprostol] [GD Searle and Co, Skokie, IL, USA]). The development of vaginal drug delivery systems for new uses, most notably as topical microbicides, continues to expand and progress at a fervent pace.

Several decades following the introduction of vaginal drug delivery, IVR development efforts began in earnest, in 1966, with the discovery that steroids could be delivered, through silicone elastomers, at constant rates for several days.Citation21 The first IVR tested clinically in the 1970s was for the delivery of medroxyprogesterone acetate for contraception.Citation22,Citation23 Also in the 1970s, the World Health Organization (WHO) developed three contraceptive rings containing either progesterone, norethisterone, or levonorgestrel (LNG). The progesterone and norethisterone rings showed higher than desired side effects (eg, menstrual irregularities) and pregnancy rates,Citation24 so the 90-day LNG IVR was selected for further development, ultimately showing a 3.6% pregnancy rate among women who wore the IVR continuously for 1 year (replacing the IVR every 90 days).Citation25 While effective, the IVR was firm and relatively inflexible, leading some women to experience expulsion, vaginal abrasions/ulcers and asymptomatic vaginal irritationCitation26,Citation27 and was subsequently redesigned to be smaller and more flexible. The redesigned LNG IVR was shown not to have adverse effects on the vaginal epithelium,Citation28 but a lack of funding precluded further testing.

Today, there are two contraceptive rings commercially available: the NuvaRing (available in more than 40 countries worldwide), and the Progering (available in Chile, Peru, Bolivia, Ecuador, Guatemala, and Honduras). The NuvaRing is made of ethylene vinyl acetate (EVA), a thermoplastic that allows the IVR to be 4 mm thick in cross-sectional diameter, which is significantly thinner than silicone IVRs. It delivers 120 μg of etonogestrel and 15 μg ethinyl estradiol (E2) per day. The NuvaRing is approved for 3 weeks of use, with removal for 1 week to allow for menses. However, a small study using vaginal ultrasound and serum progesterone as markers of ovulation supports that ovulation is suppressed for 5 weeks with the NuvaRing.Citation29

Progering, which is made of silicone elastomer and approved for use by lactating women, delivers 10 mg of progesterone per day for 3 months. Other hormonal IVRs have been developed over the years but were not brought to market for various reasons, usually related to undesirable side effects (eg, menstrual irregularities and reduction of high-density lipoprotein cholesterol levels). Work is ongoing in contraceptive IVR development, including the Population Council’s Nestorone® (NES) and NES/E2 IVRs, the latter of which is intended to last 1 year using a 21-day-in and 7-day-out dosing cycle.Citation30

Steroid-releasing IVRs have also been developed for hormone supplementation or replacement therapy, with a handful of silicone elastomer-based products commercially available. The Fertiring, similar to the Progering, releases progesterone but for the indication of hormone supplementation and pregnancy maintenance during in vitro fertilization. The Estring (7.5 μg/day estradiol) and Femring (0.05 mg/d or 0.1 mg/d estradiol acetate dose options) are both 90-day IVRs for use in estrogen replacement therapy in postmenopausal women with urogenital atrophy.

The first IVR developed preclinically for microbicide release was a silicone matrix IVR designed to release the nonspecific microbicide/spermicide surfactant nonoxynol-9.Citation31 Development of this ring was halted when a clinical trial showed that frequent use of nonoxynol-9 could increase the risk of HIV-1 transmission.Citation32 In the decade since, the development of microbicide and more recently, MPT IVRs has rapidly expanded to include the development of numerous classes of microbicide candidates, including ARVs with different mechanisms of action. Because of the physiochemical diversity of these drug candidates, this has also led to a boom in the development of new IVR designs and technologies that may better deliver these drugs or drug combinations.Citation33

Advantages of IVRs for delivery of microbicides and MPTs

The IVR is a unique and appealing vaginal drug delivery system, as it is female initiated and controlled, and provides sustained drug release, in the order of weeks to months. Some of the major advantages of IVRs for the development of microbicides and MPT products include the local delivery of microbicides, the maintenance of steady-state hormone serum concentrations, extended dosing regimens that do not require daily action, and the discreet woman-controlled use.

Local delivery of microbicides

IVRs can effectively deliver high concentrations of microbicides directly to the vagina and specifically, to the cells and tissues targeted by most of the microbicide drug candidates in development.Citation34Citation37 This local delivery also allows for substantially lower drug doses compared with oral dosing regimens. The clinical data on the delivery of TFV via an oral pill (using 300 mg dose of the prodrug tenofovir disoproxil fumarate) or via vaginally applied TFV 1% gel (~40 mg TFV) in women show that the vaginal tissue concentrations of tenofovir-diphosphate, the active metabolite of TFV, were ≥130-fold higher with vaginal dosing compared with oral dosing.Citation38 Compared with vaginal gels, the rate and duration of microbicide delivery from IVRs to the vagina is more controlled, providing reduced doses that may better maintain prophylactic concentrations for longer duration. Moreover, the local delivery of microbicides from IVRs also helps reduce the systemic exposure to these ARVs, potentially improving their safety profiles (ie, reduced systemic side effects) and minimizing the risk for developing resistance. Pharmacokinetic (PK) studies will need to confirm that the microbicide doses delivered from IVRs remain high in the genital compartment but low in the systemic circulation, to reduce the risk of developing ARV resistance, as this is a concern with oral dosing.

Maintenance of steady-state hormone serum concentrations

For microbicide/contraceptive MPTs, IVRs are capable of simultaneously providing the local delivery of microbicides and the sustained systemic delivery of contraceptive hormonal steroids. Delivery of hormones via IVRs eliminates the burst effect seen with injectable contraceptivesCitation39,Citation40 and minimizes the daily peak and trough fluctuations seen with orally administered hormones.Citation41 For example, NuvaRing use results in maximum serum hormone concentrations that are 30%–40% lower than those seen with orally administered contraceptive hormones.Citation42 Further, by avoiding first pass metabolism in the gut, IVR users typically have fewer hormonal side effects than do oral hormonal contraceptive users, including adverse effects on the coagulation system.Citation43Citation45 Many ARVs either induce or inhibit hepatic cytochrome P450 enzymes, which are required for the metabolism of commonly used contraceptive steroid hormones.Citation46,Citation47 In addition, vaginal drug absorption, unlike oral dosing, is not altered by gastrointestinal disturbances.

Extended dosing regimens possible with IVRs

The clinical evaluation of effectiveness in the microbicide and oral preexposure prophylaxis (PrEP) trials, to date, has struggled greatly with the issue of poor adherence, which might be attributed in part, to short action and daily or pericoital dosing regimens. Adherence to vaginal gel dosing in the microbicide gel trials has been a major challenge. TFV 1% gel was shown to be 39% effective overall in the CAPRISA 004 trial when dosed pericoitally, with the effectiveness increasing to 54%–90% in the high-adherence users.Citation4,Citation9,Citation10 TFV 1% gel, dosed daily in a coitally-dissociated regimen, was determined to be ineffective in the VOICE trial, with the wide consensus that poor adherence was a major contributor to the gel’s failure.Citation2 The analysis of drug levels in the VOICE participants’ blood samples showed detectable levels of TFV in only 23% of the participants in the TFV gel arm, indicating that only one out of four participants actually used the product.Citation2 Similarly, the poor adherence to daily oral PrEP dosing regimens has also affected efficacy in clinical efficacy studies.Citation48 In the FEM-PrEP study, for example, the self-report and pill count showed >90% and >80% adherence, respectively, while detection of drug in blood indicated that no more than 30% of women used the product as instructed.Citation48

Although daily dosed combined oral contraceptives are the most commonly used contraceptive method worldwide, this dosing regimen remains problematic for many women, with nonadherence and discontinuations being major contributors to contraceptive failures.Citation49 Recent initiatives have focused on long-acting reversible contraceptive (LARC) methods because it has been shown that as the uptake of LARC increases, unintended pregnancies decrease.Citation50 While not typically considered LARC methods, the currently available contraceptive IVRs have durations of action of 3 weeks–3 months,Citation51 eliminating the need for daily or coitally-dependent action. Because a woman can insert an IVR and forget about it for a determined time period, there is a great potential to improve adherence to product use with IVRs.Citation52

Though it remains to be seen whether IVRs will in fact improve adherence to microbicide use, a study comparing the adherence to contraceptive IVRs with adherence to daily oral contraceptive pills provides encouraging results.Citation52 IVR users were more likely to report perfect use compared with daily pill users.Citation52 The IVRs that are currently in contraceptive, microbicide, or MPT development are being designed to have a 30–365 day duration.Citation33

Discreet woman-controlled use

The most disadvantaged women in society are also at highest risk for HIV. These women include commercial sex workers, women who cannot negotiate safer sex, women who cannot ensure their partners’ faithfulness, and those who often cannot leave a relationship, for economic or social reasons. Therefore, the development of microbicide and MPT products that require no partner cooperation and that can be used discreetly is a priority. Many of the highly effective LARC methods, such as the contraceptive implant or the contraceptive intrauterine system, require insertion by a trained provider, which may be a barrier to access in resource-constrained areas.Citation53 IVRs, on the other hand, provide the promising balance of not requiring daily action and being woman initiated and controlled.

Microbicide and MPT IVRs in development

Microbicide IVRs

Currently, the most clinically advanced microbicide IVR is for the 28-day delivery of dapivirine (DPV), a nonnucleoside reverse transcriptase inhibitor of HIV developed by the International Partnership for Microbicides (IPM). While several types of DPV IVRs (including matrix- and reservoir-type IVRs, all comprised of silicone elastomer) were evaluated in the early clinical studies,Citation36,Citation54,Citation55 the matrix DPV ring (a simpler ring design with a 56 mm outer diameter and a 7.6 mm cross-sectional diameter) was selected for further development and is currently in Phase III testing, with results expected in 2015. IPM is also conducting clinical trials of silicone IVRs containing maraviroc, a chemokine receptor type 5 (CCR5) entry inhibitor, alone or in combination with DPV.Citation56

Building on the success of the leading topical microbicide product TFV 1% gel, which has demonstrated prophylactic effectiveness in both animal models and women (CAPRISA 004), long-acting TFV-based IVRs are being developed for the 3-month delivery of TFV, with Phase I testing planned later this year. These IVRs, developed by CONRAD in collaboration with Dr Patrick Kiser (University of Utah and Northwestern University), are comprised of polyurethane, which is a more versatile thermoplastic than the EVA used in the NuvaRing, yet have dimensions more similar to the NuvaRing than to the bulky silicone-based IVRs (the TFV IVR has a 5.5 mm cross-sectional diameter and a 55 mm outer diameter). Notably, in sheep, the drug concentrations throughout 90 days of treatment with TFV IVRs were similar to or higher than those seen with TFV 1% gel.Citation19,Citation57 A 1-month tenofovir disoproxil fumarate IVR that is similar in design to the TFV IVR is also nearing Phase I testing. This IVR has been tested in a nonhuman primate HIV efficacy model and was recently reported to demonstrate 100% protection against simian–human immunodeficiency virus (SHIV) acquisition in a repeated low-dose challenge model.Citation58

Several additional microbicide IVRs are in preclinical development and have been recently reviewed elsewhere.Citation1,Citation33 A detailed table of past, ongoing, and future microbicide trials, including IVR trials is available at http://www.avac.org/ht/a/GetDocumentAction/i/3109.

MPT IVRs

Worldwide, there is a significant unmet need for both effective contraception and medications to prevent genital acquisition of STIs, namely HIV. The most advanced MPT IVR in development is the 90-day TFV/LNG IVR for the dual purpose of HIV prevention and contraception, which is expected to enter Phase I testing later in 2013. Developed by CONRAD in collaboration with Dr Kiser, this IVR builds on the development of the TFV IVR. The combination IVR has been demonstrated preclinically to provide steady-state dosing of approximately 10 mg/d TFV (similar to the TFV-only IVR) and 20 μg/d LNG for 90 days, both in vitro and in animal models.Citation19,Citation57 This low-LNG dose was selected based on the previous work done by the WHO in the 1970s, described above, in which 20 μg/d LNG was found to be effective and acceptable, working primarily through local effects (eg, cervical mucus thickening) rather than by disrupting normal ovulation.Citation25,Citation59 Previous studies of the 20 μg/d LNG IVR found that 40% to 60% of users ovulated normally.Citation24,Citation60 The systemic levels of LNG found in the 20 μg/d LNG IVR users were in the same low range as those measured among users of the 20 μg/d LNG intrauterine system (IUS) (Mirena®; Bayer HealthCare Pharmaceuticals) (0.2–0.5 ng/mL)Citation61Citation63 and the 14 μg/d LNG IUS (Skyla™; Bayer HealthCare Pharmaceuticals) (0.1–0.2 ng/mL).Citation64 These highly effective and acceptable contraceptive products work primarily by exerting local effects on the cervical mucus,Citation65 which we believe will be the primary contraceptive mechanism of action of the proposed TFV/LNG IVR. The additional contraceptive action from the LNG IUS likely results from local effects on the endometrium,Citation63 impairment of the fallopian tube transport of sperm and ova, and some level of ovulation suppression.Citation66 In vitro data even support that the LNG levels delivered from the LNG IUS interfere with sperm–zona pellucida interactions.Citation67 We plan to assay cervical mucus LNG levels in early phase PK studies and compare these levels in TFV/LNG IVR users with the levels found in LNG IUS users.Citation65

IPM is also involved in the early development stages of a microbicide/contraceptive MPT IVR comprising the combination DPV/LNG; the LNG dose targeted for this IVR is 35 or 70 μg/d, which is expected to have both systemic (ie, anovulatory) and local contraceptive effects.

Other MPT IVRs in preclinical development include the Population Council’s Medivir-150/zinc acetate IVR (for prevention of HIV and HSV-2) and Medivir-150/zinc acetate/LNG IVR (for the same indications plus contraception). CONRAD also initiated the development of an acyclovir/TFV IVR for the treatment of HSV-2 and prevention of HIV,Citation68 but the development was halted due to reallocation of funding.

Objective assessment of product adherence with long-term use of IVRs

As discussed above, the monitoring of product adherence in microbicide/PrEP efficacy trials is a major issue facing the field currently, and while great strides are being made to develop better measures of adherence for gel products, monitoring the correct and consistent use of long-acting IVRs will present new, additional challenges. Some groups have proposed assessment of the presence of the biofilm on the surface of IVRs to monitor adherence to the dosing regimen. However, it is not known how quickly a biofilm forms on the IVR surface, as the limited number of studies have only tested 28 days of use.Citation69,Citation70 In comparison, urinary catheters have been shown to become colonized with bacteria, in the sterile bladder, within 48 hours.Citation71 Biofilms form within 60 hours on intrauterine devices, in the relatively sterile uterine cavity.Citation72 This suggests that monitoring vaginal bacteria could determine whether an IVR was inserted vaginally but would unlikely be able to differentiate hours from weeks of use or determine the duration of use.

An alternative to assessment of the vaginal biofilms on IVRs would be to quantify the level of drug substance left in an IVR after a participant returns the IVR. This method is currently being employed in the IPM Phase III study of the DPV IVR. The current TFV and TFV/LNG IVR prototypes about to enter Phase I testing are transparent, revealing a white drug-loaded core that transitions to clear as TFV is released during use; similar to determining the residual drug content in the returned IVRs, the level of pigment/opacity in these IVRs could potentially be standardized to correlate with duration of usage, potentially providing a new, rapid, and more field-ready measure of IVR adherence.

However, all of these methods described thus far have the limitation that they do not provide time-associated adherence data, and given that IVRs may be removed intermittently during use, time-stamped measures would be most useful.

Clinical assessment of IVRs

The need for microbicide and MPT products is clear. STIs and unintended/mistimed pregnancies are leading causes of morbidity and mortality among women worldwide. Progress has been made in the preclinical setting to develop both IVRs, which can deliver concentrations of contraceptive hormones, and ARVs, which are expected to be effective in preventing pregnancy and HIV. Going forward, Phase I testing in the clinic must include an evaluation of product safety and acceptability, particularly in women in developing, resource-poor countries, who are at high risk of HIV acquisition.

Acceptability

There is high acceptance of IVRs for the delivery of contraceptive hormones.Citation73,Citation74 An international study of the NuvaRing found that approximately 97% of women reported they would recommend the IVR to their friends.Citation75 Perhaps the best assessment of user acceptability was reported in a study of women who were currently using oral contraceptive pills and who reported being happy with this method.Citation73 These oral contraceptive pill users were transitioned and randomized to either the contraceptive patch or NuvaRing.Citation73 The women who transitioned to the IVR were significantly more likely to plan to continue this method than were the women who transitioned to the contraceptive patch.Citation73 The acceptability assessments for the IVR were significantly higher than for the oral contraceptive pill (P < 0.001), and the women randomized to receive the contraceptive IVR reported shorter and less painful menses than did the women randomized to the contraceptive patch.Citation73 These data are in accordance with another head-to-head study of the contraceptive IVR versus oral contraceptive pills, in which IVR users reported equal or higher acceptability with the IVR compared with the oral contraceptive pills.Citation76 Women using a NES/E2 IVR for 13 cycles, using a 21 days in/7 days out regimen, also found this extended use to be acceptable.Citation77

An IVR acceptability study conducted in Africa enrolled 157 HIV-negative women who were asked to use a placebo IVR (56 mm outer diameter, 7.7 mm thick) for 12 weeks. Based on focus group discussions with women and in-depth interviews with a subset of male partners, the IVRs were found to have low expulsion rates (3%), and most women (82%) did not remove the IVR for any time during the 12 weeks of use, with most of the removals occurring for less than a 12-hour period.Citation78 There was high acceptability of the placebo IVR in this patient cohort, with 94% of women saying that they were either interested or very interested in trying the IVR.Citation78 Very few women (2%) reported that they could feel the IVR during daily activities.Citation78 The biggest concern for women trying the placebo IVR was that it would “get lost inside their bodies,”Citation78 suggesting that counseling on product use, risks, and benefits would be important prior to delivery.

The most advanced microbicide IVR to date is the DPV IVR (developed by IPM). Early safety and acceptability studies, in African populations, of this IVR have found no product-related serious adverse events,Citation36 and the adverse events were not higher than those reported by placebo IVR users.Citation54

Side effects associated with IVRs

Breakthrough bleeding (contraceptive component)

Another major determinant of acceptability of an IVR product includes the side effects experienced by the user. For IVRs, systemic side effects are usually minimized, as first-pass metabolism through the liver is avoided.Citation79 However, a major concern in terms of the acceptability of the product, specifically, when a hormonal contraceptive is selected, is the effect on the menstrual cycle, with breakthrough bleeding being a major reason for product dissatisfaction and discontinuation.Citation25,Citation80 Breakthrough bleeding was a leading reason for the Population Council to discontinue development of its NES-only IVR for contraception.Citation30 Of note, ovulation was inhibited in approximately 97%–99% of cycles, using the NES IVR,Citation81 supporting our hypothesis that disruption of ovulation is one factor associated with breakthrough bleeding in progestin-only methods. However, with use of the previous 20 μg/d LNG IVR, during which approximately 40%–60% of women continued to ovulate normally,Citation24,Citation60 bleeding disturbances were less likely – at approximately 17%.Citation80 This hypothesis is further supported by prior studies of LNG and norethisterone IVR users,Citation82Citation84 showing that anovulation was associated with disrupted bleeding patterns. It has previously and recently been recommended that to decrease breakthrough bleeding, one should increase the progestin dose,Citation30,Citation81 but we believe that the opposite is true. By lowering the progestin dose, the main contraceptive action would be local,Citation65 and breakthrough bleeding would be minimized by allowing normal ovulatory cycles and physiologic estrogen and progesterone levels.Citation82Citation84

Vaginal expulsions, disruption of intercourse, vaginal abrasions/ulcers

Early clinical safety testing of microbicide (and MPT) IVRs includes a focus on the effect of the IVR on the subclinical and physical properties of the vaginal mucosa, as well as any systemic effects. One of the most basic safety precautions is that an IVR does not have any adverse physical effects on the vaginal epithelium. The reason for this is that women who are at risk of unintended pregnancies are similarly at risk of STIs, including HIV-1. It is known that ulcerative infections, such as syphilis and HSV-2, increase the efficiency of sexual transmission of HIV-1, likely through frank breeches in the genital epithelium or chronic inflammatory infiltrates after ulcer healing.Citation85Citation89 Any breeches in the protective mucosa caused by IVRs would be a safety concern.

As discussed above, an early LNG IVR prototype developed by the WHO had a cross-sectional diameter of 9.5 mm and released 20 μg/day of LNG for 90 days.Citation25 This was a silastic ring and was firm and relatively inflexible, requiring 6.4 N to compress the ringCitation28 (thus, approximately fourfold more rigid than the NuvaRing). These physical properties resulted in 29% of women experiencing an unintended expulsion of the IVR in the first year of use, mainly with defecation, urination, or menstruation.Citation26 Repeated unintentional expulsion of the IVR resulted in 7.1% of women discontinuing the product in the first year of use.Citation26 This bulky IVR was also electively removed by 12% of women, due to vaginal discharge, irritation, vaginal pain, or dyspareunia.Citation26 The 3.6% pregnancy rate found in the older generation 20 μg/day LNG IVR might have been primarily the result of the high rates of expulsions and removals seen with this less flexible IVR.Citation26 A later trial of 139 women using the early generation silastic IVR found that 35% had erythematous lesions, which were usually asymptomatic.Citation27 Although most of the lesions resolved quickly and spontaneously, the findings prompted considerable concern over safety, and manufacturing of that IVR ceased.Citation27

A redesigned, softer 20 μg/d 90-day LNG IVR (IVR-2), which had a smaller cross-sectional diameter (6 mm) and required only 1.3 N to compress, was later developed and tested for cervicovaginal irritation. This IVR showed no clinically significant changes in the vaginal and cervical mucosa.Citation28

Modern day IVRs have thus evolved to incorporate smaller cross-sectional diameters and more flexible polymers. The progesterone IVR (Progering) has a cross-sectional diameter of 8.4 mm and an outer diameter of 5.5 cm.Citation90 In one study, 6% of the progesterone IVR users reported frequent expulsions, and 8.1% discontinued use due to this reason.Citation91 The NuvaRing has a cross-sectional diameter of 4 mm and an outer diameter of 5.4 cm and requires approximately 0.75 N to compress through 20 mm. In studies, women have reported that this IVR was easy to insert and removeCitation74,Citation92 and did not interfere with intercourse.Citation74 Among the couples using the NuvaRing for contraception, approximately 15% of men reported being infrequently able to feel the IVR during intercourseCitation75 but noted that it was not bothersome and did not interfere with intercourse. The Population Council is currently testing a NES/estradiol IVR for contraception. The colposcopic safety data on the NES/estradiol IVR demonstrate no adverse physical effects on the vaginal epithelium. For example, in a large study of women using IVRs with various combinations of ethinyl estradiol and/or NES or norethindrone acetate, there was a low incidence of subclinical abnormal findings, with colposcopy; all of the subclinical abnormal findings resolved within 1 month, and there were no differences found in the incidence of colposcopic abnormal findings, based on the four types of IVRs evaluated.Citation93 The IVRs used in this study ranged in size from 7.6 × 56 mm to 9.0 × 56 mm.Citation94 The rigidity of these IVRs was expressed as the force, in grams, required to push the sides of the IVR together in the center, and ranged from 270–1000 grams.Citation94 The results of the IVR users in this study were also compared with those of a control group of non-IVR users and to baseline measurements. The research team concluded that IVR use, including long-term IVR use, does increase the incidence of subtle and reversible changes in the vaginal mucosa compared with non-IVR use, but the clinical significance of these findings, specifically in terms of susceptibility to STIs and HIV-1, is not known.Citation93 The most common findings in the study of IVR users were petechiae,Citation93 which are known to occur in healthy women who do not use the IVR.Citation94

While the clinical evaluation is currently pending, the TFV and TFV/LNG IVRs in development by CONRAD are specifically designed to have dimensions and mechanical properties similar to the NuvaRing and to maximize the discreetness of the IVR compared with the bulkier silicone rings.

Emergence of ARV resistance with preventative ARV regimens

There is an ongoing concern that individuals who use either oral or topical PrEP products, once exposed to HIV-1, will be more likely to acquire ARV-resistant strains.Citation95 Studies with microbicide vaginal gels support that ARVs delivered vaginally achieve higher levels in the genital tissue than in serum or peripheral blood mononuclear cells.Citation38,Citation96 HIV prevention trials utilizing vaginal ARV gels have not demonstrated, in the short term, the acquisition of ARV-resistant strains among the active drug users who acquired HIV.Citation4,Citation97 Low systemic levels and high genital compartment concentrations are the PK target for ARV-containing IVRs.Citation19 We believe that locally delivering effective concentrations of ARVs will significantly reduce the systemic side effects while maintaining protection against mucosal HIV-1 acquisition.

Subclinical safety

Alteration of vaginal microflora

Some studies have associated alterations of vaginal flora, specifically, intermediate vaginal flora or bacterial vaginosis, with HIV-1 acquisition.Citation98Citation100 Increases in anaerobic vaginal bacteria have been shown to increase local genital tract inflammation, which is reversed with normalization of the flora.Citation101 There are data to support that the NuvaRing improves the vaginal flora from baseline measurements;Citation102,Citation103 however, the methods used in these studies are less sensitive,Citation102 and there exists a gap in our knowledge of how IVRs alter the vaginal microbiome, as assessed by more detailed methods, such as 16S ribonucleic acid (rRNA) sequencing, which is considered the state of the art in assessing the mucosal microbiome.Citation104 In the bacteria-rich vagina, bio-film forms on the IVR, as would form on any foreign body located in a nonsterile environment. In vitro data support that common vaginal yeast isolates adhere to the NuvaR-ing.Citation105 A small study, using scanning electron microscopy of NuvaRing segments from one woman after 28 days of use, supports that bacteria and mucus do not penetrate or erode the IVR surface and are easily washed away with water.Citation70 A biofilm develops on contraceptive IVR segments after 28 days of use in nonhuman primates.Citation69 Early clinical studies of future microbicide and MPT IVRs will need to include assessments of the changes in the vaginal microbiome with IVR use, to confirm that no adverse changes occur with chronic IVR use.

Effect on vaginal mucosa – local safety endpoints

A major issue in hormonal contraception, which is particularly relevant to MPTs, is the possible link between systemic contraceptive hormones (mainly the intramuscular depot medroxyprogesterone acetate [DMPA]) and the incidence of HIV.Citation106Citation110 One pivotal analysis showed that women in serodiscordant relationships who used DMPA not only had a higher risk of acquiring HIV, but also had an increased risk of transmitting the virus to seronegative male partners.Citation110

Theories as to how exogenous progestins might increase genital tissue susceptibility to HIV-1 infection center on epithelial thinning and the alteration of local mucosal immunity. A decrease in the number of epithelial cell layers or the density of intercellular junction proteins potentially enhances the exposure of cervicovaginal mucosal target cells to HIV-1. In nonhuman primate models of HIV infection, high-dose progestin administration has been found to cause a dramatic atrophy of the vaginal epithelium.Citation111Citation116 However, the data regarding the effect of exogenous progestins on epithelial thickness in the human vagina are mixed, ranging from no change to either increased or decreased thickness,Citation117Citation120 in short-term (3–6 months)Citation117,Citation119 and long-term (2–3 years)Citation118 DMPA users.

Exogenous contraceptive hormones might also affect the mucosal susceptibility to HIV-1 infection, through alterations of the cervicovaginal mucosal immune response. This theory will likely continue to be tested as new contraceptive and MPT IVR products are developed. Although the cervix and vagina are likely the initial sites of entry of HIV-1 in women,Citation121Citation123 the effects of exogenous estrogen and progesterone on the local immune environment of the lower genital tract have not been clearly elucidated,Citation120,Citation124,Citation125 with most data focusing on biologic mechanisms within the endometrium.Citation126 Our group recently published a study showing that DMPA administration resulted in a significant increase in activated lymphocytes (cluster of differentiation [CD]45, CD3, CD8, CD68, human leukocyte antigen [HLA]-DR, and CCR5) in the vaginal tissues compared with baseline samples obtained in the follicular and luteal phases of the menstrual cycle.Citation127 This alteration of local immune response could increase mucosal susceptibility to HIV. Other groups have shown an association between increases in the concentration of systemic (cytokine) and natural killer (NK) cells in the genital compartment and an increased incidence of HIV-1, specifically, in the CAPRISA 004 cohort.Citation128 An increase in genital tract inflammatory mediators has also been linked to higher viral loads and lower systemic CD4 counts in HIV-1 positive women.Citation129

We hypothesize that the microdose concentrations of hormones, particularly of LNG, being proposed in MPTs would not have the same effect on the vaginal mucosa as do the high-dose injectable progestins, like DMPA. In particular, women using DMPA for contraception have serum estradiol levels similar to those of menopausal women (range 15–40 pg/mL)Citation40,Citation130,Citation131 and endogenous progesterone concentrations that are suppressed, at 3–5 nmol/L (1–2 ng/mL), secondary to anovulation.Citation40,Citation132 On the contrary, serum progestin (LNG) levels are lower, at approximately 0.6–2.2 nmol/L (0.187–0.682 ng/mL) in women using the 20 μg/d LNG IVR,Citation60,Citation63,Citation82 while serum estradiol levels remain well above menopausal levels, at 50–110 pg/mL.Citation82 In fact, the systemic levels of LNG found in the previously tested 20 μg/d LNG IVR users were in the same low range as the levels measured among users of the 20 μg/d LNG intrauterine system (IUS) (Mirena) (0.47–1.37 nmol/L [0.147–0.482 ng/mL])Citation61Citation63 and the 14 μg/d LNG IUS (Skyla) (0.06–0.19 ng/mL [0.19–0.61 nmol/L]).Citation64

The data support that the mucosal effects seen with DMPA are more a reflection of the hypoestrogenic milieu seen exclusively in DMPA users. For example, it is known that hypoestrogenic states, such as menopause, pregnancy, and lactation, result in cervical and vaginal atrophy,Citation133,Citation134 an elevated vaginal pH,Citation135 and an altered vaginal microbiome,Citation136 with a preponderance of anaerobic bacteria and a decrease in lactobacilli.Citation137,Citation138 The in vitro data indicate that epithelial tight junction proteins are altered by estrogen levels.Citation139

Based on these theories regarding the effect of exogenous hormones on mucosal susceptibility to HIV, each objective endpoint (eg, vaginal pH, microbiome, vaginal immune cells, or epithelial tight junction proteins) would be important safety endpoints to test in early Phase I studies of MPT IVRs.

Market demand

A good generalization regarding the currently available contraceptive IVRs is that few women use them, but those who do, report high acceptability. Although there has been a steady increase in the use of the NuvaRing since its release in the USA and other European countries in 2002, the current data indicate that approximately only 1.3% of reproductive-age women in the USA report using the NuvaRing as their current contraceptive method.Citation140 Worldwide use of the NuvaRing is low, with large databases not subcategorizing the IVR as a separate category.Citation141 This may be due to cost of the IVR, which ranges from $50–$120 per month without insurance coverage. The US patent for the NuvaRing expires in April of 2018, and this will allow the development of cheaper generic versions and likely an increase in the global market demand. However, an improved introduction of IVRs to regions of the world hit hardest by the HIV pandemic, particularly sub-Saharan Africa, is required for microbicide and MPT IVRs uptake to be successful in these regions.

Many of the contraceptive/microbicide MPTs currently in development utilize LNG as the contraceptive component. LNG has been endorsed by the Initiative for Multipurpose Prevention Technologies Scientific Advisory Working Group as the contraceptive hormone of choice to use in MPTs, as it is well characterized, inexpensive, generic, and has a good safety profile.

Conclusion

IVRs can successfully deliver contraceptive hormones in a highly efficient, discreet, and acceptable manner. Studies to date also show the potential of IVRs to deliver microbicides. Successfully combining the delivery of the two in a single, long-acting IVR has the potential to be a game changer, as this would provide women across the globe with an easy, discreet method of protecting themselves from both pregnancy and HIV or other STIs. With approximately 41% of global pregnancies unplannedCitation13 and 2.5 million new HIV infections per year,Citation1 an MPT could potentially prevent thousands of unintended/mistimed pregnancies and HIV infections, ultimately saving countless women’s lives.

Disclosure

The authors report no conflicts of interest in this work.

References

  • UNAIDSGlobal Report. UNAIDS Report on the Global AIDS EpidemicGenevaUNAIDS2012 Available from: http://www.unaids.org/en/media/unaids/contentassets/documents/epidemiology/2012/gr2012/20121120_UNAIDS_Global_Report_2012_en.pdfAccessed August 24, 2013
  • MarrazzoJRamjeeGNairGPre-exposure prophylaxis for HIV in women: daily oral tenofovir, oral tenofovir/emtricitabine, or vaginal tenofovir gel in the VOICE ttudy (MTN 003)Presented at: the 20th Conference of Retroviruses and Opportunistic Infections (CROI 2013)March 3–6, 2013Atlanta, GA
  • van der LindeIHIV/AIDS in South Africa: At last the glass is half fullPresented at: the 6th SA AIDS ConferenceJune 18–21, 2013Durban, South Africa
  • KarimQAAbdool KarimSSFrohlichJACAPRISA 004 Trial GroupEffectiveness and safety of tenofovir gel, an antiretroviral microbicide, for the prevention of HIV infection in womenScience Express2010329599611681174
  • GrantRMLamaJRAndersonPLiPrEx Study TeamPreexposure chemoprophylaxis for HIV prevention in men who have sex with menN Engl J Med2010363272587259921091279
  • DobardCSharmaSMartinADurable protection from vaginal simian-human immunodeficiency virus infection in macaques by tenofovir gel and its relationship to drug levels in tissueJ Virol201286271872522072766
  • BaetenJMDonnellDNdasePPartners PrEP Study TeamAntiretroviral prophylaxis for HIV prevention in heterosexual men and womenN Engl J Med2012367539941022784037
  • DentonPWOthienoFMartinez-TorresFOne percent tenofovir applied topically to humanized BLT mice and used according to the CAPRISA 004 experimental design demonstrates partial protection from vaginal HIV infection, validating the BLT model for evaluation of new microbicide candidatesJ Virol201185157582759321593172
  • KarimSSKashubaADWernerLKarimQADrug concentrations after topical and oral antiretroviral pre-exposure prophylaxis: implications for HIV prevention in womenLancet2011378978727928121763939
  • DaiJYGilbertPBHughesJPBrownEREstimating the efficacy of preexposure prophylaxis for HIV prevention among participants with a threshold level of drug concentrationAm J Epidemiol2013177325626323302152
  • World Health OrganizationUNICEFUNFPAWorld BankMaternal Mortality in 2005GenevaWorld Health Organization2007 Available from: http://whqlibdoc.who.int/publications/2007/9789241596213_eng.pdfAccessed July 15, 2010
  • SinghSWulfDHussainRBankoleASedghGInstituteGAbortion Worldwide: A Decade of Uneven ProgressNew York, NYGuttmacher Institute2009 Available from: http://www.guttmacher.org/pubs/Abortion-Worldwide.pdfAccessed August 24, 2013
  • SinghSSedghGHussainRUnintended pregnancy: worldwide levels, trends, and outcomesStud Fam Plann201041424125021465725
  • StuartJESecuraGMZhaoQPittmanMEPeipertJFFactors associated with 12-month discontinuation among contraceptive pill, patch, and ring usersObstet Gynecol20131212 Pt 133033623344283
  • FriendDRDoncelGFCombining prevention of HIV-1, other sexually transmitted infections and unintended pregnancies: Development of dual-protection technologiesAntiviral Res201088Suppl 1S47S5421109068
  • World Health OrganizationFigure 3-1 Comparing Typical Effectiveness of Contraceptive MethodsGenevaWorld Health Organization2007 Available from: http://www.contraceptivetechnology.org/table.htmlAccessed August 24, 2013
  • SeidmanDSNon-contraceptive benefits of hormonal contraception: time for renewed awarenessEur J Contracept Reprod Health Care201116640740822066889
  • PadianNSvan der StratenARamjeeGMIRA TeamDiaphragm and lubricant gel for prevention of HIV acquisition in southern African women: a randomised controlled trialLancet2007370958325126117631387
  • JohnsonTJClarkMRAlbrightTHA 90-day tenofovir reservoir intravaginal ring for mucosal HIV prophylaxisAntimicrob Agents Chemother201256126272628323006751
  • MachtDIOn the absorption of drugs and poisons through the vaginaJ Pharmacol Exp Ther1918107509522
  • DziukPJCookBPassage of steroids through silicone rubberEndocrinology19667812082115948426
  • MishellDRTalasMParlowAFMoyerDLContraception by means of a silastic vaginal ring impregnated with medroxyprogesterone acetateAm J Obstet Gynecol197010711001075443056
  • MishellDRLumkinMEContraceptive effect of varying dosages of progestogen in silastic vaginal ringsFertil Steril1970212991035414562
  • World Health OrganizationTask Force on Long-Acting Systemic Agents for Fertility RegulationIntravaginal and intracervical devices for the delivery of fertility regulating agentsJ Steroid Biochem1979111B461467491617
  • KoetsawangSJiGKrishnaUMicrodose intravaginal levonorgestrel contraception: a multicentre clinical trial. I. Contraceptive efficacy and side effects. World Health Organization. Task Force on Long-Acting Systemic Agents for Fertility RegulationContraception19904121051242107054
  • KoetsawangSJiGKrishnaUMicrodose intravaginal levonorgestrel contraception: a multicentre clinical trial. II. Expulsions and removals. World Health Organization. Task Force on Long-Acting Systemic Agents for Fertility RegulationContraception19904121251412107055
  • BoundsWSzarewskiALoweDGuillebaudJPreliminary report of unexpected local reactions to a progestogen-releasing contraceptive vaginal ringEur J Obstet Gynecol Reprod Biol19934821231258491331
  • WeisbergEFraserISBakerJA randomized comparison of the effects on vaginal and cervical epithelium of a placebo vaginal ring with non-use of a ringContraception2000622838911102592
  • MuldersTMDiebenTOBenninkHJOvarian function with a novel combined contraceptive vaginal ringHum Reprod200217102594259912351535
  • BracheVPayánLJFaundesACurrent status of contraceptive vaginal ringsContraception201387326427223040125
  • MalcolmKWoolfsonDRussellJAndrewsCIn vitro release of nonoxynol-9 from silicone matrix intravaginal ringsJ Control Release200391335536412932713
  • Van DammeLRamjeeGAlaryMCOL-1492 Study GroupEffectiveness of COL-1492, a nonoxynol-9 vaginal gel, on HIV-1 transmission in female sex workers: a randomised controlled trialLancet2002360933897197712383665
  • KiserPFJohnsonTJClarkJTState of the art in intravaginal ring technology for topical prophylaxis of HIV infectionAIDS Rev2012141627722297505
  • GuptaKMPearceSMPoursaidAEPolyurethane intravaginal ring for controlled delivery of dapivirine, a nonnucleoside reverse transcriptase inhibitor of HIV-1J Pharm Sci200897104228423918338805
  • MalcolmRKWoolfsonADTonerCFMorrowRJMcCullaghSDLong-term, controlled release of the HIV microbicide TMC120 from silicone elastomer vaginal ringsJ Antimicrob Chemother200556595495616155060
  • NelASmytheSYoungKSafety and pharmacokinetics of dapivirine delivery from matrix and reservoir intravaginal rings to HIV-negative womenJ Acquir Immune Defic Syndr200951441642319623693
  • WoolfsonADMalcolmRKMorrowRJTonerCFMcCullaghSDIntravaginal ring delivery of the reverse transcriptase inhibitor TMC 120 as an HIV microbicideInt J Pharm20063251–2828916884869
  • HendrixCWChenBAGudderaVMTN-001: randomized pharmacokinetic cross-over study comparing tenofovir vaginal gel and oral tablets in vaginal tissue and other compartmentsPLoS ONE201381e5501323383037
  • ThurmanAKimbleTHallPSchwartzJLArcherDFMedroxyprogesterone acetate and estradiol cypionate injectable suspension (Cyclofem) monthly contraceptive injection: steady-state pharmacokineticsContraception201387673874323265980
  • MishellDRPharmacokinetics of depot medroxyprogesterone acetate contraceptionJ Reprod Med199641Suppl 5S381S390
  • van den HeuvelMWvan BragtAJAlnabawyAKKapteinMCComparison of ethinylestradiol pharmacokinetics in three hormonal contraceptive formulations: the vaginal ring, the transdermal patch and an oral contraceptiveContraception200572316817416102549
  • TimmerCJMuldersTMPharmacokinetics of etonogestrel and ethinylestradiol released from a combined contraceptive vaginal ringClin Pharmacokinet200039323324211020137
  • MagnusdóttirEMBjarnadóttirRIOnundarsonPTThe contraceptive vaginal ring (NuvaRing) and hemostasis: a comparative studyContraception200469646146715157790
  • NahoulKDehenninLJondetMRogerMProfiles of plasma estro-gens, progesterone and their metabolites after oral or vaginal administration of estradiol or progesteroneMaturitas19931631852028515718
  • CoutinhoEMCoutinhoEJGonçalvesMTBarbosaICOvulation suppression in women following vaginal administration of oral contraceptive tabletsFertil Steril19823833803816811333
  • KobayashiKMimuraNFujiiHRole of human cytochrome P450 3A4 in metabolism of medroxyprogesterone acetateClin Cancer Res2000683297330310955816
  • TsengAHills-NieminenCDrug interactions between antiretrovirals and hormonal contraceptivesExpert Opin Drug Metab Toxicol20139555957223425052
  • Van DammeLCorneliAAhmedKFEM-PrEP Study GroupPreexposure prophylaxis for HIV infection among African womenN Engl J Med2012367541142222784040
  • RosenbergMJWaughMSLongSUnintended pregnancies and use, misuse and discontinuation of oral contraceptivesJ Reprod Med19954053553607608875
  • BlumenthalPDVoedischAGemzell-DanielssonKStrategies to prevent unintended pregnancy: increasing use of long-acting reversible contraceptionHum Reprod Update201117112113720634208
  • MassaiRQuinterosEReyesMVExtended use of a progesterone-releasing vaginal ring in nursing women: a phase II clinical trialContraception200572535235716246661
  • GilliamMLNeustadtAKozloskiMMistrettaSTilmonSGodfreyEAdherence and acceptability of the contraceptive ring compared with the pill among students: a randomized controlled trialObstet Gynecol2010115350351020177280
  • AlexanderNJBakerEKapteinMKarckUMillerLZampaglioneEWhy consider vaginal drug administration?Fertil Steril200482111215236978
  • RomanoJVarianoBCoplanPSafety and availability of dapivirine (TMC120) delivered from an intravaginal ringAIDS Res Hum Retroviruses200925548348819388819
  • MalcolmRKFetherstonSMMcCoyCFBoydPMajorIVaginal rings for delivery of HIV microbicidesInt J Womens Health2012459560523204872
  • FetherstonSMBoydPMcCoyCFA silicone elastomer vaginal ring for HIV prevention containing two microbicides with different mechanisms of actionEur J Pharm Sci201248340641523266465
  • ClarkJTJohnsonTJClarkMRQuantitative evaluation of a hydrophilic matrix intravaginal ring for the sustained delivery of tenofovirJ Control Release2012163224024822981701
  • SmithJRastogiRTellerRSA tenofovir disoproxil fumarate intravaginal ring completely protects against repeated SHIV vaginal challenge in nonhuman primatesPresented at: the 20th Conference of Retroviruses and Opportunistic Infections (CROI 2013)March 3–6, 2013Atlanta, GA
  • SahotaJBarnesPMMansfieldEBradleyJLKirkmanRJInitial UK experience of the levonorgestrel-releasing contraceptive intravaginal ringAdv Contracept199915431332411145373
  • LandgrenBMJohannissonEMasironiBDiczfalusyEPharmacokinetic and pharmacodynamic investigations with vaginal devices releasing levonorgestrel at a constant, near zero order rateContraception19822665675856820337
  • LockhatFBEmemboluJEKonjeJCSerum and peritoneal fluid levels of levonorgestrel in women with endometriosis who were treated with an intrauterine contraceptive device containing levonorgestrelFertil Steril200583239840415705381
  • SeeberBZiehrSCGschlieβerAQuantitative levonorgestrel plasma level measurements in patients with regular and prolonged use of the levonorgestrel-releasing intrauterine systemContraception201286434534922402256
  • HidalgoMMHidalgo-ReginaCBahamondesMVMonteiroIPettaCABahamondesLSerum levonorgestrel levels and endometrial thickness during extended use of the levonorgestrel-releasing intrauterine systemContraception2009801848919501221
  • Bayer HealthcareHighlights of Prescribing Information [Package Insert for Skyla Intrauterine System]Wayne, NJBayer HealthCare Pharmaceuticals2013 Available from: http://labeling.bayerhealthcare.com/html/products/pi/Skyla_PI.pdfAccessed August 24, 2013
  • NatavioMFTaylorDLewisRATemporal changes in cervical mucus after insertion of the levonorgestrel-releasing intrauterine systemContraception201387442643123121828
  • LähteenmäkiPRauramoIBackmanTThe levonorgestrel intrauterine system in contraceptionSteroids20006510–1169369711108878
  • MunuceMJNascimentoJARosanoGFaundesABahamondesLDoses of levonorgestrel comparable to that delivered by the levonorgestrel-releasing intrauterine system can modify the in vitro expression of zona binding sites of human spermatozoaContraception20067319710116371304
  • MossJAMaloneAMSmithTJSimultaneous delivery of tenofovir and acyclovir via an intravaginal ringAntimicrob Agents Chemother201256287588222123689
  • GunawardanaMMossJASmithTJMicrobial biofilms on the surface of intravaginal rings worn in non-human primatesJ Med Microbiol201160Pt 682883721393449
  • MillerLMacFarlaneSAMateriHLA scanning electron microscopic study of the contraceptive vaginal ringContraception2005711656715639076
  • HazanZZumerisJJacobHEffective prevention of microbial biofilm formation on medical devices by low-energy surface acoustic wavesAntimicrob Agents Chemother200650124144415216940055
  • PruthiVAl-JanabiAPereiraBMCharacterization of biofilm formed on intrauterine devicesIndian J Med Microbiol200321316116517643011
  • CreininMDMeynLABorgattaLMulticenter comparison of the contraceptive ring and patch: a randomized controlled trialObstet Gynecol20081112 Pt 126727718238962
  • DiebenTORoumenFJApterDEfficacy, cycle control, and user acceptability of a novel combined contraceptive vaginal ringObstet Gynecol2002100358559312220783
  • NovákAde la LogeCAbetzLvan der MeulenEAThe combined contraceptive vaginal ring, NuvaRing: an international study of user acceptabilityContraception200367318719412618252
  • OddssonKLeifels-FischerBde MeloNREfficacy and safety of a contraceptive vaginal ring (NuvaRing) compared with a combined oral contraceptive: a 1-year randomized trialContraception200571317618215722066
  • SivinIMishellDRJrAlvarezFContraceptive vaginal rings releasing Nestorone and ethinylestradiol: a 1-year dose-finding trialContraception200571212212915707562
  • van der StratenAMontgomeryETChengHHigh acceptability of a vaginal ring intended as a microbicide delivery method for HIV prevention in African womenAIDS Behav20121671775178622644068
  • Jung-HoffmanCKuhlHPharmacokinetics and pharmacodynamics of oral contraceptive steroids: factors influencing steroid metabolismAm J Obstet Gynecol19901636 Pt 2218321972147819
  • KoetsawangSJiGKrishnaUMicrodose intravaginal levonorgestrel contraception: a multicentre clinical trial. IV. Bleeding patterns. World Health Organization. Task Force on Long-Acting Systemic Agents for Fertility RegulationContraception19904121511672107057
  • BracheVMishellDRLahteenmakiPOvarian function during use of vaginal rings delivering three different doses of NestoroneContraception200163525726111448466
  • XiaoBLZhangXLFengDDPharmacokinetic and pharmacodynamic studies of vaginal rings releasing low-dose levonorgestrelContraception19853254554713936678
  • LandgrenBMJohannissonEMasironiBDiczfalusyEPharmacokinetic and pharmacodynamic effects of small doses of norethisterone released from vaginal rings continuously during 90 daysContraception1979193253271572278
  • JohannissonEBrosensICornillieFMorphometric study of the human endometrium following continuous exposure to levonorgestrel released from vaginal rings during 90 daysContraception19914343613741906792
  • Van de PerrePSegondyMFoulongneVHerpes simplex virus and HIV-1: deciphering viral synergyLancet Infect Dis20088849049718652995
  • GreenblattRMLukehartSAPlummerFAGenital ulceration as a risk factor for human immunodeficiency virus infectionAIDS19882147503128996
  • WasserheitJNEpidemiological synergy. Interrelationships between human immunodeficiency virus infection and other sexually transmitted diseasesSex Transm Dis199219261771595015
  • RoyceRASeñaACatesWCohenMSSexual transmission of HIVN Engl J Med199733615107210789091805
  • ThurmanARDoncelGFHerpes simplex virus and HIV: genital infection synergy and novel approaches to dual preventionInt J STD AIDS201223961361923033511
  • NathASitruk-WareRProgesterone vaginal ring for contraceptive use during lactationContraception201082542843420933116
  • SivinIDíazSCroxattoHBContraceptives for lactating women: a comparative trial of a progesterone-releasing vaginal ring and the copper T 380A IUDContraception19975542252329179454
  • RoumenFJApterDMuldersTMDiebenTOEfficacy, tolerability and acceptability of a novel contraceptive vaginal ring releasing etonogestrel and ethinyl oestradiolHum Reprod200116346947511228213
  • FraserISLacarraMMishellDRVaginal epithelial surface appearances in women using vaginal rings for contraceptionContraception200061213113810802278
  • FraserISLähteenmäkiPElomaaKVariations in vaginal epithelial surface appearance determined by colposcopic inspection in healthy, sexually active womenHum Reprod19991481974197810438412
  • MayerKHVenkateshKKAntiretroviral therapy as HIV prevention: status and prospectsAm J Public Health2010100101867187620724682
  • SchwartzJLRountreeWKashubaADMA multi-compartment, single and multiple dose pharmacokinetic study of the vaginal candidate microbicide 1% tenofovir gelPLoS One2011610e2597422039430
  • Abdool KarimSSCAPRISA 004 two years on: Ten key lessons and their implicationsPresented at: Microbicides 2012April 12, 2012Sydney, Australia
  • MartinHLRichardsonBANyangePMVaginal lactobacilli, microbial flora, and risk of human immunodeficiency virus type 1 and sexually transmitted disease acquisitionJ Infect Dis199918061863186810558942
  • LagaMManokaAKivuvuMNon-ulcerative sexually transmitted diseases as risk factors for HIV-1 transmission in women: results from a cohort studyAIDS199371951028442924
  • MyerLDennyLTelerantRSouzaMDWrightTCJrKuhnLBacterial vaginosis and susceptibility to HIV infection in South African women: a nested case-control studyJ Infect Dis200519281372138016170754
  • ValoreEVWileyDJGanzTReversible deficiency of antimicrobial polypeptides in bacterial vaginosisInfect Immun200674105693570216988245
  • DaviesGCFengLXNewtonJRDiebenTOCoelingh-BenninkHJThe effects of a combined contraceptive vaginal ring releasing ethinyloestradiol and 3-ketodesogestrel on vaginal floraContraception19924555115181623721
  • De SetaFRestainoSDe SantoDEffects of hormonal contraception on vaginal floraContraception201286552652922520642
  • RavelJGajerPAbdoZVaginal microbiome of reproductive-age womenProc Natl Acad Sci U S A2011108Suppl 1S4680S4687
  • CamachoDPConsolaroMEPatussiEVDonattiLGasparettoASvidzinskiTIVaginal yeast adherence to the combined contraceptive vaginal ring (CCVR)Contraception200776643944318061701
  • UngchusakKRehleTThammapornpilapPSpiegelmanDBrinkmannUSiraprapasiriTDeterminants of HIV infection among female commercial sex workers in northeastern Thailand: results from a longitudinal studyJ Acquir Immune Defic Syndr Hum Retrovirol19961255005078757428
  • MartinHLJrNyangePMRichardsonBAHormonal contraception, sexually transmitted diseases, and risk of heterosexual transmission of human immunodeficiency virus type 1J Infect Dis19981784105310599806034
  • BaetenJMBenkiSChohanVHormonal contraceptive use, herpes simplex virus infection, and risk of HIV-1 acquisition among Kenyan womenAIDS200721131771177717690576
  • MorrisonCSChenPLKwokCHormonal contraception and HIV acquisition: reanalysis using marginal structural modelingAIDS201024111778178120588106
  • HeffronRDonnellDReesHPartners in Prevention HSV/HIV Transmission Study TeamUse of hormonal contraceptives and risk of HIV-1 transmission: a prospective cohort studyLancet Infect Dis2012121192621975269
  • MarxPASpiraAIGettieAProgesterone implants enhance SIV vaginal transmission and early virus loadNat Med1996210108410898837605
  • SmithSMBaskinGBMarxPAEstrogen protects against vaginal transmission of simian immunodeficiency virusJ Infect Dis2000182370871510950763
  • JiangYTianBAgyMBSaifuddinMTsaiCCMacaca fascicularis are highly susceptible to an RT-SHIV following intravaginal inoculation: a new model for microbicide evaluationJ Med Primatol200938Suppl 1S39S46
  • PalRNuttallJGalminLWeissDChungHKRomanoJCharacterization of vaginal transmission of a simian human immunodeficiency virus (SHIV) encoding the reverse transcriptase gene from HIV-1 in Chinese rhesus macaquesVirology2009386110210819195672
  • SalléBBrochardPBourryOInfection of macaques after vaginal exposure to cell-associated simian immunodeficiency virusJ Infect Dis2010202333734420569157
  • VeazeyRSShattockRJPopeMPrevention of virus transmission to macaque monkeys by a vaginally applied monoclonal antibody to HIV-1 gp120Nat Med20039334334612579198
  • MillerLPattonDLMeierAThwinSSHootonTMEschenbachDADepomedroxyprogesterone-induced hypoestrogenism and changes in vaginal flora and epitheliumObstet Gynecol200096343143910960638
  • BahamondesLTrevisanMAndradeLThe effect upon the human vaginal histology of the long-term use of the injectable contraceptive Depo-ProveraContraception2000621232711024225
  • MauckCKCallahanMMBakerJThe effect of one injection of Depo-Provera on the human vaginal epithelium and cervical ectopyContraception1999601152410549448
  • IldgrubenAKSjöbergIMHammarströmMLInfluence of hormonal contraceptives on the immune cells and thickness of human vaginal epitheliumObstet Gynecol2003102357158212962945
  • HaaseATEarly events in sexual transmission of HIV and SIV and opportunities for interventionsAnnu Rev Med20116212713921054171
  • PudneyJQuayleAJAndersonDJImmunological microenvironments in the human vagina and cervix: mediators of cellular immunity are concentrated in the cervical transformation zoneBiol Reprod20057361253126316093359
  • HladikFSakchalathornPBallweberLInitial events in establishing vaginal entry and infection by human immunodeficiency virus type-1Immunity200726225727017306567
  • PattonDLThwinSSMeierAHootonTMStapletonAEEschenbachDAEpithelial cell layer thickness and immune cell populations in the normal human vagina at different stages of the menstrual cycleAm J Obstet Gynecol2000183496797311035348
  • WieserFHosmannJTschugguelWCzerwenkaKSedivyRHuberJCProgesterone increases the number of Langerhans cells in human vaginal epitheliumFertil Steril20017561234123511384659
  • WiraCRFaheyJVGhoshMPatelMVHickeyDKOchielDOSex hormone regulation of innate immunity in the female reproductive tract: the role of epithelial cells in balancing reproductive potential with protection against sexually transmitted pathogensAm J Reprod Immunol201063654456520367623
  • ChandraNThurmanARAndersonSDepot medroxyprogesterone acetate increases immune cell numbers and activation markers in human vaginal mucosal tissuesAIDS Res Hum Retroviruses201329359260123189932
  • NaranbhaiVAbdool KarimSSAltfeldMCAPRISA004 TRAPS teamInnate immune activation enhances hiv acquisition in women, diminishing the effectiveness of tenofovir microbicide gelJ Infect Dis20122067993100122829639
  • RobertsLPassmoreJAMlisanaKGenital tract inflammation during early HIV-1 infection predicts higher plasma viral load set point in womenJ Infect Dis2012205219420322190580
  • ClarkMKSowersMLevyBTTenhundfeldPMagnitude and variability of sequential estradiol and progesterone concentrations in women using depot medroxyprogesterone acetate for contraceptionFertil Steril200175587187711334896
  • FotherbyKSaxenaBNShrimankerKA preliminary pharmacokinetic and pharmacodynamic evaluation of depot-medroxyprogesterone acetate and norethisterone oenanthateFertil Steril19803421311397409232
  • OrtizAHirolMStanczykFZGoebelsmannUMishellDRSerum medroxyprogesterone acetate (MPA) concentrations and ovarian function following intramuscular injection of depo-MPAJ Clin Endocrinol Metab19774413238833262
  • WisniewskiPMWilkinsonEJPostpartum vaginal atrophyAm J Obstet Gynecol19911654 Pt 2124912541659199
  • NilssonKRisbergBHeimerGThe vaginal epithelium in the postmenopause – cytology, histology and pH as methods of assessmentMaturitas199521151567731384
  • RoySCaillouetteJCRoyTFadenJSVaginal pH is similar to follicle-stimulating hormone for menopause diagnosisAm J Obstet Gynecol200419051272127715167829
  • PetricevicLDomigKJNierscherFJCharacterisation of the oral, vaginal and rectal Lactobacillus flora in healthy pregnant and postmenopausal womenEur J Obstet Gynecol Reprod Biol20121601939922088236
  • PabichWLFihnSDStammWEScholesDBoykoEJGuptaKPrevalence and determinants of vaginal flora alterations in postmenopausal womenJ Infect Dis200318871054105814513427
  • GondoFda SilvaMGPolettiniJVaginal flora alterations and clinical symptoms in low-risk pregnant womenGynecol Obstet Invest201171315816221160139
  • GorodeskiGIEstrogen modulation of epithelial permeability in cervical-vaginal cells of premenopausal and postmenopausal womenMenopause20071461012101917572644
  • JonesJMosherWDanielsKDivision of vital statisticsCurrent contraceptive use in the United States, 2006–2010 and changes in patterns of use since 1995Natl Health Stat Report201260126 Available from: http://www.cdc.gov/nchs/data/nhsr/nhsr060.pdfAccessed August 24, 2013
  • un.org [homepage on the Internet]World contraceptive use 2010United Nations, Department of Economic and Social Affairs, Population Division2010 [updated February 2011; cited July 15, 2011]. Available from: http://www.un.org/esa/population/publications/wcu2010/Data/UNPD_WCU_2010_Contraceptive_prevalence_method.xlsAccessed August 24, 2013