Abstract
Despite the identification of HIV-1 as the etiological agent responsible for AIDS nearly 30 years ago, a sterilizing vaccine capable of preventing transmission of the virus remains elusive. In response to struggles on the vaccine development front, significant effort has been devoted to preventing the transmission of HIV with alternative products, technologies, and strategies. One of the early alternative HIV prevention strategies was microbicides, which are topical products that can be used to prevent sexual transmission of HIV either vaginally or rectally. First generation microbicide products were designed to be simple gel formulations comprised of readily available active agents that were inexpensive and broadly active (i.e., non-specific). Unfortunately, despite the clinical investigation of multiple product concepts satisfying these requirements, none were shown to be efficacious in pivotal trials. More recently, microbicide and oral prevention strategies involving highly specific and potent anti-retroviral (ARV) drugs have shown to be efficacious in trials. Although building on these successes continues, these products have a number of issues including potential toxicity with long term use, selection of HIV resistance, and cost. Further, all of the original justifications for non-specific microbicide products remain valid. This review provides a brief history of non-specific microbicide development, outlines the evolution to, and limitations of, ARV based microbicides, and summarizes the current activity on non-specific microbicide product development.
Keywords: Microbicides, HIV, prevention, product development
INTRODUCTION
The need for HIV prevention strategies dates back to the earliest days of the epidemic. Identification of HIV as the pathogenic cause of AIDS [1,2] spawned hopes for a speedy path to a preventive vaccine. Nearly 30 years later, despite some recent positive results in vaccine development [3], a highly efficacious vaccine for HIV remains elusive. Over that time period, highly effective treatment strategies have emerged that can largely control HIV infection and have greatly enhanced survivability and quality of life for HIV infected patients [4]. However, these treatment strategies can be costly and have been challenging to fund and apply broadly in low resource countries. With neither a highly potent vaccine or effective treatment readily available in resource poor settings, the epidemic has progressed to a devastating level in parts of the world least equipped to address the situation. Today more than 30 million people are living with HIV, and there were more than 2.6 million new infections in 2009, most of which occur in sub-Saharan Africa [5]. Further, the incidence of new infections in women continues to be a major health problem in these settings, and is likely based in both biological and socioeconomic factors.
Nearly 20 years ago, the notion of an alternative prevention strategy based on the use of vaginally dosed products to inhibit HIV infection in women was formalized [6,7,8]. The concept of a “microbicide” product was born out of the unavailability of a vaccine and the limited capacity for women to negotiate the use of condoms by their male partners, which compounds their apparently elevated biological susceptibility to sexual transmission of HIV. Women in resource poor parts of the world desperately needed a means of controlling their risk of infection and to be empowered to make their own decisions regarding their sexual and reproductive health. Consequently, to best serve this population, microbicide products would need to be safe and effective at preventing sexual transmission of HIV, acceptable to the at risk populations, inexpensive and easily distributed, and women initiated. Further, in view of the rapidly expanding nature of the epidemic (particularly in women) and slow progress on vaccine development, microbicide products needed to be available as soon as possible. In the earliest days of microbicide development options that satisfied the microbicide target product profile were extremely limited. Anti-retroviral (ARV) agents used in treatment were expensive, and generally, their developer pharmaceutical companies had limited interest in pursuing their use for prevention. Therefore, the earliest microbicide product development efforts attempted to take advantage of inexpensive and available agents that could be readily formulated for vaginal (and eventually rectal) use, and had the additional potential to serve as inhibitors of other sexually transmitted infections (STI) and as contraceptives. The prevention of other STI and pregnancy remain important to the base goal of HIV prevention, given that the former are co-factors increasing susceptibility to HIV infection [e.g., 9], and that contraceptive efficacy provides a non-stigmatized additional motivation for microbicide uptake and use, which does not require women to self-identify as “high risk”. This review summarizes the early attempts to develop such microbicide products and how those first generation product development efforts led to the current state of the microbicide field. Table 1 provides a summary of the early generation, non-specific microbicide candidate products.
Table 1.
Product | Agent Class | Mechanism of Action |
---|---|---|
Nonoxynol-9 | Detergent | Virucide/Disrupts viral lipid envelope |
C31G (Savvy) | Detergent | Virucide/Disrupts viral lipid envelope |
Carraguard | Polyanion | Entry Inhibitor/Blocks virus-target cell interaction |
Cellulose Sulfate | Polyanion | Entry Inhibitor/Blocks virus-target cell interaction |
PRO 2000 | Polyanion | Entry Inhibitor/Blocks virus-target cell interaction |
BufferGel | Acidifier | Virucide/Presumed to denature viral proteins |
THE USE OF SURFACE ACTIVE AGENTS
Nonoxynol 9 (N-9) is a non-ionic surfactant that has been used as an over-the-counter (OTC) spermicide to prevent pregnancy for decades. The spermicidal effect of N-9 is achieved via solubilization of the sperm cell membrane [10]. A number of contraceptive vaginal formulations of N-9 (2–12%) are commercially available. In addition to its spermicidal activity, a number of in vitro studies have demonstrated that not only could N-9 inhibit HIV [11,12], but it was also effective against several other STIs [13,14] including Herpes Simplex Virus (HSV) [15]. Further, an animal model had been used to demonstrate that N-9 could inhibit transmission of SIV in vivo [16]. The in vitro and in vivo anti-microbial and anti-viral activity associated with N-9, along with the fact that vaginal formulations for contraception were available as OTC products suggested that evaluation of N-9 containing formulations for the prevention of HIV transmission in the clinic was reasonable. This was supported by other safety data for N-9 available at the time [17].
Vaginal gel formulations with N-9 were evaluated in a number of safety, acceptability, and efficacy studies as a potential microbicide product for the prevention of HIV transmission and STI [18–22]. Perhaps the most relevant of these trials was the evaluation of the COL-1492 formulation of N-9 (3.5%, 52.5 mg dose) in female sex workers [22]. This study was conducted in four countries (three in sub-Saharan Africa and Thailand) with 765 women randomized 1:1 with a placebo gel control. HIV incidence in the N-9 group was 16% versus 12% in the placebo group (hazard ratio adjusted for center 1.5; 95% CI 1.0–3.2; p=0.047). Thirty-two percent of the women reported mean use of more than 3.5 applicators per day (instructions were to use within one hour prior to sex), and the HIV infection rate in this group was nearly twice that in placebo users. Thus, not only did this N-9 product fail to show efficacy against HIV transmission in this high risk population, intensive use of the product was likely increasing the risk of HIV infection. Although previous studies had shown this formulation to be both safe [19] and acceptable [18] in similar populations of women, the results from the efficacy study clearly demonstrated that N-9 was not a viable candidate for microbicide development. In rectal safety studies, N-9 formulations were shown to cause severe epithelial damage, indicating this compound was not appropriate for rectal use [23].
Another surface active drug, C31G, was also developed as a vaginal microbicide and contraceptive product. Savvy® is a vaginal gel formulated with 1% C31G, an amphoteric surfactant that functioned similar to N-9 in terms of cellular membrane disruption. C31G was shown to be a potent inhibitor of HIV in vitro [24] and had an acceptable cytotoxicity profile [25]. Like N-9, C31G was also inexpensive, potentially broad spectrum [26], and contraceptive [27]. Therefore, a series of phase 1 safety studies were conducted with the final vaginal gel formulation of C31G, including male and female tolerance and acceptability trials [28,29]; a post-coital testing study [30], and a daily use dose escalation study [31]. These studies demonstrated that C31G gel formulation was generally safe and well tolerated. Consequently, the Savvy gel was progressed into two HIV prevention phase 3 efficacy studies (one in Ghana [32] and one in Nigeria [33]), and a third contraceptive efficacy trial [34]. In the Ghana study, a total of 17 seroconversions occurred; eight in the Savvy group and nine in the placebo group. In the Nigeria study, there were 21 seroconversions in the Savvy group and 12 in the placebo group. The number of adverse events (AE), including reproductive tract and pelvic findings were similar in the two groups in the Nigeria study, however more reproductive tract AE were observed in the Savvy arm versus placebo in the Ghana trial (13.0% versus 9.4%, respectively). Both of these studies failed to reach the required number of HIV endpoints to achieve power to determine an effect by Savvy, and both studies were consequently stopped early for futility.
With multiple phase 3 trials showing lack of effectiveness and possible harm, and a series of in vitro and animal studies linking surfactants to inflammatory changes in the cervicovaginal mucosa [35–39], the microbicide field was ready to move away from surfactant based product options.
DEVELOPMENT AND CLINICAL EVALUATION OF BUFFERING AGENTS
Microbicides based on acid buffering were developed in light of reports based on experiments indicating that HIV-1 was inactivated at acid pH [40,41], and that lymphocytes and macrophages, potential vectors for entry of HIV [42–46], can be immobilized and killed [46] by acidity. In addition, HSV-2, certain bacterial STI pathogens, and sperm can also be inhibited at acid pH and their transmission inhibited by acid buffering microbicides [47–51]. Further supporting the potential utility of vaginal acidification are the consistent epidemiological observations that women with lactobacillus-dominated flora and vaginal acidity are at reduced risk of HIV, HSV, and other STIs compared to women with bacterial vaginosis (BV), a condition characterized by the loss of lactobacilli and vaginal acidity [52–56].
BufferGel® [51], Amphora™ (formerly Acidform™) [57], and lime juice are acid buffering agents that have been studied clinically. Development of lime juice as a microbicide has been abandoned due to cervicovaginal epithelial toxicity [58, 59]. Amphora has shown protection against vaginal transmission of Neisseria gonorrhoeae [49] and HSV [50] in animal models, and has been evaluated in human safety studies [60,61]. Amphora is being studied in a Phase 3 contraceptive trial (ClinicalTrials.gov Identifier NCT0130633), but has not entered HIV/STI efficacy studies.
BufferGel is the most extensively studied of the acid buffering products. BufferGel was protective against Chlamydia trachomatis, HSV-2, and cottontail rabbit papilloma virus in mouse and rabbit models [47,48], and was contraceptive in the rabbit [47]. In the pigtailed macaque vaginal model, BufferGel had a good safety profile, but did not protect against chlamydia challenge [62]. It was effective in blocking vaginal infection with an HIV-infected-cell challenge in the huSCID mouse model [46] but did not protect macaques from cell-free SIV challenge (CC Tsai, University of Washington unpublished).
In human Phase 1 safety trials, BufferGel was safe and acceptable [63,64]. Moreover, BufferGel significantly reduced BV prevalence in women with BV at enrollment [64]. BufferGel was chosen for inclusion in a Phase 2b HIV effectiveness trial (HPTN 035) due to its theoretical safety (reinforcing a natural vaginal protective mechanism), its observed safety in animal models [62] and Phase 1 trials [63,64], and its activity in some vaginal challenge models. It was also chosen for inclusion in the trial because its mechanism of action was distinct from the second agent studied in the trial, the polyanion PRO 2000. Finally, BufferGel’s contraceptive potential (later confirmed in a Phase 3 efficacy trial [65]) was considered a potential benefit in providing multipurpose protection, and providing a non-stigmatized motivation for product use.
However, BufferGel was ineffective in reducing the incidence of HIV and other STIs in the HPTN 035 trial [66]. What factors might explain it’s failure? Recent studies of non-clade B HIV-1 have been reported showing lower sensitivity to acid inactivation [67] than those of clade B virus [40,41]. BufferGel has a relatively short duration of action [47], thus likely requiring close adherence to precoital dosing timing, and thus increasing the difficulty of adherence. Duration may be particularly limited in women lacking an intrinsically acidic vaginal environment, since here BufferGel’s acidity would more rapidly decline. And, unlike the outcome during twice-daily BufferGel dosing in Phase 1 where BV prevalence was substantially reduced [64], BufferGel did not reduce BV prevalence in HPTN 035, where dose frequency was <3/week. Finally, BufferGel effectiveness is lost after modest dilution [51]. Challenge studies with a variety of microbicide candidates indicate that in vivo efficacy often require microbicide concentrations two to three orders of magnitude higher than those effective in vitro [68,17]. These factors, individually or in combination likely are responsible for the lack of BufferGel effectiveness against HIV/STI in HPTN 035.
THE USE OF POLYANIONS FOR THE PREVENTION OF HIV INFECTION
Early in the HIV epidemic, polyanions, electronegatively charged polymers, were shown to inhibit HIV-1 replication, primarily by preventing viral entry through interference with gp120-cell receptor interactions [69–71]. In vitro and animal studies proved that, in general, these compounds were safe, particularly for topical use [72,73]. With varying degrees of activity, most of them also inhibited other sexually transmitted viruses such as HSV-2 and HPV [74,75]. Higher potency against HIV-1 and HSV-2, better safety profiles, ease of manufacturing in gels and lack of restrictive patents made polyanions reasonable candidates to follow surfactants in the quest for a successful microbicide. Among all the compounds with demonstrated anti-HIV activity, three were fully developed and entered clinical effectiveness trials. These were cellulose sulfate (UsherCell), carrageenan (Carraguard), and a sulfonated naphthalene derivative (PRO 2000).
Cellulose sulfate (CS) was developed by Rush University in collaboration with CONRAD and Polydex, Inc. CS is a sulfate ester of β1,4-glucose polymer with an average molecular weight around 2x106 Daltons, which displays inhibitory activity against HIV-1/2, HSV-2, HPV, N. gonorrhoeae, C. trachomatis, and other STI-causing pathogens [49, 75–77]. Numerous phase 1/2 clinical studies support CS cervicovaginal and penile safety [78–80]. CS is the only polyanion with clinically proven contraceptive efficacy [81]. Two clinical HIV effectiveness trials were run concomitantly, sponsored by CONRAD and FHI360, respectively [82,83]. The final, intent-to-treat analysis of the results of the CONRAD trial, which was stopped early due to the independent data monitoring committee’s recommendation, revealed 25 and 16 new infections in the CS and placebo arms, respectively, with a relative risk (RR) of 1.61 (0.86–3.01), which was not statistically significant (p=0.1). Acting on the possibility of harm, the FHI360 trial was also stopped early, showing 10 and 13 new seroconversions in the CS and placebo arms, respectively, with a RR=0.8 (0.3–1.9; p=0.56).
Carraguard, developed by the Population Council, is a mixture of lambda and kappa carrageenans, which are seaweed-derived compounds with demonstrated antiviral and antibacterial activity and a track-record of safety as food additives [49, 72, 84–87]. Recent macaque studies of Carraguard have shown statistically significant partial protection against RT-SHIV [88]. These results seem to contradict in vitro data suggesting weak potency against R5 viruses and a possible enhancement of infection of RT-SHIV at low concentrations [88,89]. A randomized, placebo-controlled trial of Carraguard in South Africa revealed similar HIV incidence in both Carraguard and placebo groups with a hazard ratio of 0.87 (0.69–1.09) and no significant difference in the time to seroconversion (p=0.30) [90]. Gel use based on applicator testing was about 42% on average, suggesting poor protocol adherence.
PRO 2000, developed successively by Procept, Indevus and Endo Pharmaceutical Solutions, is a synthetic naphthalene sulfonate derivative (~5 kDa) displaying antiviral and antibacterial activities [49,91,92]. Macaque studies revealed partial protection against X4 and R5 SHIV [93,94]. Phase 1/2 clinical studies demonstrated safety for PRO 2000 0.5 and 2.0% gels [95–97]. Two randomized, placebo-controlled studies were performed to test the effectiveness and safety of PRO 2000 gel against HIV-1 [98,99]. The HPTN-035 (phase 2b) compared PRO 2000 0.5% gel and BufferGel against placebo and no gel (condom-only), while the MDP-301 compared PRO 2000 0.5 and 2.0% gels with placebo gel. Although the HIV incidence was lower for PRO 2000 0.5% in the HPTN trial (HR=0.7; p=0.1), the more powerful phase 3 MDP trial did not show any statistically significant difference, demonstrating no protection against HIV-1 acquisition.
Other polyanionic compounds were developed as microbicides but never made it past preclinical stages or phase 1 clinical trials [100]. Notwithstanding their differences, all three polyanions tested in effectiveness trials failed to protect women against HIV-1. Reasons for this failure may be found in their low potency (in comparison to antiretrovirals), negligible absorption with the consequent need to act at the surface of the mucosa, reduction of activity in seminal plasma, short duration of effect, and possible induction of mucosal and microflora changes facilitating HIV-1 initial infection.
THE EVOLUTION TO ARV BASED MICROBICIDES
The early generation non-specific microbicides were a diverse collection of compounds and formulations that shared some fundamental attributes as microbicide candidate products. They were all inexpensive, readily available, potentially broad spectrum, and were shown to be safe and acceptable in early stage trials. However, low potency, short duration of action, challenges to consistent adherence to product use, and in some cases product alteration of epithelial barrier function all contributed to the conclusion that this set of early microbicide compounds was not viable for continued development. Efforts with these early compounds led to initiation of development of ARV based microbicide products, which were far more potent and specific, and likely to be safe based on extensive clinical experience with systemic use. Fortunately, there have been a number of successes recently reported with ARV based prevention strategies [e.g., 101,102], including the results in the CAPRISA 004 trial with tenofovir gel, which showed a statistically significant protective effect [103]. However ARV based prevention products are not without their own issues. For example, there are concerns regarding the emergence of resistance associated with the use of such products, especially for those already in use in front line HIV therapies. This concern will likely dictate the need for regular testing for HIV status in women using ARVs. Thus, a higher level of health care management will be associated with their use and there is likely a higher hurdle to reach OTC status with these products. There are also concerns regarding toxicity with long term use of these compounds used orally, or after absorption following topical use, consistent with what has been seen in therapy, but less acceptable in this new role of prophylaxis for healthy individuals. These compounds are more expensive than the early generation non-specific compounds, and the individual physical-chemical properties of ARV can limit the available formulation options. Thus, the original justifications for the non-specific microbicide products remain valid today, even in the context of emerging successful ARV based prevention options.
CURRENT STATE OF THE “NON-SPECIFIC” MICROBICIDE PIPELINE
There remains a critical need to identify broad-acting microbicide formulations that (i) are potent against HIV, as well as other STIs, (ii) are safe when used repeatedly and/or intensively (as will occur in real life), (iii) have a long duration of activity to help circumvent adherence issues and provide users with a more flexible dosing regimen, (iv) limit the development and/or transmission of drug-resistant HIV, and (v) are effective both vaginally and rectally to provide greater protection for women and men.
A partial summary of the current non-specific product pipeline is provided in Table 2. This includes formulations that are in clinical or preclinical testing, as well as novel agents that may represent the next APIs advancing for formulation and more extended testing.
Table 2.
Product | Mechanism of Action/Targets | Stage of Development | References |
---|---|---|---|
VivaGel (SPL7013 dendrimer) | Attachment/Entry Inhibitor; HIV/HSV | Clinical | [104–110] |
Praneen (Polyherbal Tablet) | HIV/Pregnancy; Possibly HSV | Clinical | [111–116] |
Zinc Acetate (±MIV-150) in Carrageenan Gel | Antiviral/Virucidal/Immunomodulatory; HIV/HSV | Pre-Clinical | [138] |
Aptamers | Attachment/Entry Inhibitor; HIV/HSV | Pre-Clinical | [149,150] |
Aptamers-siRNA Hybrid | CCR5 knockdown; HIV/HSV | Pre-Clinical | [151] |
Sydencan-Fc Hybrid | Attachment/Entry Inhibitor; HIV/HSV | In vitro | [152] |
Monoclonal Antibodies | Broadly Neutralizing; HIV/HSV | Pre-Clinical | [153–155] |
Clinical Testing
VivaGel® is the SLP7013 dendrimer-containing gel (3% or 30mg/ml SLP7013) developed by Starpharma, which has activity against HIV and HSV-2 (in vitro) [104,105]). Macaque testing demonstrated that the 3% gel protected 5 of 6 macaques (16.7% infection) from vaginal infection when applied 20 min prior to challenge with SHIV89.6P (compared to 100% infection in the controls and 66.7% infection in animals treated with the 1% gel) [106]. Initial clinical testing of VivaGel following penile [107] or vaginal [108] application once a day for 7d suggested that the gel was safe. However, more recent Phase 1 studies of twice-daily vaginal application for 14d revealed evidence of mild irritation after this more extensive repeated vaginal use [109,110]. Notably, Phase 2 testing demonstrated VivaGel is effective in the treatment of bacterial vaginosis (BV), which has been linked to HIV [52]. Phase 3 testing of VivaGel’s efficacy against BV is being planned.
Praneem is a polyherbal tablet being developed in India as a multipurpose prevention technology for the prevention of HIV and pregnancy. Initial in vitro studies also suggest that it may have activity against HSV [111]. Phase 1 and 2 testing of Praneem suggests that it is safe, acceptable for use [112–115], and efficacious in relieving abnormal vaginal discharge (caused by various STIs) [116]. However, two of the Phase 2 studies reported that participants experienced transient genital discomfort (though there were no discontinuations of product use) [114], and revealed a concern with adherence (and, therefore, ultimately efficacy) [115].
Preclinical Testing of Established Formulations
The next most advanced products are the Population Council’s zinc acetate-containing gels, which are in the final stages of preclinical testing, with Phase 1 testing slated to commence in 2012. Although hampered by zinc’s toxic effects in cell assays, in vitro studies to date suggest it has anti-viral and virucidal activity against HIV and other viruses, including HSV-2 [117–125] (and MR, unpublished). It is likely that the immunomodulatory properties of zinc [126–127] also contribute to limiting HIV spread, by rendering the mucosal milieu more resistant to infection and/or impeding virus-driven responses that are critical to the onset of HIV infection [128]. As a result, zinc is expected to provide protection against wild type and drug-resistant HIV, underscoring its potential as a broad-acting API, also potentiating the potent activity of MIV-150 (a non-nucleotide reverse transcriptase inhibitor) when used together. Moreover, zinc has been reported to have activity against other STI pathogens such as T. vaginalis [129,130].
Carrageenan was chosen as a safe and acceptable vehicle [90,131–136], with excellent properties to deliver APIs [137]. This formulation was found to be safe and provided mice significant protection against high-dose HSV-2 challenge via the vaginal and rectal routes, where carrageenan is ineffective; (Fernández-Romero et al., 2011, in press). Zinc in solution or formulated in HEC was also ineffective. Macaque studies also revealed that zinc acetate-containing gels are extremely effective at preventing immunodeficiency virus infection (Table 3) [138]. Repeated application of 14 mM (0.3% or 3mg/ml) zinc acetate dihydrate in carrageenan which is effective against HSV-2 in mice, was shown to be safe and provide significant (70%) protection against vaginal SHIV-RT infection. Protection is further enhanced when zinc acetate is combined with only 50 μM MIV-150 (0.00185% or 18.5μg/ml; ~90% protection, compared to the carrageenan vehicle controls). Notably, the protection afforded by these zinc acetate-containing gels lasts for at least 24 h, unlike other preparations tested in macaques that often only provide partial protection for minutes to a few hours, even when using considerably higher doses of drugs [106,139–145].
There is emerging evidence that carrageenan can reduce HPV infection in mice [87,146] and reduces the incidence of HPV infection in women [147]. A Phase 2b study being carried out by researchers at the Albert Einstein School of Medicine in collaboration with the Population Council should provide the first insight as to whether carrageenan is effective against HPV in humans. While clinical testing is necessary to verify the safety and activity of zinc-containing gels, they represent an exciting new generation formulation that could be used repeatedly by women (and potentially men) independent of the time of intercourse (if desired) to protect them against HIV, HSV-2, and possibly other STIs. While multicomponent formulations pose regulatory challenges, both carrageenan and zinc acetate are generally recognized as safe (GRAS), MIV-150 has completed all preclinical toxicology testing, and the final preclinical testing of the zinc-containing formulations is underway – thus positioning these gels to advance for clinical testing.
To provide an alternative delivery vehicle for the promising zinc acetate/MIV-150 API combination, a vaginal ring loaded with these two API is currently being developed by Population Council researchers. Proof-of-concept macaque studies have confirmed that MIV-150 delivered from a vaginal ring is effective at preventing SHIV-RT infection (Singer et al., in preparation). Preclinical formulation research is also underway to explore the potential of combining the zinc acetate-containing formulations with a contraceptive levonorgestrel (LNG; in gels and/or vaginal rings), similar to the strategies being pursued elsewhere in the field [148].
Next Generation Non-Specific Drugs in Preclinical Testing
Although in much earlier stages of investigation, several promising non-specific API candidates are emerging in the field (most recent references relevant to microbicides research are cited in Table 2). These include a variety of agents derived from natural sources (e.g. praneem, carragaurd combinations), novel synthesized agents, and neutralizing monoclonal antibodies expressed in plants, and used in combinations for expanded spectrum against HIV-1, HSV, and potentially other pathogens). All have activity against HIV and some also against HSV. To date, all of these have demonstrated potent anti-HIV activity in vitro and some have even been shown to have activity in vivo (macaques or mice), thereby providing the proof-of-concept for advancing their development. However, most of these products have not yet been formulated and need further testing as formulated products (stability, efficacy, safety, etc). It will be critical to determine which will advance further as a promising formulation.
SUMMARY
Despite recent success with highly specific ARV based HIV prevention strategies and products, the original drivers for non-specific microbicide product development remain valid. Products which have broad spectrum activity, are less toxic, have reduced risks associated with the emergence of resistance, and are inexpensive remain important to the HIV prevention field. Although there is significant focus and investment today in ARV based HIV prevention products, there remain a number of non-specific product candidates in development. Hopefully, the development of these new non-specific products can be empowered by the lessons learned from earlier generation non-specific products, and the successes of recent generation ARV based products.
Table 3.
Gel | Time of Virus Challenge After the Last Gel (Infected/Challenged) | Total (Infected/Challenged) | Protection vs Carrageenan | P Value vs Carrageenan | ||
---|---|---|---|---|---|---|
4h | 8h | 24h | ||||
MIV-150/ZA | 0/7 | 0/7 | 2/14* | 2/28 | 89% | <0.0002 |
ZA | ND | 1/7 | 3/14* | 4/21 | 70% | <0.012 |
MIV-150 | ND | 2/7 | 4/7 | 6/14 | 33% | <0.5 |
Carrageenan | 9/14^ |
Data from animals receiving a daily dosing of gel for 2 weeks and dosing every other day for 4 weeks has been pooled.
Data from real time controls for each challenge are pooled with data from historical controls.
Depo-Provera-treated Chinese rhesus macaques were challenged with 103 TCID50 SHIV-RT at the indicated times relative to repeated gel application. This virus dose routinely infects 60–65% of placebo/untreated animals.
ND = not done.
From reference [138].
Acknowledgments
MR is supported by NIH grants R37 AI040877, R01s DE018293 and AI084133, the Swedish Ministry of Foreign Affairs, and the USAID Cooperative Agreement GPO-00-04-00019-00. MR is a 2002 Elizabeth Glaser Scientist.
GFD’s work was funded by USAID (GPO-A-8-00-08-00005-00) and the Bill and Melinda Gates Foundation (41266) grants. The views and opinions of the authors do not necessarily reflect those of the funding agencies or their institutions.
Footnotes
CONFLICTS OF INTEREST
As an employee of the non-profit organization, The Population Council, MR is involved in the research on the ZA-containing formulations, but has no financial interest in any of the organization’s research or products.
References
- 1.Barré-Sinoussi F, Chermann JC, Rey F, et al. Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS) Science. 1983;220:868–71. doi: 10.1126/science.6189183. [DOI] [PubMed] [Google Scholar]
- 2.Popovic M, Sarngadharan MG, Read E, et al. Detection, isolation, and continuous production of cytopathic retroviruses (HTLV-III) from patients with AIDS and pre-AIDS. Science. 1984;224:497–500. doi: 10.1126/science.6200935. [DOI] [PubMed] [Google Scholar]
- 3.Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med. 2009;36:2209–20. doi: 10.1056/NEJMoa0908492. [DOI] [PubMed] [Google Scholar]
- 4.Palmisano L, Vella S. A brief history of antiretroviral therapy of HIV infection: success and challenges. Ann Ist Super Sanita. 2011;47:44–8. doi: 10.4415/ANN_11_01_10. [DOI] [PubMed] [Google Scholar]
- 5.UNAIDS Report on the Global AIDS Epidemic. 2010. [Google Scholar]
- 6.Hodgen GD. HIV and Contraception: Research Priorities. In: Alexander NJ, Gabelnick HL, Spieler JM, editors. Heterosexual Transmission of AIDS. New York: Wiley-Liss; 1990. [Google Scholar]
- 7.Elias CJ, Heise L. The development of microbicides: A new method of HIV prevention for women. New York: The Population Council; 1993. [Google Scholar]
- 8.Stein ZA. Vaginal microbicides and prevention of HIV infection. Lancet. 1994;343:362–63. doi: 10.1016/s0140-6736(94)91205-x. [DOI] [PubMed] [Google Scholar]
- 9.Abu-Raddad LJ, Magaret AS, Celum C, et al. Genital herpes has played a more important role than any other sexually transmitted infection in driving HIV prevalence in Africa. PLoS One. 2008;3(5):e2230. doi: 10.1371/journal.pone.0002230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Buttar HS, Swierenga SHH, Matula TI. Evaluation of the cytotoxicity and genotoxicity of the spermicides nonoxynol-9 and Octoxynol-9. ToxicolLett. 1986;1:65–73. doi: 10.1016/0378-4274(86)90195-5. [DOI] [PubMed] [Google Scholar]
- 11.Hicks DR, Martin LS, Getchell JP, et al. Inactivation of HTLV-III/Lav infected cultures of normal human lymphocytes by nonoxynol-9 in vitro. Lancet. 1985;2:1422–23. doi: 10.1016/s0140-6736(85)92584-x. [DOI] [PubMed] [Google Scholar]
- 12.Malkovsky M, Newell A, Dalgleish AG. Inactivation of HIV by nonoxynol-9. Lancet. 1988;1:645. doi: 10.1016/s0140-6736(88)91440-7. [DOI] [PubMed] [Google Scholar]
- 13.Singh B, Cutler JC, Utidjian HMD. Studies on the development of a vaginal preparation providing both prophylaxis against venereal disease and other genital infections and contraception. II. Effect in vitro of vaginal contraceptive and non-contraceptive preparations on Trepenemapallidum and Neisseria gonorrhoeae. Br J Vener Dis. 1972;48:57–64. doi: 10.1136/sti.48.1.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Benes M, McCormick WM. Inhibition of growth of Chlamydia Trachomatis by nonoxynol-9 in vitro. Antimicrob Agents Chemother. 1985;27:724–6. doi: 10.1128/aac.27.5.724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Asculai SS, Weis MT, Rancourt MW, et al. Inactivation of herpes simplex viruses by nonionic surfactants. Antimicrob Agents Chemother. 1978;13:686–90. doi: 10.1128/aac.13.4.686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Miller CJ, Alexander NJ, Gettie A, et al. The effect of contraceptives containing nonoxynol-9 on the genital transmission of simian immunodeficiency virus in rhesus macaques. FertilSteril. 1992;259:1851–53. [PubMed] [Google Scholar]
- 17.Hiller SL, Moench T, Shattock R, Black R, Reichelderfer P, Veronese F. In vitro and In vivo: The story of nonoxynol-9. JAcquir Immune DeficSyndr. 2005;39:1–8. doi: 10.1097/01.qai.0000159671.25950.74. [DOI] [PubMed] [Google Scholar]
- 18.Vandebosch A, Goetghebeur E, Ramjee G, et al. Acceptability of COL-1492, a vaginal gel, among sex workers in one Asian and three African cities. Sex Transm Infect. 2004;80:241–3. doi: 10.1136/sti.2003.005934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Van Damme L, Chandeying V, Ramjee G, et al. Safety of multiple applications of COL-1492, a nonoxynol-9 vaginal gel, among female sex workers. COL-1492 Phase II Study Group. AIDS. 2000;14:85–8. doi: 10.1097/00002030-200001070-00010. [DOI] [PubMed] [Google Scholar]
- 20.Roddy RE, Zekeng L, Kelley R, et al. A controlled trial of nonoxynol 9 film to reduce male-to-female transmission of sexually transmitted diseases. New Eng J Med. 1998;339:504–10. doi: 10.1056/NEJM199808203390803. [DOI] [PubMed] [Google Scholar]
- 21.Richardson BA, Lavreys L, Martin HL, et al. Evaluation of a low-dose Nonoxynol-9 gel for prevention of sexually transmitted diseases. Sex Transm Dis. 2001;28:394–400. doi: 10.1097/00007435-200107000-00006. [DOI] [PubMed] [Google Scholar]
- 22.Van Damme L, Ramjee G, Vuuylsteke N, et al. Effectiveness of COL-1492, a nonoxynol-9 vaginal gel, on HIV-1 transmission in female sex workers: A randomized controlled trial. Lancet. 2002;360:971–77. doi: 10.1016/s0140-6736(02)11079-8. [DOI] [PubMed] [Google Scholar]
- 23.Phillips DM, Sudol KM, Taylor CL, et al. Lubricants containing N-9 may enhance rectal transmission of HIV and other STIs. Contraception. 2004;70(2):107–10. doi: 10.1016/j.contraception.2004.04.008. [DOI] [PubMed] [Google Scholar]
- 24.Krebs FC, Miller SR, Malamud D, Howett MK, Wigdahl B. Inactivation of human immunodeficiency virus type 1 by nonoxynol-9, C31G, or an alkyl sulfate, sodium dodecyl sulfate. Antiviral Res. 1999;43:157–73. doi: 10.1016/s0166-3542(99)00044-3. [DOI] [PubMed] [Google Scholar]
- 25.Krebs FC, Miller SR, Catalone BJ, et al. Sodium dodecyl sulfate and C31G as microbicidal alternatives to nonoxynol-9; comparative sensitivity of primary human vaginal keratinocytes. Antimicron Agents Chemother. 2000:1954–60. doi: 10.1128/aac.44.7.1954-1960.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Wyrick PB, Knight ST, Gerbig DG, Jr, et al. The microbicidal agent C31G inhibits Chlamydia trachomatis infectivity in vitro. Antimicrob Agents Chemother. 1997;41:1335–44. doi: 10.1128/aac.41.6.1335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Thompson KA, Malamud D, Storey BT. Assessment of the antimicrobial agent C31G as a spermicide: Comparison with nonoxynol-9. Contraception. 1996;53:313–8. [PubMed] [Google Scholar]
- 28.Mauck CK, Frezieres RG, Walsh TL, et al. Male tolerance study of 1% C31G. Contraception. 2004a;70:221–5. doi: 10.1016/j.contraception.2003.12.008. [DOI] [PubMed] [Google Scholar]
- 29.Bax R, Douville K, McCormick D, et al. Microbicides--evaluating multiple formulations of C31G. Contraception. 2002;66(5):365–8. doi: 10.1016/s0010-7824(02)00389-x. [DOI] [PubMed] [Google Scholar]
- 30.Mauck CK, Creinin MD, Barnhart KT, et al. A phase I comparative postcoital testing study of three concentrations of C31G. Contraception. 2004b;70:227–31. doi: 10.1016/j.contraception.2004.02.001. [DOI] [PubMed] [Google Scholar]
- 31.Mauck CK, Weiner DH, Creinin MD, et al. A randomized Phase I vaginal safety study of three concentrations of C31G vsExtra Strength Gynol II. Contraception. 2004c;70:233–40. doi: 10.1016/j.contraception.2004.04.010. [DOI] [PubMed] [Google Scholar]
- 32.Peterson L, Nanda K, Opoku BK, et al. SAVVY (C31G) gel for prevention of HIV infection in women: A phase 3, double-blind, randomized, placebo-controlled trial in Ghana. PLos ONE. 2007;2:e1312. doi: 10.1371/journal.pone.0001312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Feldblum PJ, Adeiga A, Bakare R, et al. SAVVY vaginal gel (C31G) for prevention of HIV infection: A randomized controlled trial in Nigeria. PLoS One. 2008;3:e1474. doi: 10.1371/journal.pone.0001474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Burke AE, Barnhart K, Jensen JT. Contraceptive efficacy, acceptability, and safety of C31G and nonoxynol-9 spermicidal gels: a randomized controlled trial. Obstet Gynecol. 2010;116:1265–73. doi: 10.1097/AOG.0b013e3181fc3b1a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Fichorova RN, Tucker LD, Anderson DJ. The molecular basis of nonoxynol-9-induced vaginal inflammation and its possible relevance to human immunodeficiency virus type 1 transmission. J Infect Dis. 2001;184:418–8. doi: 10.1086/322047. [DOI] [PubMed] [Google Scholar]
- 36.Fichorova RN, Bajpai M, Chandra N, et al. Interleukin (IL)-1, IL-6, and IL-8 predict mucosal toxicity of vaginal microbicidal contraceptives. Biol Reprod. 2004;71:761–9. doi: 10.1095/biolreprod.104.029603. [DOI] [PubMed] [Google Scholar]
- 37.Catalone BJ, Kish-Catalone TM, Budgeon LR, et al. Mouse model of cervicovaginal toxicity and inflammation for preclinical evaluation of topical vaginal microbicides. Antimicrob Agents Chemother. 2004;48:1837–47. doi: 10.1128/AAC.48.5.1837-1847.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wilson SS, Cheshenko N, Fakioglu E, et al. Susceptibility to genital herpes as a biomarker predictive of increased HIV risk: expansion of a murine model of microbicide safety. Antivir Ther. 2009;14:1113–2. doi: 10.3851/IMP1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Zalenskaya IA, Cerocchi OG, Joseph T, et al. Increased COX-2 Expression in Human Vaginal Epithelial Cells Exposed to Nonoxynol-9, a Vaginal Contraceptive Microbicide that Failed to Protect Women from HIV-1 Infection. Am J Reprod Immunol. 2011;65:569–77. doi: 10.1111/j.1600-0897.2010.00964.x. [DOI] [PubMed] [Google Scholar]
- 40.Martin LS, Loskoski SL, McDougal JS. Inactivation of human T-lymphotropic virus type III/lymphadenopathy-associated virus by formaldehyde-based reagents. Appl Environ Microbiol. 1987;53:708–9. doi: 10.1128/aem.53.4.708-709.1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ongradi J, Ceccherini-Nelli L, Pistello M, et al. Acid sensitivity of cell-free and cell-associated HIV-1: clinical implications. AIDS Res Hum Retroviruses. 1990;6:1433–36. doi: 10.1089/aid.1990.6.1433. [DOI] [PubMed] [Google Scholar]
- 42.Anderson DJ, Yunis EJ. “Trojan Horse” leukocytes in AIDS. N Engl J Med. 1983;309:984–5. [PubMed] [Google Scholar]
- 43.Quayle AJ, Zu C, Mayer KH, et al. T lymphocytes and macrophages, but not motile spermatozoa, are a significant source of human immunodeficiency virus in semen. J Infect Dis. 1995;176:960–8. doi: 10.1086/516541. [DOI] [PubMed] [Google Scholar]
- 44.Khanna KV, Whaley KJ, Zeitlin L, et al. Vaginal transmission of cell-associated HIV-1 in the mouse is blocked by a topical, membrane-modifying agent. J Clin Invest. 2002;109:205–11. doi: 10.1172/JCI13236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Sallé B, Brochard P, Bourry O, et al. Infection of macaques after vaginal exposure to cell-associated simian immunodeficiency virus. J Infect Dis. 2010;202:337–44. doi: 10.1086/653619. [DOI] [PubMed] [Google Scholar]
- 46.Olmsted SS, Khanna KV, Ng EM, et al. Low pH immobilizes and kills human leukocytes and prevents transmission of cell-associated HIV in a mouse model. BMC Infect Dis. 2005;5:79. doi: 10.1186/1471-2334-5-79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zeitlin L, Hoen TE, Achilles SL, et al. Tests of Buffergel for contraception and prevention of sexually transmitted diseases in animal models. Sex Transm Dis. 2001;28:417–23. doi: 10.1097/00007435-200107000-00010. [DOI] [PubMed] [Google Scholar]
- 48.Achilles SL, Shete PB, Whaley KJ, et al. Microbicide efficacy and toxicity tests in a mouse model for vaginal transmission of Chlamydia trachomatis. Sex Transm Dis. 2002;29:655–64. doi: 10.1097/00007435-200211000-00007. [DOI] [PubMed] [Google Scholar]
- 49.Spencer SE, Valentin-Bon IE, Whaley K, et al. Inhibition of Neisseria gonorrhoeae genital tract infection by leading-candidate topical microbicides in a mouse model. J Infect Dis. 2004;189:410–9. doi: 10.1086/381125. [DOI] [PubMed] [Google Scholar]
- 50.Tuyama AC, Cheshenko N, Carlucci MJ, et al. ACIDFORM inactivates herpes simplex virus and prevents genital herpes in a mouse model: optimal candidate for microbicide combinations. J Infect Dis. 2006;194:795–803. doi: 10.1086/506948. [DOI] [PubMed] [Google Scholar]
- 51.Olmsted SS, Dubin NH, Cone RA, et al. The rate at which human sperm are immobilized and killed by mild acidity. FertilSteril. 2000;73:687–93. doi: 10.1016/s0015-0282(99)00640-8. [DOI] [PubMed] [Google Scholar]
- 52.Atashili J, Poole C, Ndumbe PM, et al. Bacterial vaginosis and HIV acquisition: a meta-analysis of published studies. AIDS. 2008;22:1493–501. doi: 10.1097/QAD.0b013e3283021a37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Nagot N, Ouedraogo A, Defer MC, et al. Association between bacterial vaginosis and Herpes simplex virus type-2 infection: implications for HIV acquisition studies. Sex Transm Infect. 2007;83:365–8. doi: 10.1136/sti.2007.024794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gallo MF, Warner L, Macaluso M, et al. Risk factors for incident herpes simplex type 2 virus infection among women attending a sexually transmitted disease clinic. Sex Transm Dis. 2008;35:679–85. doi: 10.1097/OLQ.0b013e31816fcaf8. [DOI] [PubMed] [Google Scholar]
- 55.Cherpes TL, Meyn LA, Krohn MA, et al. Association between acquisition of herpes simplex virus type 2 in women and bacterial vaginosis. Clin Infect Dis. 2003;37:319–25. doi: 10.1086/375819. [DOI] [PubMed] [Google Scholar]
- 56.Kaul R, Nagelkerke NJ, Kimani J, et al. Prevalent herpes simplex virus type 2 infection is associated with altered vaginal flora and an increased susceptibility to multiple sexually transmitted infections. J Infect Dis. 2007;196:1692–7. doi: 10.1086/522006. [DOI] [PubMed] [Google Scholar]
- 57.Garg S, Anderson RA, Chany CJ, 2nd, et al. Properties of a new acid-buffering bioadhesive vaginal formulation (ACIDFORM) Contraception. 2001;64:67–75. doi: 10.1016/s0010-7824(01)00217-7. [DOI] [PubMed] [Google Scholar]
- 58.Fletcher PS, Harman SJ, Boothe AR, et al. Preclinical evaluation of lime juice as a topical microbicide candidate. Retrovirology. 2008;11(5):3. doi: 10.1186/1742-4690-5-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Mauck CK, Ballagh SA, Creinin MD, et al. Six-day randomized safety trial of intravaginal lime juice. J Acquir Immune DeficSyndr. 2008;49:243–50. doi: 10.1097/QAI.0b013e318186eae7. [DOI] [PubMed] [Google Scholar]
- 60.Amaral E, Faúndes A, Zaneveld L, et al. Study of the vaginal tolerance to Acidform, an acid-buffering, bioadhesive gel. Contraception. 1999;60:361–6. doi: 10.1016/s0010-7824(99)00102-x. [DOI] [PubMed] [Google Scholar]
- 61.Schwartz JL, Poindexter A, Schmitz SW, et al. Male tolerance of ACIDFORM gel. Contraception. 2005;71:443–6. doi: 10.1016/j.contraception.2004.11.008. [DOI] [PubMed] [Google Scholar]
- 62.Patton DL, Sweeney YC, Cummings PK, et al. Safety and efficacy evaluations for vaginal and rectal use of BufferGel in the macaque model. Sex Transm Dis. 2004;31:290–6. doi: 10.1097/01.olq.0000124614.91448.d4. [DOI] [PubMed] [Google Scholar]
- 63.Mayer KH, Peipert J, Fleming T, et al. Safety and tolerability of BufferGel, a novel vaginal microbicide, in women in the United States. Clin Infect Dis. 2001;32:476–82. doi: 10.1086/318496. [DOI] [PubMed] [Google Scholar]
- 64.van De Wijgert J, Fullem A, Kelly C, et al. Phase 1 trial of the topical microbicideBufferGel: safety results from four international sites. J Acquir Immune DeficSyndr. 2001;26:21–7. doi: 10.1097/00126334-200101010-00003. [DOI] [PubMed] [Google Scholar]
- 65.Barnhart KT, Rosenberg MJ, MacKay, et al. Contraceptive efficacy of a novel spermicidal microbicide used with a diaphragm: a randomized controlled trial. Obstet Gynecol. 2007;110:577–86. doi: 10.1097/01.AOG.0000278078.45640.13. [DOI] [PubMed] [Google Scholar]
- 66.Abdool Karim SS, Richardson BA, Ramjee G, et al. HIV Prevention Trials Network (HPTN) 035 Study Team. Safety and effectiveness of BufferGel and 0. 5% PRO 2000 gel for the prevention of HIV infection in women. AIDS. 2011;25:957–66. doi: 10.1097/QAD.0b013e32834541d9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Connor RI. Sensitivity of non-clade B primary HIV-1 isolates to mildly acidic pH. J Acquir Immune DeficSyndr. 2006;43:499–501. doi: 10.1097/01.qai.0000243048.50451.18. [DOI] [PubMed] [Google Scholar]
- 68.Zeitlin L, Whaley KJ, Hegarty TA, et al. Tests of vaginal microbicides in the mouse genital herpes model. Contraception. 1997;56:329–35. doi: 10.1016/s0010-7824(97)00154-6. [DOI] [PubMed] [Google Scholar]
- 69.Ito M, Baba M, Sato A, et al. Inhibitory effect of dextran sulfate and heparin on the replication of human immunodeficiency virus (HIV) in vitro. Antiviral Res. 1987;7:361–7. doi: 10.1016/0166-3542(87)90018-0. [DOI] [PubMed] [Google Scholar]
- 70.Mitsuya H, Looney DJ, Kuno S, et al. Dextran sulfate suppression of viruses in the HIV family: inhibition of virion binding to CD4+ cells. Science. 1988;240:646–9. doi: 10.1126/science.2452480. [DOI] [PubMed] [Google Scholar]
- 71.Baba M, Nakajima M, Schols D, et al. Pentosan polysulfate, a sulfated oligosaccharide, is a potent and selective anti-HIV agent in vitro. Antiviral Res. 1988;9:335–43. doi: 10.1016/0166-3542(88)90035-6. [DOI] [PubMed] [Google Scholar]
- 72.Pearce-Pratt R, Phillips DM. Sulfated polysaccharides inhibit lymphocyte-to-epithelial transmission of human immunodeficiency virus-1. Biol Reprod. 1996;54:173–82. doi: 10.1095/biolreprod54.1.173. [DOI] [PubMed] [Google Scholar]
- 73.Patton DL, Cosgrove-Sweeney YT, Paul KJ. A summary of preclinical topical microbicide vaginal safety and chlamydial efficacy evaluations in a pigtailed macaque model. Sex Transm Dis. 2008;35:889–97. doi: 10.1097/OLQ.0b013e31817dfdb8. [DOI] [PubMed] [Google Scholar]
- 74.Witvrouw M, De Clercq E. Sulfated polysaccharides extracted from sea algae as potential antiviral drugs. Gen Pharmacol. 1997;29:497–511. doi: 10.1016/s0306-3623(96)00563-0. [DOI] [PubMed] [Google Scholar]
- 75.Christensen ND, Reed CA, Culp TD, et al. Papillomavirus microbicidal activities of high-molecular-weight cellulose sulfate, dextran sulfate, and polystyrene sulfonate. Antimicrob Agents Chemother. 2001;45:3427–32. doi: 10.1128/AAC.45.12.3427-3432.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Anderson RA, Feathergill KA, Diao XH, et al. Preclinical evaluation of sodium cellulose sulfate (Ushercell) as a contraceptive antimicrobial agent. J Androl. 2002;23:426–38. [PubMed] [Google Scholar]
- 77.Saifuddin M, Doncel GF, Tsai L, et al. Intravaginal administration of 6% cellulose sulfate (CS) gel prevented systemic infection in rhesus macaques in a multiple dose R5/X4 SHIV vaginal challenge model. Abstract presented at Microbicides; 2008; New Delhi, India. 2008. [Google Scholar]
- 78.Mauck C, Weiner DH, Ballagh S, et al. Single and multiple exposure tolerance study of cellulose sulfate gel: a Phase I safety and colposcopy study. Contraception. 2001;64:383–91. doi: 10.1016/s0010-7824(01)00271-2. [DOI] [PubMed] [Google Scholar]
- 79.Malonza IM, Mirembe F, Nakabiito C, et al. Expanded Phase I safety and acceptability study of 6% cellulose sulfate vaginal gel. AIDS. 2005;19:2157–63. doi: 10.1097/01.aids.0000194797.59046.8f. [DOI] [PubMed] [Google Scholar]
- 80.Doh AS, Ngoh N, Roddy R, et al. Safety and acceptability of 6% cellulose sulfate vaginal gel applied four imes per day for 14 days. Contraception. 2007;76:245–9. doi: 10.1016/j.contraception.2007.05.083. [DOI] [PubMed] [Google Scholar]
- 81.Mauck CK, Freziers RG, Walsh TL, et al. Noncomparative contraceptive efficacy of cellulose sulfate gel. Obstet Gynecol. 2008;111:739–46. doi: 10.1097/AOG.0b013e3181644598. [DOI] [PubMed] [Google Scholar]
- 82.Van Damme L, Govinden R, Mirembe FM, et al. Lack of effectiveness of cellulose sulfate gel for the prevention of vaginal HIV transmission. N Engl J Med. 2008;359:463–72. doi: 10.1056/NEJMoa0707957. [DOI] [PubMed] [Google Scholar]
- 83.Halpern V, Ogunsola F, Obunge O, et al. Effectiveness of cellulose sulfate vaginal gel for the prevention of HIV infection: results of a Phase III trial in Nigeria. PLoS One. 2008;3:e3784. doi: 10.1371/journal.pone.0003784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Phillips DM. Intravaginal formulations to prevent HIV infection. Perspectives in Drug Discovery and Design. 1996;5:213–24. [Google Scholar]
- 85.Zacharopoulos VR, Phillips DM. Cell-mediated HTLV-I infection of a cervix-derived epithelial cell line. Microb Pathog. 1997;23:225–33. doi: 10.1006/mpat.1997.0151. [DOI] [PubMed] [Google Scholar]
- 86.Zaretzky FR, Pearce-Pratt R, Phillips DM. Sulfated polyanions block Chlamydia trachomatis infection of cervix-derived human epithelia. Infect Immun. 1995;63:3520–6. doi: 10.1128/iai.63.9.3520-3526.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Buck CB, Thompson CD, Roberts JN, et al. Carrageenan is a potent inhibitor of papillomavirus infection. PLoS Pathog. 2006;2:e69. doi: 10.1371/journal.ppat.0020069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Turville SG, Aravantinou M, Miller T, et al. Efficacy of Carraguard-based microbicides in vivo despite variable in vitro activity. PLoS One. 2008;3:e3162. doi: 10.1371/journal.pone.0003162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Dezzutti CS, James VN, Ramos A, et al. In vitro comparison of topical microbicides for prevention of human immunodeficiency virus type 1 transmission. Antimicrob Agents Chemother. 2004;48:3834–4. doi: 10.1128/AAC.48.10.3834-3844.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Skoler-Karpoff S, Ramjee G, Ahmed K, et al. Efficacy of Carraguard for prevention of HIV infection in women in South Africa: a randomised, double-blind, placebo-controlled trial. Lancet. 2008;372:1977–87. doi: 10.1016/S0140-6736(08)61842-5. [DOI] [PubMed] [Google Scholar]
- 91.Rusconi S, Moonis M, Merrill DP, et al. Naphthalene sulfonate polymers with CD4-blocking and anti-human immunodeficiency virus type 1 activities. Antimicrob Agents Chemother. 1996;40:234–6. doi: 10.1128/aac.40.1.234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Cheshenko N, Keller MJ, MasCasullo V, et al. Candidate topical microbicides bind herpes simplex virus glycoprotein B and prevent viral entry and cell-to-cell spread. Antimicrob Agents Chemother. 2004;48:2025–36. doi: 10.1128/AAC.48.6.2025-2036.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Weber J, Nunn A, O’Connor T, et al. ‘Chemical condoms’ for the prevention of HIV infection: evaluation of novel agents against SHIV(89. 6PD) in vitro and in vivo. AIDS. 2001;15:1563–8. doi: 10.1097/00002030-200108170-00014. [DOI] [PubMed] [Google Scholar]
- 94.Lewis M. Efficacy of PRO 2000 in macaques. Abstract presented at Microbicides; 2008; New Delhi, India. 2008. [Google Scholar]
- 95.Van Damme L, Wright A, Depraetere K, et al. A phase I study of a novel potential intravaginal microbicide, PRO 2000, in healthy sexually inactive women. Sex Transm Infect. 2000;76:126–30. doi: 10.1136/sti.76.2.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Mayer KH, Karim SA, Kelly C, et al. Safety and tolerability of vaginal PRO 2000 gel in sexually active HIV-uninfected and abstinent HIV-infected women. AIDS. 2003;17:321–9. doi: 10.1097/00002030-200302140-00005. [DOI] [PubMed] [Google Scholar]
- 97.Kamali A, Byomire H, Muwonge C, et al. A randomised placebo-controlled safety and acceptability trial of PRO 2000 vaginal microbicide gel in sexually active women in Uganda. Sex Transm Infect. 2010;86:222–6. doi: 10.1136/sti.2009.038372. [DOI] [PubMed] [Google Scholar]
- 98.Karim SS, Richardson BA, Ramjee G, et al. Safety and effectiveness of BufferGel and 0. 5% PRO 2000 gel for the prevention of HIV infection in women. AIDS. 2011;25:957–66. doi: 10.1097/QAD.0b013e32834541d9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.McCormack S, Ramjee G, Kamali A, et al. PRO 2000 vaginal gel for prevention of HIV-1 infection (Microbicides Development Programme 301): a phase 3, randomised, double-blind, parallel-group trial. Lancet. 2010;376:1329–37. doi: 10.1016/S0140-6736(10)61086-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Pirrone V, Wigdahl B, Krebs FC. The rise and fall of polyanionic inhibitors of the human immunodeficiency virus type 1. Antiviral Res. 2011;90:168–82. doi: 10.1016/j.antiviral.2011.03.176. [DOI] [PubMed] [Google Scholar]
- 101.Grant RM, Lama JR, Anderson PL, et al. Preexposure chemoprophylaxis for HIV prevention in men who have sex with men. N Engl J Med. 2010;363:2587–99. doi: 10.1056/NEJMoa1011205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Baeten J, Cellum C, et al. Antiretroviral pre-exposure prophylaxis for HIV-1 prevention among women: The Partners PrEP study. IAS; July 17–20, 2011; Rome, Italy. p. abstract MOAX0106. [Google Scholar]
- 103.Abdool Karim Q, Abdool Karim SS, et al. Effectiveness and safety of tenofovir gel, an antiretroviral microbicide, for the prevention of HIV infection in women. Science. 2010;329:1168–74. doi: 10.1126/science.1193748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.McCarthy TD, Karellas P, Henderson SA, et al. Dendrimers as drugs: discovery and preclinical and clinical development of dendrimer-based microbicides for HIV and STI prevention. Mol Pharm. 2005;2:312–8. doi: 10.1021/mp050023q. [DOI] [PubMed] [Google Scholar]
- 105.Rupp R, Rosenthal SL, Stanberry LR. VivaGel (SPL7013 Gel): a candidate dendrimer-microbicide for the prevention of HIV and HSV infection. Int J Nanomedicine. 2007;2:561–6. [PMC free article] [PubMed] [Google Scholar]
- 106.Jiang YH, Emau P, Cairns JS, et al. SPL7013 gel as a topical microbicide for prevention of vaginal transmission of SHIV89. 6P in macaques. AIDS Res Hum Retroviruses. 2005;21:207–13. doi: 10.1089/aid.2005.21.207. [DOI] [PubMed] [Google Scholar]
- 107.Chen 2MY, Millwood IY, Wand H, et al. A randomized controlled trial of the safety of candidate microbicide SPL7013 gel when applied to the penis. J Acquir Immune Defic Syndr. 2009;50:375–80. doi: 10.1097/QAI.0b013e318198a7e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.O’Loughlin J, Millwood IY, McDonald HM, et al. Safety, tolerability, and pharmacokinetics of SPL7013 gel (VivaGel): a dose ranging, phase I study. Sex Transm Dis. 2010;37:100–4. doi: 10.1097/OLQ.0b013e3181bc0aac. [DOI] [PubMed] [Google Scholar]
- 109.Cohen CR, Brown J, Moscicki AB, et al. A phase I randomized placebo controlled trial of the safety of 3% SPL7013 Gel (VivaGel®) in healthy young women administered twice daily for 14 days. PLoS One. 2011;6:e16258. doi: 10.1371/journal.pone.0016258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.McGowan I, Gomez K, Bruder K, et al. Phase 1 randomized trial of the vaginal safety and acceptability of SPL7013 gel (VivaGel) in sexually active young women (MTN-004) AIDS. 2011;25:1057–64. doi: 10.1097/QAD.0b013e328346bd3e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Talwar GP, Raghuvanshi P, Mishra R, et al. Polyherbal formulations with wide spectrum antimicrobial activity against reproductive tract infections and sexually transmitted pathogens. Am J Reprod Immunol. 2000;43:144–51. doi: 10.1111/j.8755-8920.2000.430303.x. [DOI] [PubMed] [Google Scholar]
- 112.Joshi SN, Katti U, Godbole S, et al. Phase I safety study of Praneem polyherbal vaginal tablet use among HIV-uninfected women in Pune, India. Trans R Soc Trop Med Hyg. 2005;99:769–74. doi: 10.1016/j.trstmh.2005.01.007. [DOI] [PubMed] [Google Scholar]
- 113.Joglekar NS, Joshi SN, Navlakha SN, et al. Acceptability of Praneem polyherbal vaginal tablet among HIV uninfected women & their male partners in Pune, India-Phase I study. Indian J Med Res. 2006;123:547–52. [PubMed] [Google Scholar]
- 114.Joshi SN, Dutta S, Kumar BK, et al. Expanded safety study of Praneem polyherbal vaginal tablet among HIV-uninfected women in Pune, India: a phase II clinical trial report. Sex Transm Infect. 2008;84:343–7. doi: 10.1136/sti.2007.029207. [DOI] [PubMed] [Google Scholar]
- 115.Joglekar NS, Joshi SN, Deshpande SS, et al. Acceptability and adherence: findings from a Phase II study of a candidate vaginal microbicide, ‘Praneem polyherbal tablet’, in Pune, India. Trans R Soc Trop Med Hyg. 2010;104:412–5. doi: 10.1016/j.trstmh.2009.12.007. [DOI] [PubMed] [Google Scholar]
- 116.Salhan S, Tripathi V, Sehgal R, et al. A phase II randomized controlled trial to evaluate the safety and efficacy of Praneem polyherbal vaginal tablets compared with betadine vaginal pessary in women with symptoms of abnormal vaginal discharge. Asia Pac J Public Health. 2009;21:461–8. doi: 10.1177/1010539509344610. [DOI] [PubMed] [Google Scholar]
- 117.Kumel G, Schrader S, Zentgraf H, et al. The mechanism of the antiherpetic activity of zinc sulphate. J Gen Virol. 1990;71:2989–97. doi: 10.1099/0022-1317-71-12-2989. [DOI] [PubMed] [Google Scholar]
- 118.Haraguchi Y, Sakurai H, Hussain S, et al. Inhibition of HIV-1 infection by zinc group metal compounds. Antivir Res. 1999;43:123–33. doi: 10.1016/s0166-3542(99)00040-6. [DOI] [PubMed] [Google Scholar]
- 119.Arens M, Travis S. Zinc salts inactivate clinical isolates of herpes simplex virus in vitro. J Clin Microbiol. 2000;38:1758–62. doi: 10.1128/jcm.38.5.1758-1762.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Bourne N, Stegall R, Montano R, Meador M, et al. Efficacy and toxicity of zinc salts as candidate topical microbicides against vaginal herpes simplex virus type 2 infection. Antimicrob Agents Chemother. 2005;49:1181–3. doi: 10.1128/AAC.49.3.1181-1183.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Geist FC, Bateman JA, Hayden FG. In vitro activity of zinc salts against human rhinoviruses. Antimicrob Agents Chemother. 1987;31:622–4. doi: 10.1128/aac.31.4.622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Lazarczyk M, Favre M. Role of Zn2+ ions in host-virus interactions. J Virol. 2008;82:11486–94. doi: 10.1128/JVI.01314-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Prasad AS, Beck FW, Bao B, et al. Duration and severity of symptoms and levels of plasma interleukin-1 receptor antagonist, soluble tumor necrosis factor receptor, and adhesion molecules in patients with common cold treated with zinc acetate. J Infect Dis. 2008;197:795–802. doi: 10.1086/528803. [DOI] [PubMed] [Google Scholar]
- 124.Suara RO, Crowe JE., Jr Effect of zinc salts on respiratory syncytial virus replication. Antimicrob Agents Chemother. 2004;48:783–90. doi: 10.1128/AAC.48.3.783-790.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Zhang ZY, Reardon IM, Hui JO, et al. Zinc inhibition of renin and the protease from human immunodeficiency virus type 1. Biochemistry. 1991;30 :8717–21. doi: 10.1021/bi00100a001. [DOI] [PubMed] [Google Scholar]
- 126.Shankar AH, Prasad AS. Zinc and immune function: the biological basis of altered resistance to infection. Am J Clin Nutr. 1998;68:447S–463S. doi: 10.1093/ajcn/68.2.447S. [DOI] [PubMed] [Google Scholar]
- 127.Rink L, Kirchner H. Zinc-altered immune function and cytokine production. J Nutr. 2000;130:1407S–11S. doi: 10.1093/jn/130.5.1407S. [DOI] [PubMed] [Google Scholar]
- 128.Li Q, Estes JD, Schlievert PM, et al. Glycerol monolaurate prevents mucosal SIV transmission. Nature. 2009;458:1034–8. doi: 10.1038/nature07831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Krieger JN, Rein MF. Zinc sensitivity of Trichomonas vaginalis: in vitro studies and clinical implications. J Infect Dis. 1982;146:341–5. doi: 10.1093/infdis/146.3.341. [DOI] [PubMed] [Google Scholar]
- 130.Andrews BJ, Mylvaganam H, Yule A. Sensitivity of Trichomonas vaginalis, Tritrichomonas foetus and Giardia intestinalis to bacitracin and its zinc salt in vitro. Trans R Soc Trop Med Hyg. 1994;88:704–6. doi: 10.1016/0035-9203(94)90239-9. [DOI] [PubMed] [Google Scholar]
- 131.Kilmarx PH, Blanchard K, Chaikummao S, et al. A randomized, placebo-controlled trial to assess the safety and acceptability of use of carraguard vaginal gel by heterosexual couples in Thailand. Sex Transm Dis. 2008;35:226–32. doi: 10.1097/OLQ.0b013e31815d6e0d. [DOI] [PubMed] [Google Scholar]
- 132.Kilmarx PH, van de Wijgert JH, Chaikummao S, et al. Safety and acceptability of the candidate microbicide Carraguard in Thai Women: findings from a Phase II Clinical Trial. J Acquir Immune Defic Syndr. 2006;43:327–34. doi: 10.1097/01.qai.0000243056.59860.c1. [DOI] [PubMed] [Google Scholar]
- 133.Cummins JE, Jr, Guarner J, Flowers L, et al. Preclinical testing of candidate topical microbicides for anti-human immunodeficiency virus type 1 activity and tissue toxicity in a human cervical explant culture. Antimicrob Agents Chemother. 2007;51:1770–9. doi: 10.1128/AAC.01129-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Whitehead SJ, Kilmarx PH, Blanchard K, et al. Acceptability of Carraguard vaginal gel use among Thai couples. AIDS. 2006;20 :2141–8. doi: 10.1097/QAD.0b013e32801086c9. [DOI] [PubMed] [Google Scholar]
- 135.Martin S, Blanchard K, Manopaiboon C, et al. Carragaurd acceptability among men and women in a couples study in Thailand. J Womens Health. 2010;19:1–7. doi: 10.1089/jwh.2009.1362. [DOI] [PubMed] [Google Scholar]
- 136.McLean CA, van de Wijgert JH, Jones HE, et al. HIV genital shedding and safety of Carraguard use by HIV-infected women: a crossover trial in Thailand. Aids. 2010;24:717–22. doi: 10.1097/QAD.0b013e328333bf89. [DOI] [PubMed] [Google Scholar]
- 137.Fernández-Romero JA, Thorn M, Titchen K, et al. Carrageenan/MIV-150 (PC-815), A Combination Microbicide. Sexually Transmitted Diseases. 2007;34:9–14. doi: 10.1097/01.olq.0000223287.46097.4b. [DOI] [PubMed] [Google Scholar]
- 138.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. doi: 10.1371/journal.pone.0015835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Veazey RS, Ketas TA, Klasse PJ, et al. Tropism-independent protection of macaques against vaginal transmission of three SHIVs by the HIV-1 fusion inhibitor T-1249. Proc Natl Acad Sci USA. 2008;105:10531–6. doi: 10.1073/pnas.0802666105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Veazey RS, Shattock RJ, Pope M, et al. Prevention of virus transmission to macaque monkeys by a vaginally applied monoclonal antibody to HIV-1 gp120. Nat Med. 2003;9:343–6. doi: 10.1038/nm833. [DOI] [PubMed] [Google Scholar]
- 141.Veazey RS, Klasse PJ, Schader SM, et al. Protection of macaques from vaginal SHIV challenge by vaginally delivered inhibitors of virus-cell fusion. Nature. 2005;438:99–102. doi: 10.1038/nature04055. [DOI] [PubMed] [Google Scholar]
- 142.Lederman MM, Veazey RS, Offord R, et al. Prevention of vaginal SHIV transmission in rhesus macaques through inhibition of CCR5. Science. 2004;306:485–7. doi: 10.1126/science.1099288. [DOI] [PubMed] [Google Scholar]
- 143.Parikh UM, Dobard C, Sharma S, et al. Complete protection from repeated vaginal simian-human immunodeficiency virus exposures in macaques by a topical gel containing tenofovir alone or with emtricitabine. J Virol. 2009;83:10358–65. doi: 10.1128/JVI.01073-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Cranage M, Sharpe S, Herrera C, et al. Prevention of SIV rectal transmission and priming of T cell responses in macaques after local pre-exposure application of tenofovir gel. PLoS Med. 2008;5:e157. doi: 10.1371/journal.pmed.0050157. discussion e157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Veazey RS, Ling B, Green LC, et al. Topically applied recombinant chemokine analogues fully protect macaques from vaginal simian-human immunodeficiency virus challenge. J Infect Dis. 2009;199:1525–7. doi: 10.1086/598685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Roberts JN, Buck CB, Thompson CD, Kines R, Bernardo M, et al. (2007) Genital transmission of HPV in a mouse model is potentiated by nonoxynol-9 and inhibited by carrageenan. Nat Med. 2007;13:857–61. doi: 10.1038/nm1598. [DOI] [PubMed] [Google Scholar]
- 147.Marais D, Gawarecki D, Allan B, et al. The effectiveness of Carraguard, a vaginal microbicide, in protecting women against high risk Human Papillomavirus infection. Antiviral Therapy. 2007 doi: 10.3851/IMP1890. In Press. [DOI] [PubMed]
- 148.Thurman AR, Clark MR, Doncel GF. Multipurpose Prevention Technologies: Biomedical Tools to Prevent HIV-1, HSV-2, and Unintended Pregnancies. Infect Dis Obstet Gynecol. 2011;2011:429403. doi: 10.1155/2011/429403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Moore MD, Cookson J, Coventry VK, et al. Protection of HIV neutralizing aptamers against rectal and vaginal nucleases: implications for RNA-based therapeutics. J Biol Chem. 2011;286:2526–35. doi: 10.1074/jbc.M110.178426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Moore MD, Bunka DH, Forzan M, et al. Generation of neutralizing aptamers against herpes simplex virus type 2: potential components of multivalent microbicides. J Gen Virol. 2011;92:1493–9. doi: 10.1099/vir.0.030601-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Wheeler LA, Trifonova R, Vrbanac V, et al. Inhibition of HIV transmission in human cervicovaginal explants and humanized mice using CD4 aptamer-siRNA chimeras. J Clin Invest. 2011;121:2401–12. doi: 10.1172/JCI45876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Bobardt MD, Chatterji U, Schaffer L, et al. Syndecan-Fc hybrid molecule as a potent in vitro microbicidal anti-HIV-1 agent. Antimicrob Agents Chemother. 2010;54:2753–66. doi: 10.1128/AAC.01606-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Zeitlin L, Whaley KJ. Microbicides for preventing transmission of genital herpes. Herpes. 2002;9:4–9. [PubMed] [Google Scholar]
- 154.Sexton A, Harman S, Shattock RJ, et al. Design, expression, and characterization of a multivalent, combination HIV microbicide. FASEB J. 2009;23:3590–600. doi: 10.1096/fj.09-131995. [DOI] [PubMed] [Google Scholar]
- 155.Walker LM, Huber M, Doores KJ, et al. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature. 2011 Aug 17; doi: 10.1038/nature10373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Mori T, O’Keefe BR, Sowder RC, 2nd, et al. Isolation and characterization of griffithsin, a novel HIV-inactivating protein, from the red alga Griffithsia sp. J Biol Chem. 2005;280:9345–53. doi: 10.1074/jbc.M411122200. [DOI] [PubMed] [Google Scholar]
- 157.Cole AM, Patton DL, Rohan LC, et al. The formulated microbicide RC-101 was safe and antivirally active following intravaginal application in pigtailed macaques. PLoS One. 2010;5:e15111. doi: 10.1371/journal.pone.0015111. [DOI] [PMC free article] [PubMed] [Google Scholar]