Skip to content

July 2017

By Jeremiah Johnson and Richard Jefferys

INTRODUCTION

Recent advances in the research, development, and implementation of biomedical HIV prevention—primarily in the form of treatment as prevention (TasP) and tenofovir disoproxil fumarate (TDF)/ emtricitabine (FTC) as pre-exposure prophylaxis (PrEP)—already appear to be bearing fruit in addressing complex HIV epidemics. At this year’s Conference on Retroviruses and Opportunistic Infections (CROI), the Centers for Disease Control and Prevention (CDC) presented their first HIV incidence estimates in six years, showing declines in new infections overall, including among white men who have sex with men (MSM).1 Last year, a Lancet article looking at incidence in Danish MSM found that, thanks to very high levels of viral suppression among HIV-positive MSM, new infections have been declining since 1996, nearly reaching the World Health Organization (WHO) elimination threshold by 2013.2 A 42 percent decline in HIV diagnoses among MSM in London’s Dean Street STI clinic, which diagnoses one in four of London’s HIV infections, also seems strongly linked to increased testing, treatment, and community advocacy to connect men to PrEP in spite of National Health Service England’s ongoing refusal to cover PrEP.3 Given the persistence of HIV epidemics among MSM, these successes indicate that we may at last have prevention tools that can end some of the most stubborn epidemics.

Not all of the news is rosy, of course. Racial disparities in the United States in new incidence rates—including stagnant rates of infections among black MSM and rising infections in Latino MSM—are a reminder that we are far from dismantling the systemic racism that underlies disparate health outcomes in communities of color. The struggle to firmly establish the visibility of transgender men and women in research and data collection continues to leave gender-nonconforming individuals exceptionally vulnerable compared with other key populations.4 With over 200 documented new infections, largely attributed to injection drug use, since the end of 2014 in an Indiana town of only 4,200 people, we are reminded of the fragility of earlier victories in epidemics among people who inject drugs.5 UNAIDS has also sounded the alarm about declining international investments in HIV, which are happening at a time when HIV infections among adults have stopped declining and are rising in some regions.6 While the science of HIV prevention has never been more productive, unfortunately many of our triumphs continue to be overshadowed by the social, political, and economic barriers that greatly limit access for marginalized communities.

Ongoing HIV prevention research remains hopeful, however, with many possibilities for expanding and improving our current toolbox in the pipeline. A number of highly anticipated studies have launched in the past year to build upon recent exciting breakthroughs related to oral PrEP, long-acting injectable PrEP, and vaginal rings. Gilead Sciences began recruitment in the fall of 2016 to study the efficacy of Descovy, their new tenofovir alafenamide (TAF)-based version of Truvada, as PrEP. After a number of missteps that led community advocates to call for a halt to the study—including lack of transparency and community oversight—the phase III trial is now moving forward with separate community advisory groups being convened for North American and European trial sites. Despite concerns related to the long pharmacokinetics (PK) “tail” observed with long-actingy injectable cabotegravir (CAB LA), a phase III trial looking at its efficacy in MSM and transgender women launched in December of last year. A primary challenge for implementation would be that individuals may need to commit to taking oral PrEP for a  year or more following their final injection in order to avoid becoming infected with HIV and developing resistance as a result of the subtherapeutic levels of cabotegravir.

The International Partnership for Microbicides (IPM) is moving ahead with follow-up assessments and analyses related to their vaginal ring containing dapivirine, which last year was reported to reduce new infections in two simultaneous studies by approximately one-third overall, with greater protection occurring in both trials among women 22 years of age and older and little to no protection among women 21 years of age and younger.7

One new concept for prevention of bacterial sexually transmitted infections (STIs) that has gained more attention in recent years has been the use of doxycycline as a PrEP or post-exposure prophylaxis (PEP) for gonorrhea, chlamydia, and syphilis. Although the real-world possibilities for implementation remain unclear, particularly considering ongoing concerns related to drug-resistant gonorrhea, more research is being planned to assess doxycycline for prevention.

A few short years ago, passive immunization—the infusion or injection of antibodies—was the tiniest of blips on the biomedical prevention radar. Today it represents a busy and expanding area of research, due to the discovery and characterization of an ever-increasing number of broadly neutralizing antibodies (bNAbs), which are capable of potently inhibiting diverse HIV variants from multiple global clades.8 Several bNAbs have been manufactured for clinical testing, and the furthest along the developmental pathway, VRC01, is the subject of two large efficacy trials known as the AMP studies.9 The rise of passive immunization provides an important example of how a technological breakthrough can revolutionize research: the identification of the new generation of potent bNAbs was made possible by techniques that can isolate and clone the antibodies being produced by individual B cells among many millions sampled from an individual.10,11,12 The U.S. National Institutes of Health (NIH), whose funding is now under serious threat from the Trump administration, provided the support for much of this critical work. In an example of cross-pollination between biomedical prevention fields, bNAbs are also undergoing evaluation in microbicide formulations.13

The immunological process that leads to the generation of bNAbs in some HIV-positive individuals is typically long and complex, proceeding over several years,14 and reproducing this process with a vaccine—which remains the ultimate goal for researchers—presents a stern challenge. Incremental progress has continued in preclinical studies over the past year, and trials of vaccine constructs that may have the potential to guide B cells along the first steps toward bNAb production are expected to begin in 2018.15

In the meantime, vaccine candidates capable of inducing other types of immune responses that might lead to at least some level of protection—based on lessons learned from the RV144 trial in Thailand16—have advanced into an efficacy trial in South Africa, HIV Vaccine Trials Network (HVTN) 702, which began enrolling last fall.17

Table 1. PrEP and Microbicides Pipeline 2017

Agent Class/Type Manufacturer/Sponsor Delivery Status
ORAL FORMULATIONS
TAF + FTC NtRTI/NRTI Gilead Sciences Oral PrEP Phase III
TAF + FTC NtRTI/NRTI CONRAD Oral PrEP Phase I (in cisgender women)
Genvoya (EVG + COBI + FTC + TAF) INSTI/NtRTI/NRTI Emory University Oral PrEP Phase I
LONG-ACTING FORMULATIONS
Cabotegravir INSTI ViiV Healthcare IM Phase IIb/III
Rilpivirine NNRTI PATH IM Phase II
MICROBICIDE RINGS, GELS, ENEMAS, FILMS, AND OTHER INSERTABLES
Dapivirine NNRTI

 

 

 

 

IPM (vaginal ring and rectal gel also with MTN)

 

Vaginal ring Phase IIIb
Vaginal gel Phase II
Rectal gel Phase I (planned)
Vaginal film Phase I
Tenofovir NtRTI

 

 

CONRAD
Vaginal ring Phase I
Vaginal tablet Phase I
Tenofovir NtRTI Johns Hopkins University Enema Phase I
MIV150 NNRTI MTN Rectal gel Phase I (planned)
Elvitegravir INSTI MTN Rectal insert Phase I (planned)
IQP-0528 NNRTI ImQuest U19 Rectal gel Phase I
Griffithsin Cell-viral fusion–blocking agent U19 University of Louisville Rectal gel Phase I
PC-1005 NNRTI, ZA, CGN Population Council Vaginal and rectal gel Phase I
MVC EI IPM Vaginal and rectal gel Phase I
MVC + dapivirine EI/NNRTI IPM Vaginal ring Phase I
MK-2048 + vicriviroc CCR5 inhibitor/ INSTI MTN Vaginal ring Phase I
Dapivirine + darunavir NNRTI/PI CHAARM Vaginal gel Phase I
DS003 EI IPM Vaginal tablet Phase I
Dapivirine + DS003 NNRTI/EI IPM Vaginal ring Preclinical
Darunavir PI IPM Vaginal ring Preclinical
MULTIPURPOSE TECHNOLOGIES
Tenofovir + levonorgestrel NtRTI/HC CONRAD Vaginal ring Phase I
Dapivirine + levonorgestrel NNRTI/HC IPM + MTN Vaginal ring Phase I
MB66 Anti-HIV + anti-HSV antibodies LeafBio, Inc. Vaginal film Phase I

CGN, carrageenan; COBI, cobicistat; EI, entry inhibitor; EVG, elvitegravir; FTC, emtricitabine; HC, hormonal contraception; HSV, herpes simplex virus; IM, intramuscular; IPM, International Partnership for Microbicides; MVC, maraviroc; MTN, Microbicide Trials Network; INSTI, integrase strand transfer inhibitor; NNRTI, non-nucleoside analogue reverse transcriptase inhibitor; NRTI, nucleoside analogue reverse transcriptase inhibitor; NtRTI, nucleotide analogue reverse transcriptase inhibitor; PI, protease inhibitor; PrEP, pre-exposure prophylaxis; TAF,  tenofovir  alafenamide; ZA, zinc acetate

ORAL FORMULATIONS

With scale-up initiatives to bolster TDF/FTC awareness and utilization where it is approved as PrEP under way—along with ongoing efforts to see that the coformulation is registered and covered by national health programs in other countries—additional oral products are making their way down the biomedical prevention pipeline.

The advantages of these compounds, which include Gilead’s TAF plus FTC (Descovy) and possibly its other TAF-based single-tablet regimen product that includes elvitegravir, cobicistat, and FTC (E/C/F/TAF; Genvoya)—as PrEP remain unclear.18,19 Possibilities include improved markers of renal and bone safety relative to TDF-inclusive regimens. Although kidney and bone problems remain uncommon and mild and are almost always reversible following drug cessation among long-term TDF/FTC PrEP users in clinical trial and demonstration project cohorts, new oral compounds may prove to be useful for those with other risk factors (e.g., underlying renal insufficiency, baseline bone mineral deficiency, concomitant use of nephrotoxic or bone-mineral-depleting medications, and advancing age).20,21,22,23,24,25

Updates for PEP have also been in the works. Last year, the CDC updated its guidelines for non- occupational PEP (nPEP).26 Researchers are also looking at dolutegravir (Tivicay), elvitegravir/cobicistat/ FTC/TDF (Stribild), and E/C/F/TAF as alternative PEP regimens that may improve adherence to and completion of the 28-day course of prophylaxis (see Text Box, page 41). The use of doxycycline as a PrEP and/or PEP for bacterial STIs has also gained interest in recent years, with studies presented at CROI 2015 and 2017 showing an effective reduction in STIs when doxycycline was used among MSM for prevention (see Text Box, page 43).

TAF and FTC

Like TDF, TAF is a prodrug formulation of tenofovir. Unlike TDF, which is converted in the blood to the active drug tenofovir diphosphate (TFV-DP) and then taken up into cells, TAF is primarily metabolized and converted to TFV-DP inside of cells. Using a much lower dose (25 mg), TAF achieves plasma tenofovir levels that are roughly 90 percent lower but intracellular concentrations that are approximately four- to sevenfold higher.27,28 The reduced systemic exposure has the potential for fewer renal- and bone-related toxicities compared with TDF. TAF’s low-milligram dosing also has the potential for reduced generic production costs and, ultimately, greater affordability versus TDF/FTC in low-income countries. Hence, TAF/FTC is also being eyed as an alternative to Truvada.

Enrollment for a phase III safety and efficacy trial comparing TAF/FTC to TDF/FTC for the prevention of HIV infections in HIV-negative men and transgender women who have sex with men is underway, with an estimated study completion date of September 2020.29 The DISCOVER trial is being run by Gilead Sciences, the manufacturer of both Descovy and Truvada, and began recruitment in September of 2016 with an estimated 5,000 participants set to be enrolled from across the United States, Canada, and

Western Europe. Participants will be randomized to two arms, one receiving active TAF/FTC and placebo TDF/FTC and the other receiving active TDF/FTC and placebo TAF/FTC. Following community pushback, Gilead modified the initial study protocol, which called for a 30-day washout period for individuals already on Truvada for PrEP.30,31 After at least 96 weeks of blinded treatment, and provided that TAF/  FTC shows sufficient efficacy, the study will be unblinded and participants will be offered the option to continue as part of an open-label extension of DISCOVER.

With TDF set to go off patent in the United States at the end of this year and FTC going off patent in 2021, there is little mystery as to why Gilead has taken TAF off the shelf—after development was inexplicably delayed for the past decade32—and is now following up on the FDA approval of Descovy or HIV treatment by aggressively pursuing a phase III trial of F/TAF as PrEP before generics can come to market. If F/TAF is shown to be noninferior, its improved safety profile may give a competitive edge to Descovy over generic TDF/3TC or, eventually, TDF/FTC. Given the already excellent safety profile of TDF/FTC, health care professionals and potential PrEP users should be wary of this scheme. While F/TAF as PrEP will be the better option for some, particularly individuals with decreases in renal function, for the vast majority of PrEP users the additional financial costs of Descovy will greatly outweigh the additional benefits compared with generics.

Researchers are confident that Descovy will be noninferior to Truvada as PrEP, given positive outcomes in nonhuman primate trials. Results from CDC evaluations of TAF plus FTC in rhesus macaques that were rectally challenged with simian-human immunodeficiency virus (SHIV) were published last year and more thoroughly covered in last year’s Pipeline Report.33 None of the TAF-treated macaques were infected after 19 exposures—100 percent protection—whereas the previous macaque studies of TDF/FTC suggested 94 percent protection after 14 SHIV exposures.

Making heads or tails of macaque and human tissue studies has been difficult. Despite apparent protection, rectal concentrations of TFV-DP of macaques treated with TAF were lower than those of the macaques treated in previous studies with TDF. In another study presented at CROI 2016 that looked at TFV and TFV-DP concentrations in the mucosal tissues of eight HIV-negative cisgender women, the plasma levels of TFV were 19-fold lower and peripheral blood mononuclear cell levels of TFV-DP were ninefold higher than those seen following single-dose TDF 300 mg dosing in an earlier study.34 Conversely, intracellular concentrations in biopsied tissues proved to be significantly lower: twofold in cervicovaginal samples and 13-fold in rectal samples. And, compared with TDF, TAF administration resulted in a higher percentage of tissue samples with undetectable drug levels: 63 percent of the rectal and 75 percent of genital tract samples had TFV and TFV-DP concentrations below the level of detection.

While DISCOVER will seek to answer lingering questions and determine efficacy for men and transgender women who have sex with men, CONRAD has launched additional investigation aimed at assessing the pharmacology of TAF in cervicovaginal tissues as a next step for understanding the potential value of TAF as PrEP for cisgender women.35 The phase I trial is estimated to be completed by October of this year and will give greater insights into whether TAF/FTC is likely to show efficacy in a larger trial.

PEP Updates

A 2014 meta-analysis of randomized and nonrandomized studies reporting completion rates for PEP revealed low levels of completion of PEP in the 28 days following a possible exposure to HIV.36 Researchers have been looking for alternative regimens that might have better completion outcomes. Last year, the CDC released an update to its 2005 nPEP guidelines listing TDF/FTC and raltegravir as the preferred regimen for nPEP, with TDF/FTC, darunavir, and ritonavir as possible alternatives.37 Research has shown, however, that the second daily dose of raltegravir may be challenging for people taking nPEP to remember.38 Ritonavir, with its well- known gastrointestinal side effects, may also complicate nPEP completion.

A recent study with results published in March of this year found that dolutegravir with TDF/FTC was a safe and well-tolerated option for once-daily PEP in 100 gay and bisexual Australian men in need of PEP.39 PEP completion was 90 percent (95% confidence interval [CI]: 84–96%). For the 10 men who did not complete dosing, nine were lost to follow-up and one discontinued due to headache. No participant was found to acquire HIV through week 12.

Another study looking at Stribild as PEP published favorable results in November, showing that among 234 participants who effectively received PEP, 215 (92%) completed 28 days of PEP, with only three switching from Stribild to another PEP because of side effects. More than 60 percent of participants reported at least one adverse event, which were mild to moderate. Fatigue and central neurological and abdominal side effects were the most frequently reported.40 Another study is preparing to evaluate Genvoya, Gilead’s TAF-based version of Stribild, as PEP.41 Researchers are hopeful that these single-tablet regimens will be capable of further improving adherence and completion.

TDF/FTC and Pregnancy

A number of studies are looking at the role of TDF/FTC in the lives of pregnant or postpartum women and for serodiscordant couples looking to conceive. Conception and pregnancy pose unique circumstances for HIV prevention; in women trying to conceive, condoms are obviously not a viable option for protection from HIV infection, and pregnant and postpartum women have been shown to be at increased risk of HIV infection largely due to reduced condom use.42 While treatment as prevention may itself be enough to protect the HIV-negative partner while trying to conceive,43 PrEP may contribute to peace of mind and presents a simpler, potentially cheaper, and less invasive solution than methods such as sperm washing or in vitro fertilization. It may also be a safe alternative to condoms for women during and just after pregnancy.

PrEP during pregnancy has not been specifically studied as part of randomized controlled trials; however, the safety of TDF/FTC for pregnant women and fetuses has been fairly well established. For years, HIV-positive women who have become pregnant have safely taken TDF/FTC as part of treatment with no increased likelihood of birth defects or adverse pregnancy outcomes reported in the Antiretroviral Pregnancy Registry.44 Additionally, researchers in the Partners PrEP study observed no statistically significant pregnancy-related complications among the 288 pregnancies that happened among study participants.45 Although pregnancy led to discontinuation from the trial and the study was not meant to specifically research PrEP during pregnancy, investigators estimate that fetuses may have been exposed to either tenofovir or TDF/FTC for a maximum of six weeks each. A study presented at HIV R4P last year also found that it was safe to breastfeed while still on PrEP.46 Still, there has previously been some indication that pregnant women taking TDF/FTC may give birth to slightly smaller babies with reduced bone density,47 making it preferable to reduce unnecessary exposure to TDF/FTC until better information becomes available.

The Microbicide Trials Network’s ongoing EMBRACE study (MTN-016), an HIV prevention agent pregnancy exposure registry that compiles information from pregnancies that occur during biomedical prevention trials, will hopefully shed further light on the effects of TDF/FTC in expectant mothers.48 In the meantime, some studies are attempting to develop better screening methodologies that will help limit uptake of PrEP in pregnant women with low risk of seroconversion.49

Two ongoing observational studies are looking specifically at PrEP as an option for safer conception.50,51 One from the University of California, San Francisco will compare uptake, adherence, and efficacy of PrEP, sperm washing, and/or artificial vaginal insemination offered to serodiscordant couples looking to conceive. Results from the study are expected in March 2019. Another study headed up by the University of Washington will look at pregnancy rates and HIV incidence when serodiscordant couples looking to conceive are counseled on TasP, PrEP, and timed condomless sex: results will be forthcoming in summer 2018.

Doxycycline for the Prevention of Bacterial STIs

Bacterial STIs have been shown to increase the likelihood that an individual will acquire or transmit HIV.52  Traditional STI prevention approaches, including behavior change related to frequency/number of sexual partners and levels of condom use, appear to be largely ineffective from a public health perspective. Syphilis rates among MSM in the United States and Western Europe have also been increasing since before the turn of the century—well before iPrEx demonstrated the efficacy of Truvada as PrEP—adding to the urgency for better, evidence-based options for the prevention of bacterial STIs.53

A small pilot study released in 2015 demonstrated that the antibiotic doxycycline provided as a PrEP may be effective in reducing STI incidence.54 The study was small, with only 30 gay men and transgender women, but it showed a statistically significant 70 percent decrease in STIs when half the participants were assigned doxycycline as PrEP and half the participants were offered financial incentives to avoid infections. Absolute numbers of syphilis, gonorrhea, and chlamydia infections were all lower in the doxycycline arm; however, the study was too small to provide statistically significant reductions when infections were broken down by specific disease.

A study presented at CROI 2017 showed that doxycycline provided as a PEP in oral HIV PrEP users led to a 47 percent reduction in bacterial STIs, with a 70 percent drop in chlamydia and a 73 percent drop in syphilis, but no reduction in gonorrhea.55 The study randomized 232 MSM from the French Ipergay PrEP study, with half of them being provided with doxycycline for STI PEP. Those in the treatment arm were told to take a 200 mg pill up to 72 hours after each episode, though nearly every participant who took a pill did so within 24 hours. Participants were followed for 8.7 months, with 212 participants-,106 in each arm, completing the study. Notably, STI percentages were extremely high in each arm, though the 38 percent annual STI incidence rate in the doxycycline arm was a significant improvement compared with 70 percent in the control arm.

Two new studies looking at doxycycline for STI prevention are being conducted by the British Columbia Centre for Disease Control.56,57 One is a smaller pilot study that will look at the feasibility and tolerability of using daily doxycycline for syphilis PrEP in a group of 50 HIV- negative MSM who are also taking Truvada as HIV PrEP. The second study is an early phase I study to determine whether the daily use of doxycycline is an efficacious and acceptable intervention for syphilis prevention in a group of 288 HIV-positive MSM. The study focusing on HIV-negative men currently has an estimated completion date of December 2017, whereas the study of HIV-positive men is set to run through May 2020. Antibiotic resistance, specifically in the case of gonorrhea, will be one of the major factors in considering the future of doxycycline as STI PrEP or PEP. Although doxycycline has not been recommended as treatment for gonorrhea for years, the threat of additional resistance remains a concern given that there are so few new antibiotics in the treatment pipeline for gonorrhea.

IMPLANTS AND INJECTABLE LONG-ACTING FORMULATIONS

Improving the acceptability of PrEP is one approach to strengthening adherence rates among populations at risk for HIV infection. Investment in subcutaneous implants to deliver antiretrovirals for PrEP has increased in the last year, including significant investment by the NIH and the Bill & Melinda Gates Foundation.61 Particular focus is also being placed on the development of long-acting nanosuspension formulations of antiretrovirals with PrEP potential, which may allow for doses that are separated by weeks or months. The drug furthest along the development path is CAB LA, ViiV Healthcare’s integrase strand transfer inhibitor (and dolutegravir analog); however, the unexpectedly long persistence of CAB LA in a significant minority of ECLAIR trial participants, possibly tied to higher body mass index (BMI), has led to some uncertainty about how to manage the long PK tail in some individuals. A long-acting injectable version of rilpivirine (RPV LA), Janssen’s non-nucleoside analogue reverse transcriptase inhibitor (NNRTI), remains on an uncertain course.

As long-acting formulations become more likely candidates for real-world use, it is imperative that researchers and key stakeholders begin actively looking at implementation challenges early. An NIH- funded review article published in 2015 looked at the importance of addressing long-acting formulation implementation issues at three levels: patient, provider, and system.62 Patient-level factors include targeted education and messaging, tailored supports to enhance acceptability and uptake, and effective strategies for promoting adherence/persistence and retention in care. Provider-level factors include engaging a broad mix of providers while ensuring adequate training and support for patient assessment, counseling, and follow-up. Systems-level factors include optimal delivery modalities, resource allocation, and ensuring access to populations most in need of new prevention options.

CAB LA

Encouraging preliminary results presented at CROI 2016 from the ECLAIR trial, which looked at the safety and tolerability of CAB LA as a PrEP, have led the HIV Prevention Trials Network (HPTN) to launch the first of two planned phase III studies looking at efficacy. However, significant questions remain about optimal dosing and feasibility of implementation given the unexpectedly long persistence of CAB LA in the plasma of a minority of ECLAIR participants.

Last year’s Pipeline Report gave a detailed review of the outcomes of the ECLAIR trial.63 The study randomized 127 HIV-negative men between 18 and 65 years of age and at low risk of acquiring HIV at screening to either CAB (N = 106) or placebo (N = 21). For the first four weeks of the trial, oral CAB (30 mg) or placebo were administered, followed by a seven-day washout period. The injection phase began at week 5 and ended at week 41, with CAB LA 800 mg or saline being administered via intramuscular (IM) injections during visits at weeks 5, 17, and 29. CAB LA was found to be well tolerated in comparison to placebo, although a minority of participants withdrew due to injection tolerability (4%) and a small proportion experienced grade 2 events such as fever, injection site itching, and injection site swelling. Two seroconversions were reported: one in the placebo group at week 23 and one in the CAB LA group at week 53, 24 weeks after the participant’s final injection; however, the participant in the CAB LA group who ultimately seroconverted had no detectable CAB in blood plasma at week 53. CAB PK data throughout each 12-week dosing interval were reported. Results showed trough concentrations  to be lower than the prespecified ideal at the end of the dosing intervals in approximately two-thirds of participants. On the basis of these findings, a new dosing strategy of 600 mg IM injections every eight weeks has been selected for CAB LA’s continued development.

The study also included a follow-up phase with preliminary results presented at the HIVR4P conference in October 2016. There, researchers reported that in 14 out of 86 participants (17%), drug levels of CAB LA remained above the lower limit of quantification but below the protein-adjusted 90% inhibitory concentration (PA-IC90) a year after their last injection.64 Persistence of CAB LA was associated with a higher range of BMIs, with higher BMIs leading to a longer PK tail. Additional covariate evaluation is warranted; however, these findings raise questions about CAB LA discontinuation and the possibility of drug resistance should individuals become infected with HIV while they maintain subtherapeutic yet quantifiable levels of CAB LA a year or more beyond their last injection.

To better understand the impact of CAB LA’s prolonged PK, a companion phase IIa study to ECLAIR, HPTN 077, has been extended by 24 weeks.65 The study will aim to find out how long measurable drug levels persist and if smaller and more frequent injections of 600 mg every 8 weeks may shorten the tail. HPTN 077 has enrolled approximately 200 HIV-negative volunteers in the United States, South America, and sub-Saharan Africa. The estimated primary completion date is now set for July 2017.

Despite ongoing questions related to CAB LA persistence, HPTN 083, a phase IIb/III head-to-head safety and efficacy trial of CAB LA versus oral TDF/FTC, was launched in December 2016.66 In step 1 of the trial, lasting five weeks, participants will receive oral TDF/FTC or oral CAB 30 mg daily, depending on the randomization. In step 2, participants will receive a daily oral placebo plus active CAB LA 3 mL injections at two time points four weeks apart and every eight weeks thereafter, or active daily oral TDF/ FTC plus placebo injections, for up to 180 weeks. In step 3, to cover the prolonged PK tail associated with CAB LA dosing, all participants will be required to take daily oral TDF/FTC for at least one year, starting no later than eight weeks after the last injection. The HPTN 083 trial has a planned enrollment of 4,500 trangender and MSM individuals 18 years of age and older who are at high risk for sexually acquiring HIV infection. The estimated study completion date is June 2020.

A companion study to HPTN 083, HPTN 084, is in the final stages of development, and a final protocol was posted on the HPTN website in March 2017 with plans to begin recruitment later this year.67 Approximately 3,200 HIV-uninfected cisgender women from sub-Saharan Africa will be enrolled and randomized 1:1 to active CAB LA and placebo TDF/FTC versus active TDF/FTC and placebo CAB LA in order to measure safety and efficacy of CAB LA in women. The study duration is expected to be 4.6 years. After the study reaches the required number of incident HIV endpoints, participants will begin an open-label daily oral TDF/FTC extension for approximately 48 weeks. As part of HPTN 084, an injectable contraceptive substudy will run simultaneously for 100 evaluable participants to study the effect of CAB LA on depot medroxyprogesterone acetate and norethisterone enanthate.

RPV LA

Encouraging phase I results from the SSAT 040 study evaluating the PK of RPV LA in plasma, the genital tract in women, and the rectum in men were published in 2014.68 Later that year, however, preliminary data from the MWRI-01 phase I study suggested that RPV LA’s activity in rectal versus cervicovaginal tissues may differ considerably.69 Although RPV levels following single 600 mg and 1,200 mg (2 × 600 mg) doses were higher in vaginal fluids versus rectal fluids, rectal tissues were found to have twice the concentration of RPV compared with vaginal tissues. In fact, rectal cell explants were fully resistant to HIV nearly two months after the 1,200 mg RPV LA injections were given, whereas vaginal and cervical cell explants appeared to be no better protected from HIV following either dose of the RPV LA.

A more recent study characterized the concentrations of RPV needed to prevent HIV infection in mucosal tissue.70 Although rectal tissue RPV levels appeared to be sufficient to block HIV infection—concentrations were approximately fivefold higher than what would be required to suppress viral infection—2.5-fold more drug was needed in female genital tissue to demonstrate similar inhibition. These data, the authors noted, support the explant findings from MWRI-01, in which HIV infection was suppressed in rectal tissue but not in cervicovaginal tissues.

Still under way is HPTN 076, a phase II safety and acceptability evaluation of RPV LA compared with placebo. The study is set to continue through October 2017, although preliminary results were presented at CROI 2017.71 A total of 136 (100 African, 36 U.S.) women were enrolled with a median age of 31 years. Among participants, 46 percent were married, 94 percent were black, and 60 percent were unemployed. The women were randomized (2:1) to receive either oral rilpivirine 25 mg or placebo daily for four weeks. In the absence of any safety signals, the participants received either 1,200 mg RPV LA (2 mL IM injections in both gluteal muscles) or placebo every eight weeks for a total of six injections.

Acceptability, safety, and PK data were collected throughout the study. The product was paused for any participant with a grade 2 or greater related adverse event or grade 3 or greater unrelated adverse event. Ten women withdrew (eight RPV vs. two placebo) and four had product discontinued (three RPV vs. one placebo) during the oral phase (weeks 0–4). A total of 122 (80 RPV LA vs. 42 placebo) women received one or more injections; 98 (64 RPV LA vs. 34 placebo) received all six injections. During the injection phase (weeks 4–52), one woman withdrew in the placebo group and 16 product discontinuations (10 RPV LA vs. 6 placebo) occurred. Of the product discontinuations, six (8%) RPV LA and two (5%) placebo were due to adverse events, including one placebo arm participant with prolonged QTc interval. Transient grade 2 or greater liver abnormalities occurred in nine (11%) of  the RPV LA participants compared with four (10%) in the placebo arm. Three RPV LA arm participants developed grade 3 or greater injection site reactions compared with none in the placebo arm. No significant difference in adverse events was observed between the two arms. Among participants who received one or more injections, the median trough concentration (Ctrough) of RPV was 68.2 ng/mL. At week 52 (eight weeks after last injection), the Ctrough was 91.9 ng/mL. The concentration two weeks after the first and second injections (at weeks 6 and 14) was 85.5 ng/mL and 113 ng/mL, respectively. At the last injection visit, 61 percent of women strongly agreed that they would definitely use and 73 percent that they would think about using a PrEP injectable in the future.

Overall the injections were safe, well tolerated, and acceptable. The lower-quartile RPV concentrations were consistently above the PA-IC90 at all times through eight weeks post-injection. However, based on the conflicting PK and explant infection data reported to date, compounded by the formulation’s need for cold-chain storage, there is no indication of RPV LA moving into phase III trials for PrEP.

Implantable Devices

Intarcia Therapeutics, a Boston-based company developing an implantable minipump about the size of a matchstick to deliver a drug for control of blood sugar in people with type 2 diabetes, has received a $50 million grant from the Bill & Melinda Gates Foundation to develop minipump technology to deliver antiretroviral drugs for PrEP, with an additional $90 million available if they are successful.72

Other researchers have looked at extended-release implants containing TAF. The Oak Crest Institute for Science (Monrovia, California) published encouraging animal PK data from a study of a subdermal delivery system similar to that used for removable contraceptive rods (e.g., Norplant).73 Auritec, a Pasadena drug delivery company, received NIH funding to test an implant containing TAF in dogs.74  The 40-day study found that the implant maintained drug levels 30 times higher than those needed to protect against HIV infection throughout the study period. The Sustained Long-Acting Protection from HIV (SLAP-HIV) partnership, based at Chicago’s Northwestern University and supported by a $17 million NIH grant, is working to develop an implant that can deliver either cabotegravir, rilpivirine, TAF, or the tenofovir analogue tenofovir exalidex.75

MICROBICIDES

Intravaginal Rings

With a growing body of data suggesting that antiretroviral-based prevention modalities are effective for women who are vulnerable to HIV infection, provided that adherence levels that are consistent, there has been considerable interest in more user-friendly and longer-acting technologies. Polymeric intravaginal rings (IVRs), similar to those used to control the release of estrogens or progestogens that provide contraceptive protection, are one such technology and are currently in various stages of development.

IPM’s dapivirine ring, which showed limited efficacy in sub-Saharan African women in the ASPIRE and Ring studies, has generated the most excitement; CONRAD has also completed a phase I trial for a tenofovir-containing ring.76 IPM and CONRAD are also both looking at versions of their rings that also contain the contraceptive levonorgestrel as a multipurpose prevention tool that may better meet the needs of women seeking to avoid both HIV and unwanted pregnancies.

Dapivirine

The most clinically advanced candidate is a silicone elastomer IVR containing 25 mg dapivirine (TMC120), an NNRTI licensed to IPM by Janssen Sciences Ireland UC. Data from two registrational trials, the Microbicide Trials Network’s ASPIRE study (MTN-020) and the International Partnership for Microbicides’ Ring Study (IPM 027), were reported at CROI 2016, with the final ASPIRE results being simultaneously published in the New England Journal of Medicine.77,78,79

ASPIRE, a phase III trial conducted at sites in Malawi, South Africa, Uganda, and Zimbabwe, randomized 2,629 HIV-negative women between 18 and 45 years of age to receive the dapivirine IVR or a matching placebo IVR, which were self-inserted and removed once a month for a year. The Ring Study, a phase II/III evaluation at six South African sites and one Ugandan site, compared the dapivirine IVR to a placebo IVR, inserted once every month over 24 months, in 1,959 HIV-negative women between 18 and 45.

Results from both studies, presented more comprehensively in last year’s Pipeline Report, suggested that the dapivirine IVR is safe and moderately effective at reducing incident HIV in African women. HIV infection rates were reduced by approximately one-third overall, with greater protection occurring in both trials among women 22 years of age and older: 56 percent in ASPIRE and 37 percent in the Ring Study, with little to no protection among women 21 years of age and younger—most likely due to lower levels of adherence.

An updated adherence analysis from ASPIRE presented at the 21st International AIDS Conference in Durban, South Africa, found that consistent users of the ring experienced 65 percent fewer infections compared to placebo.80 Rather than looking at blood levels of dapivirine, which may be influenced by participants reinserting the ring shortly before a follow-up visit, researchers refined their analysis by looking at the level of drug left behind in rings that were returned to researchers. A ring that has been worn for a full month should have 20–21 mg of drug remaining. Any level below 22 mg was treated as indicating medium to high adherence, whereas a ring with 23.5 mg or more indicated nonadherence. Of the 2,629 women enrolled in ASPIRE, 2,359 were included in this analysis. Compared to placebo, higher adherence to the active dapivirine ring was associated with a 65 percent (95% CI 23-84, p=0.009) reduction in HIV-1 risk. Results were similar both for the full-study population and when excluding the two sites with lower adherence/retention (risk reduction 67%, 95% CI: 23–86), and point estimates suggested HIV-1 protection for both women >21 years (risk reduction 72%, 95% CI: 21–90) and ≤21 years of age (risk reduction 50%, 95% CI: -78–86). Partial/low adherence was not significantly associated with HIV-1 protection (relative risk reduction 35%, 95% CI -10–61, P = .12).

Qualitative interviews with 214 participants were also published last year, providing insight into important issues related to adherence.81 The rings were largely acceptable to women; however, concerns about side effects, the appearance of the rings, and the experimental nature of the rings were highlighted as barriers. At clinical visits, women were asked, “How worried are you about having a vaginal ring inside you every day for at least a year?” While 29 percent of women reported this concern at the start of the study, only four percent of participants did so at their final follow-up clinic visit. Specific concerns related to use, health, hygiene, sexual enjoyment, and social approval also decreased significantly between the start and the end of the study.

Additionally, possible detection by male partners during sex and partner opinions were of importance to the women interviewed. Although fewer than five percent of all ASPIRE study participants reported incidents of intimate-partner-related violence or other social harms, women who did report violence or social harm within a month of the interview were nearly 2.5 times more likely to have low adherence to the ring. Younger age at enrollment, having a new primary partner, and not disclosing study participation or ring use to the primary partner were significantly associated with reporting social harms. Additional new data revealed that a majority of women—64 percent—disclosed the use of the ring to their male partners at the outset of the study, but 13 percent of study participants never revealed that they were using the ring. The investigators found that neither disclosing nor concealing use of the ring affected women’s adherence to the product.

IPM plans to submit the dossier of dapivirine IVR evidence required for licensure —ASPIRE and the Ring Study are only a part of an extensive research portfolio—to regulatory agencies. Two open- level evaluations of the dapivirine IVR are in the works.82,83 MTN-025, the HIV Open-Label Prevention Extension (HOPE) trial, is an ASPIRE follow-on study to assess continued safety and adherence, and it is currently enrolling. IPM hopes to conduct its own open-label extension follow-on study to provide former Ring Study participants with the dapivirine IVR.

Several follow-up safety studies are planned and being implemented. A trial looking at compatibility between the dapivirine ring and an antifungal clotrimazole cream commonly used to treat vaginal yeast infections is ongoing as is a trial to assess the presence of dapivirine in the breast milk of lactating women.84,85 A trial looking at tampon use and menses in women using the ring has been completed.

Plans to investigate the potential impact of bacterial vaginosis on ring efficacy are also underway after a substudy of 41 women from the FAME-04 vaginal microbicide study, presented at CROI 2017, found a significant correlation between higher levels of non-Lactobacillus bacteria and lower tenofovir levels in vaginal fluid and cervical tissue.86

Rectal Microbicide Gel and Enemas

Researchers are largely moving away from tenofovir-based rectal gels, partially due to concerns with developing an acceptable applicator. Instead, several phase I studies are set to look at other compounds for possible gel, insert, and suppository formulations.

MTN-026/IPM 038 is a phase I, randomized, double-blind, multi-site, placebo-controlled trial designed to evaluate the safety and acceptability of dapivirine gel (0.05%) when administered rectally to healthy, HIV-1–uninfected men and women.87 Another study, MTN-33/IPM 044, is a planned phase I study looking at the PK of the dapivirine gel when administered rectally via a vaginal applicator and a coital simulation device to healthy, HIV-1–uninfected men and transgender women. Participants will be randomized to administer a single dose of study product using an applicator of up to 10 mL of gel applied as a rectal lubricant using a phallic device to simulate anal sex. Specimens will be collected at multiple time points to assess drug concentrations, ex vivo efficacy, and biomarkers of safety. MTN-037 is a phase I trial looking at a rectal gel formulation for MIV150, a new NNRTI; MTN-039 is a phase I trial set to look at the integrase inhibitor elvitegravir as a rectal gel; and ImQuest is looking at another NNRTI- IQP-0528- in its own phase I study.88 The cell-viral fusion–blocking agent Griffithsin, which has been shown to inhibit both HIV and herpes simplex virus (HSV) infection, is also being assessed as a possible rectal gel at the University of Louisville.89

For at least five years, scientists have been looking at a rectal douche as a possible microbicide delivery system for protection during anal sex. Enemas, already frequently used in preparation for receptive anal sex, have the added benefit of achieving more comprehensive coverage compared with rectal gels.

A challenge with developing enemas is finding the right formulation with an osmolarity that is likely to lead to cellular uptake of the ARV. At HIVR4P in October, researchers presented promising results from a nonhuman primate study involving a tenofovir-containing gel that is hypo-osmolar.90 Four formulations were tested: two were iso-osmolar and two were hypo-osmolar. Two concentrations of tenofovir were tested: 1.76 and 5.28 mg/mL. Nonhuman primates were given a simple dose via rectal insertion and evacuation of the TFV liquid medium; researchers then measured concentrations of tenofovir in their blood and in rectal tissue biopsies an hour, a day, and three days after the dose. Explant challenges with simian immunodeficiency virus (SIV) were also conducted in each case. Hypo-osmolar formulations led to faster uptake of tenofovir, with the higher dose leading to drug concentrations both in blood and inside cells that were 5–11 times higher than any of the other formulations, with no indication of damage to rectal tissues with any formulation. Biopsies taken one hour after dosing with the high-dose hypo-osmolar formulation were completely protected from infection; 24 hours after dosing, two out of six samples became infected, compared with infections in biopsies from all other microbicide doses.

A study out of Johns Hopkins University is moving forward with this concept in humans. DREAM-01 is an early phase I open-label dose-escalation and variable-osmolarity study to compare the safety, PK, pharmacodynamics, and acceptability of three formulations of a TFV enema.91 Eighteen men will be enrolled, with results expected in October of this year. The goal of the study will be to identify the dose and osmolarity of a TFV enema for HIV PrEP that achieves the desired tenofovir diphosphate target concentrations in colonic mucosal mononuclear cells that have previously been shown to confer protection from HIV acquisition in MSM.

Vaginal Microbicide Gels

The future of vaginal microbicides remains uncertain following the disappointing data from both the FACTS 001 and VOICE studies evaluating 1% tenofovir gel.92,93 Given these results, CONRAD is reportedly moving away from tenofovir gels, although IVRs containing tenofovir remain in the pipeline. Although adherence, rather than potency, was believed to be the primary factor associated with poor efficacy in the FACTS 001 and VOICE studies, a number of gel-based microbicides containing alternative compounds—dapivirine, maraviroc, and a broad-spectrum coformulation of MIV-150, zinc acetate, and carrageenan (see below)—are at various stages of early development. Several of these products are also being evaluated for rectal use and protection.

PC-1005

The Population Council is developing PC-1005, a combination gel containing the NNRTI MIV-150, zinc acetate, and carrageenan. PC-1005 potentially offers protection not just against HIV but also against HSV-2 and human papillomavirus. Phase I safety, PK, acceptability, and adherence data were presented at CROI 2016 and published in JAIDS in December of last year.94,95,96 The trial enrolled 25 HIV-negative women between 19 and 44 years of age. Following a three-day open-label evaluation of PC-1005 in five participants, 20 women were randomized to apply PC-1005 4 mL or placebo once daily for 14 days. Seventeen women completed the randomized phase of the trial (two were lost to follow-up and one withdrew before dosing). There were no severe adverse events or early discontinuations because of adverse events. MIV-150 was absorbed systemically at low levels, and there was no measurable HIV and HPV activity in cervicovaginal lavages. Acceptability was also high: 94 percent of participants reported a willingness to use the gel in the future. Additional data also indicate that PC-1005 inhibits HIV and HSV-2 infection in cervical explants in a dose-dependent manner.

PrEP Breakthrough Infections

TDF/FTC (Truvada) as PrEP remains the most effective, thoroughly researched, evidence- based option for preventing sexual acquisition of HIV. Out of tens of thousands of individuals taking PrEP to date, only three cases of likely breakthrough infections have been documented, validating earlier mathematical modeling indicating that Truvada was up to 99 percent effective in preventing sexual infections if taken consistently in HIV-negative individuals. However, extremely rare instances of breakthrough infections tend to gain considerable—and disproportionate—media attention when they occur.

The first and most well-documented case of a breakthrough infection was reported in Boston at CROI 2016 regarding a Toronto gay man who reported high adherence to PrEP and consistently maintained three-month checkups with his physician.58 Dried blood spot (DBS) analysis of tenofovir levels in red blood cells showed excellent adherence leading up to the infection, as did high plasma concentrations of tenofovir at the patient’s follow-up visit, though these assessments could not completely rule out the potential for a brief lapse. Despite a high likelihood of consistent adherence, the man tested positive for HIV in April 2015— two years after starting PrEP. Resistance testing indicated that the man’s virus was totally resistant to FTC and carried mutations that conferred at least partial resistance to TDF. A similar second breakthrough infection coming out of New York City with convincing, though less conclusive, documentation was reported in October in Chicago at the HIV Research for Prevention (HIVR4P) conference.59 A gay man taking PrEP with reported good adherence was diagnosed with a strain of HIV resistant to both TDF and FTC. Due to a five-month break between visits, the man’s physician was unable to fully assess adherence for the entire period, though DBS testing did indicate excellent adherence over the prior three months. Both cases indicate that TDF/FTC may not be able to prevent infection from extremely rare viruses with resistance to both medications.

A third case reported at CROI 2017, involving a gay man from Amsterdam with a strain of HIV showing no resistance mutations, has raised the possibility that on extremely rare occasions even nonresistant strains might establish infection in spite of evidence of good PrEP adherence.60 There are many mysteries and questions in this case, however. The infection occurred in the six weeks following the man’s last doctor’s visit, meaning that a lapse in adherence cannot be ruled out. Also, the man reported two instances of injection drug use over the period in question, though he insisted that he had used sterile equipment.

These three cases stress the importance of routine provider visits while taking PrEP and provide greater insight into the conditions that could potentially lead to breakthrough infection. The extreme rarity of breakthrough infections confirms that although PrEP is not 100 percent effective, it remains the most effective prevention option for sexual acquisition of HIV to date.

PREVENTIVE VACCINES, PASSIVE IMMUNIZATION, AND ANTIBODY GENE TRANSFER

Table 2. HIV Vaccines, Passive Immunization, and Antibody Gene Transfer Pipeline 2017

Agent Class/Type Manufacturer/Sponsor Status
HIV VACCINES
ALVAC-HIV (vCP2438) + bivalent clade C gp120/MF59 Canarypox vector encoding HIV-1 clade C gp120, clade B gp41, Gag, and protease + protein boost comprising two clade C Env proteins (TV1.Cgp120 and 1086.Cgp120) NIAID/HVTN/Bill & Melinda Gates Foundation/ South African Medical Research Council/Sanofi Pasteur/GlaxoSmithKline Phase IIb/III
pGA2/JS7 DNA + MVA/HIV62 Prime: DNA vaccine Boost: MVA vector

Both encoding Gag, Pol, and Env proteins from HIV-1

clade B

GeoVax/NIAID Phase IIa
ALVAC-HIV vCP1521 Canarypox vector encoding HIV-1 CRF01_AE Env, clade B Gag, the protease-encoding portion of the Pol protein, and a synthetic polypeptide encompassing several known CD8+ T-cell epitopes from the Nef and Pol proteins Sanofi Pasteur/MHRP/NIAID Phase II
AIDSVAX B/E AIDSVAX B/E recombinant protein vaccine containing gp120 from HIV-1 clades B and CRF01_AE U.S. Army Medical Research and Materiel Command Phase II
HIVIS 03 DNA + MVA-CMDR Prime: HIVIS DNA encoding Env (A, B, C), Gag (A, B), reverse transcriptase (B), and Rev (B) proteins

Boost: MVA-CMDR encoding Env (E), Gag (A), and Pol

(E) proteins

Vecura/Karolinska Institutet/SMI/MHRP Phase II
LIPO-5 Five lipopeptides composed of CTL epitopes from Gag, Pol, and Nef proteins INSERM-ANRS Phase II
VICHREPOL Chimeric recombinant protein composed of C-terminal p17, full p24, and immunoreactive fragment of gp41 with polyoxidonium adjuvant Moscow Institute of Immunology/Russian Federation Ministry of Education and Science Phase II
Ad26.Mos.HIV

MVA-Mosaic gp140 protein

Ad26 vectors encoding mosaic Env, Gag, and Pol MVA vectors encoding mosaic Env, Gag, and Pol gp140 protein boost Janssen Vaccines & Prevention B.V./NIAID/MHRP/ IAVI/Beth Israel Deaconess Medical Center Phase I/IIa
ALVAC-HIV (vCP2438)

Bivalent clade C gp120/MF59 Bivalent clade C gp120/ASO1B

Canarypox vector encoding HIV-1 clade C gp120, clade B gp41, Gag, and protease + protein boost comprising two clade C Env proteins (TV1.Cgp120 and 1086.Cgp120) with either MF59 or AS01B adjuvant NIAID/GlaxoSmithKline/Sanofi Pasteur Phase I/IIa
DNA-C + NYVAC-C Prime: DNA vaccine encoding clade C Env, Gag, Pol, and Nef proteins

Boost: NYVAC-C attenuated vaccinia vector encoding

clade C Env, Gag, Pol, and Nef proteins

GENEART/Sanofi Pasteur/CAVD Phase I/II
MYM-V101 Virosome-based vaccine designed to induce mucosal IgA antibody responses to HIV-1 Env Mymetics Phase I/II
DNA-HIV-PT123 + AIDSVAX B/E DNA vectors encoding HIV-1 clade C Gag, gp140, and Pol-Nef

AIDSVAX B/E recombinant protein vaccine containing

gp120 from HIV-1 clades B and CRF01_AE

NIAID Phase Ib
Cervicovaginal CN54gp140-Hsp70 conjugate (TL01) HIV-1 clade C gp140 protein with Hsp70 adjuvant, delivered intravaginally St George’s, University of London/European Union Phase I
DCVax + poly-ICLC + MVA-CMDR Recombinant protein vaccine including a fusion protein comprising a human monoclonal antibody specific for the dendritic cell receptor DEC-205 and the HIV Gag p24 protein, plus poly-ICLC (Hiltonol) adjuvant, followed by  a boost with MVA-CMDR encoding Env, Gag, and Pol proteins Rockefeller University Phase I
DNA-HIV-PT123, NYVAC-HIV-PT1, NYVAC-HIV-PT4, AIDSVAX B/E DNA and NYVAC vectors encoding HIV-1 clade C Gag, gp140, and Pol-Nef

AIDSVAX B/E recombinant protein vaccine containing

gp120 from HIV-1 clades B and CRF01_AE

NIAID/IPPOX/EuroVacc/HVTN Phase I
DNA + Tiantan vaccinia vector Prime: DNA vector, with or without electroporation Boost: replication-competent recombinant Tiantan vaccinia strain vector

Both encoding Gag, Pol, and Env proteins from HIV-1

CN54

Chinese Center for Disease Control and Prevention/ National Vaccine and Serum Institute/Peking Union Medical College Phase I
EN41-FPA2 Gp41-based vaccine delivered intranasally and intramuscularly PX’Therapeutics/European Commission Phase I
GEO-D03 DNA + MVA/HIV62B Prime: DNA vaccine with GM-CSF adjuvant Boost: MVA vector

Both vaccines encode Gag, Pol, and Env proteins from

HIV-1 clade B and produce VLPs

GeoVax/NIAID Phase I
GSK HIV vaccine 732461 (F4) Gag, Pol, and Nef fusion protein in proprietary adjuvant AS01 GlaxoSmithKline Phase I Prime-boost Phase I with Ad35-GRIN
MAG-pDNA, Ad35-GRIN/ENV Multi-antigen DNA vaccine encoding the Env, Gag, Pol, Nef, Tat, and Vif proteins of HIV-1 and GENEVAX, IL-12 pDNA adjuvant, delivered using the electroporation-based TriGrid delivery system + two Ad35 vectors, one encoding HIV-1 clade A Gag, reverse transcriptase, integrase, and Nef, and the other encoding HIV-1 clade A Env (gp140) IAVI/Profectus Biosciences/Ichor Medical Systems Phase I
MAG-pDNA, rVSVIN HIV-1 Gag Multiantigen DNA vaccine encoding the Env, Gag, Pol, Nef, Tat, and Vif proteins of HIV-1 and GENEVAX, IL-12 pDNA adjuvant, attenuated replication-competent rVSV vector encoding HIV-1 Gag Profectus Biosciences/HVTN Phase I
MV1-F4-CT1 Recombinant measles vaccine vector encoding HIV-1 clade B Gag, Pol, and Nef Institut Pasteur Phase I
MVA.HIVA MVA vector encoding HIV-1 clade A Gag protein and 25 CD8+ T-cell epitopes IDT/University of Oxford/Medical Research Council/University of Nairobi/Kenya AIDS Vaccine Initiative Phase  I  in infants born to HIV-positive (PedVacc002) and HIV-negative (PedVacc001) mothers
MVA HIV-B MVA vector encoding HIV-1 Bx08 gp120 and HIV-1 IIIB Gag, Pol, and Nef Hospital Clinic of Barcelona Phase I
PENNVAX-G DNA + MVA-CMDR Prime: DNA vaccine encoding HIV-1 clade A, C, and D Env proteins and consensus Gag protein

Boost: MVA-CMDR live attenuated MVA vector encoding HIV-1 clade CRF_AE-01 Env and Gag/Pol proteins

DNA component administered intramuscularly via either

Biojector 2000 or CELLECTRA electroporation device

NIAID/MHRP/Walter Reed Army Institute of Research Phase I
PolyEnv1 EnvDNA Vaccinia viruses encoding 23 different Env proteins and DNA vaccine encoding multiple Env protein St. Jude Children’s Research Hospital Phase I
pSG2.HIVconsv DNA + ChAdV63. HIVconsv, or MVA.HIVconsv Prime: DNA vaccine pSG2

Boost: chimpanzee adenovirus vector ChAdV63 or MVA vector

All contain the HIVconsv immunogen,

designed to induce cross-clade T-cell responses by focusing on conserved parts of HIV-1

University of Oxford Phase I
Ad35-ENVA Ad35 vector encoding HIV-1 clade A Env Vaccine Research Center/NIAID Phase I
rVSVIN HIV-1 Gag Attenuated replication-competent rVSV vector encoding HIV-1 Gag Profectus Biosciences/HVTN Phase I
SAAVI DNA-C2, SAAVI MVA-C, clade C gp140/MF59 SAAVI DNA and MVA vectors encoding an HIV-1 clade C polyprotein including Gag-reverse transcriptase-Tat-Nef and an HIV-1 clade C truncated Env + Novartis protein subunit vaccine comprising a clade C oligomeric V2 loop- deleted gp140 given with MF59 adjuvant SAAVI/HVTN/Novartis Phase I
SeV-G(NP), Ad35-GRIN Sendai virus vector encoding HIV-1 Gag protein delivered intramuscularly or intranasally, Ad35 vector encoding HIV- 1 clade A Gag, reverse transcriptase, integrase, and Nef IAVI/DNAVEC Phase I
LIPO-5, MVA HIV-B, GTU-MultiHIV Five lipopeptides comprising CTL epitopes from Gag, Pol, and Nef proteins

MVA vector encoding Env, Gag, Pol, and Nef proteins from HIV clade B

DNA vector encoding fusion protein comprising elements from six different HIV proteins

Given in four different prime-boost combinations

INSERM-ANRS Phase I
Phase II
Ad4-mgag, Ad4-EnvC150 Live, replication-competent recombinant Ad4 vectors encoding HIV-1 clade C Env and HIV-1 mosaic Gag proteins

Formulated either as enteric-coated capsules for oral administration or as an aqueous formulation for tonsillar

administration

NIAID/PaxVax Phase I
DNA Nat-B Env, NYVAC Nat-B Env

DNA CON-S Env, NYVAC CON-S Env

DNA mosaic Env, NYVAC mosaic Env

Prime: DNA vector encoding Nat-B, CON-S, or mosaic Env proteins

Boost: NYVAC vectors encoding Nat-B, CON-S, or mosaic

Env proteins

HVTN/IPPOX/CHAVI Phase I
DNA, MVA-C, CN54rgp140 + GLA-AF DNA vectors encoding a Gag-Pol-Nef polypeptide and gp140 Env protein, both from clade C

MVA-C vector encoding Gag-Pol-Nef and gp120 Env protein from clade C

HIV-1 clade C gp140 protein and GLA-AF delivered

intramuscularly

Imperial College London/Medical Research Council/ Wellcome Trust Phase I
GTU-MultiHIV DNA vector encoding fusion protein comprising elements from six different HIV proteins, administered by intramuscular, intradermal, or transcutaneous routes Imperial College London/European Commission- CUT’HIVAC Consortium Phase I
DNA Nat-B Env DNA CON-S Env

DNA mosaic Env

MVA-CMDR

Prime: DNA vector encoding Nat-B, CON-S, or mosaic Env proteins

Boost: MVA vector encoding Env (E), Gag (A), and Pol

(E) proteins

NIAID/CHAVI/IPPOX/MHRP/HVTN Phase I
Trimeric gp140 Protein vaccine consisting of a trimeric gp120 Crucell/NIAID/Beth Israel Deaconess Medical Center Phase I
MVA mosaic MVA vectors encoding HIV-1 mosaic proteins Crucell/MHRP/NIAID/Beth Israel Deaconess Medical Center Phase I
DNA-HIV-PT123 AIDSVAXB/E DNA vectors encoding HIV-1 clade C Gag, gp140, and Pol-Nef

AIDSVAX B/E recombinant protein vaccine containing

gp120 from HIV-1 clades B and CRF01_AE

EuroVacc/IAVI/Uganda Medical Research Council/ UVRI Uganda Research Unit on AIDS/Centre Hospitalier Universitaire Vaudois Phase I
Oral Ad26 Orally administered replicating Ad26 vector encoding mosaic Env protein IAVI/University of Rochester/Beth Israel Deaconess Medical Center Phase I
PENNVAX-GP HIV-1 DNA vaccine

IL-12 DNA adjuvant

DNA vector encoding Gag, Pol, and Env proteins + DNA vector encoding IL-12 adjuvant, delivered via intradermal or intramuscular electroporation NIAID Phase I
IHV01 (FLSC-001) Full-length single-chain gp120-CD4 complex vaccine University of Maryland/Bill & Melinda Gates Foundation/Profectus BioSciences, Inc. Phase I
HIV DNA-C CN54ENV + recombinant HIV CN54gp140 DNA vector encoding HIV-1 clade C Env delivered intramuscularly and intradermally

Clade C Env protein boost

Imperial College London Phase I
Ad26.Mos.HIV + clade C gp140 Ad26 vectors encoding mosaic HIV-1 Env, Gag, and Pol + clade C HIV Env protein boost Janssen Vaccines & Prevention B.V. Phase I
HIV-1 Nef/Tat/Vif, Env pDNA + HIV-1 rVSV envC DNA vector encoding HIV-1 Nef/Tat/Vif and Env Attenuated replication-competent rVSV vector encoding HIV-1 clade C Env NIAID Phase I
Ad4-mgag, Ad4-EnvC150 + AIDSVAX

B/E

Orally administered replication-competent Ad4 HIV vaccine in combination with AIDSVAX B/E recombinant protein vaccine containing gp120 from HIV-1 clades B and CRF01_AE PaxVax, Inc./NIAID Phase I
Trivalent Ad26.Mos.HIV, tetravalent Ad26.Mos4.HIV + clade C gp140 Ad26 vectors encoding mosaic HIV-1 Env, Gag, and Pol or Ad26 vectors encoding two mosaic HIV-1 Envs, mosaic Gag, and Pol + clade C HIV Env protein boost Janssen Vaccines & Prevention B.V. Phase I
Tetravalent Ad26.Mos4.HIV + clade C gp140 ± mosaic gp140 Ad26 vectors encoding two mosaic HIV-1 Envs and mosaic Gag and Pol + clade C HIV Env protein boost ± mosaic HIV Env protein boost Janssen Vaccines & Prevention B.V. Phase I
MVA/HIV62B + AIDSVAX B/E MVA vector encoding Gag, Pol, and Env proteins from HIV-1 clade B to produce VLPs + AIDSVAX B/E recombinant protein vaccine containing gp120 from HIV-1 clades B and CRF01_AE NIAID Phase I
DNA-HIV-PT123

Bivalent clade C gp120/MF59 Bivalent clade C gp120/ASO1B

DNA vaccine encoding HIV-1 clade C Gag, gp140, and Pol-Nef + protein boost comprising two clade C Env proteins (TV1.Cgp120 and 1086.Cgp120) with either MF59 or AS01B adjuvant NIAID Phase I
DNA-HIV-PT123 + clade C gp120/ MF59 DNA vaccine encoding HIV-1 clade C Gag, gp140, and Pol-Nef + protein boost comprising two clade C Env proteins (TV1.Cgp120 and 1086.Cgp120) in MF59 adjuvant
PASSIVE IMMUNIZATION
VRC01 Monoclonal bNAb administered intravenously NIAID/HVTN/HPTN Phase IIb
10-1074 Monoclonal bNAb administered intravenously Rockefeller University Phase I
3BNC117 + 10-1074 Monoclonal bNAbs administered intravenously Rockefeller University Phase I
P2G12 Monoclonal neutralizing antibody administered intravenously St George’s, University of London Phase I
PGT121 Monoclonal bNAb administered intravenously IAVI Phase I
VRC01 Monoclonal bNAb administered subcutaneously or intravenously NIAID Phase I (adults and HIV-exposed infants)
VRC01LS LA monoclonal bNAb administered subcutaneously or intravenously NIAID Phase I
VRC07-523LS LA monoclonal bNAb administered intravenously or subcutaneously NIAID Phase I
ANTIBODY GENE TRANSFER
rAAV1-PG9DP Recombinant AAV vector encoding the PG9 broadly neutralizing antibody IAVI/NIAID/CHOP Phase I

AAV, adeno-associated virus Ad4, adenovirus serotype 4 Ad26, adenovirus serotype 26 Ad35, adenovirus serotype 35
BNAb, broadly neutralizing antibody
CAVD, Collaboration for AIDS Vaccine Discovery CHAVI, Center for HIV/AIDS Vaccine Immunology CHOP, Children’s Hospital of Philadelphia
CMDR, Chiang Mai double recombinant CTL, cytotoxic T lymphocyte
GLA-AF, glucopyranosyl lipid adjuvant (aqueous formulation) GM-CSF, granulocyte-macrophage colony–stimulating factor Hsp70, heat shock protein 70
HVTN, HIV Vaccine Trials Network
IAVI, International AIDS Vaccine Initiative IDT, Impfstoffwerk Dessau-Tornau
IL, interleukin
INSERM-ANRS, French National Institute for Health and Medical Research-French National Agency for Research on AIDS and Viral Hepatitis
LA, long-acting
MHRP, U.S. Military HIV Research Program MVA, modified vaccinia Ankara strain
NIAID, U.S. National Institute of Allergy and Infectious Diseases
Poly-ICLC, polyinosinic-polycytidylic acid with polylysine and carboxymethylcellulose
rVSV, recombinant vesicular stomatitis virus SAAVI, South Africa AIDS Vaccine Initiative
SMI, Swedish Institute for Infectious Disease Control UVRI, Uganda Virus Research Institute
VLP, virus-like particle

PASSIVE  IMMUNIZATION/ANTIBODY  GENE  TRANSFER

The Antibody-Mediated Prevention (AMP) trials represent a collaborative effort between the NIH-funded HVTN and the HPTN. The efficacy of the bNAb VRC01 will be assessed in two populations: HVTN 704/ HPTN 085 aims to enroll 2,700 MSM and transgender individuals who have sex with men at sites in Brazil, Peru, and the United States, whereas HVTN 703/HPTN 081 will recruit 1,500 sexually active women at sites in Botswana, Kenya, Malawi, Mozambique, South Africa, Tanzania, and Zimbabwe. The antibody is delivered by inpatient infusion every eight weeks, which is not ideal, but a key goal of the studies is to define protective bNAb levels and thus inform the development of potentially more potent and convenient bNAb formulations. Results are anticipated by 2022.

In addition to these large efficacy trials, there are a growing number of early-phase studies of more recently discovered bNAbs that have been demonstrated to have greater breadth and potency than VRC01. These include 3BNC11797, 10-107498, PGT12199, and VRC07-523LS.100

A combination of 3BNC117 and 10-1074 is also being tested, which may be an augury of the  future because resistance to individual bNAbs could limit their efficacy when used alone. VRC07- 523LS represents a derivative of a parent bNAb, VRC07, modified to enhance potency, breadth, and persistence in the body, thereby reducing dosing frequency—another strategy that may become more common as researchers seek ways to make passive immunization with bNAbs more user-friendly. The phase I VRC07-523LS trial is evaluating both intravenous and subcutaneous delivery.

Over the past several years, there has been considerable attention given to a potential one-shot bNAb delivery approach known as antibody gene transfer. The method draws from gene therapy research, employing adeno-associated virus (AAV) vectors modified with the genetic code for producing the bNAb of interest. Upon injection into muscle tissue, the AAV vector acts as a factory for persistent generation  of the bNAb.101 Promising results have been reported in the SIV/macaque model,102,103 and the first human trial—a collaboration between the scientist Phil Johnson and the International AIDS Vaccine Initiative (IAVI)—is ongoing, involving the bNAb PG9. A recent macaque study has illuminated a potential downside, however—the approach can induce the production of antibodies against the bNAbs, significantly reducing the levels that are maintained.104 Additional research will be required to better understand this problem and develop ways to address it.

Several research groups are exploring the possibility of administering bNAbs in microbicide formulations. A combination of three of the earliest generation of bNAbs to be discovered, 4E10, 2F5, and 2G12, has been evaluated in a phase I clinical trial and found to be safe.105 Antibody levels capable of inhibiting HIV were detectable in cervicovaginal secretions for up to eight hours after administration, and no systemic absorption was observed. A first-in-human trial launched last year is testing the bNAb VRC01 and an antibody against HSV106 delivered in a vaginal film (see table 1); the product is named MB66, and the antibodies are being produced in a new system using genetically modified tobacco plants.107

The potential for delivering MB66 via vaginal ring is also under investigation.108 A separate group of researchers has also used tobacco plants to produce a version of the 2G12 antibody designated P2G12; a single vaginal administration has been shown to be safe,109 and an ongoing trial at St George’s, University of London is now assessing intravenous delivery.

HIV VACCINES

The most significant recent news for the vaccine field has been the launching of HVTN 702, the first HIV vaccine efficacy trial to be conducted in seven years.110 Led by principal investigator Glenda Gray, the protocol plan is to enroll 5,400 men and women between the ages of 18 and 35 years who are at risk for HIV infection at 15 sites in South Africa. Participants will be randomized to receive placebo or ALVAC vCP2438 (a canarypox vector encoding HIV-1 clade C gp120, clade B gp41, Gag, and protease) plus a boost consisting of two clade C HIV gp120 proteins in MF59 adjuvant. The ALVAC vector is administered alone at baseline and after one month, and then in combination with the gp120 boost at months 3, 6, and 12.

The rationale for the study is derived from RV144, a large efficacy trial conducted in Thailand, which demonstrated that vaccination with similar candidates led to a small but statistically significant 31.2 percent reduction in risk of HIV acquisition.111 Of potential importance, the final boost in RV144 was given at six months, and there is evidence that protection may have peaked at around 60 percent after one year of follow-up and then declined as vaccine-induced immune responses waned—this has led to the inclusion of an additional booster after 12 months in HVTN 702.

The vaccine regimen has been tailored for the South African setting, where HIV-1 clade C is prevalent. A preparatory trial conducted in South Africa, HVTN 100, evaluated whether the vaccines induced the types of immune responses that were associated with protection in RV144 in the majority of South African recipients, in order to decide whether the larger efficacy trial was justified. As reported at the International AIDS Conference in Durban last year, the immune response criteria—which included binding antibodies to clade C gp120 antigens, V1V2 antibodies to clade gp70 scaffold antigens, and CD4+ T-cell responses to HIV Env—were all met.112

There is one aspect of HVTN 702 that has proven slightly controversial, and that is the selection of the adjuvant for the gp120 protein boost. The purpose of adjuvants is to help stimulate the induction of immune responses against the antigens in the vaccine, and in RV144 the gp120 protein boost was delivered with the common adjuvant alum. In HVTN 702, a proprietary squalene-based adjuvant developed by Novartis Vaccines (since acquired by GlaxoSmithKline) named MF59 is being used.

The controversy derives from a macaque experiment conducted by the research group of Genoveffa Franchini at the U.S. National Cancer Institute, which aimed to recapitulate the RV144 results in animals. The researchers reported that while the RV144 vaccine regimen showed some protective efficacy against an SIV challenge, this was not seen in a group of macaques that received the gp120 protein boost with MF59 instead of alum.113 Analyses also indicated that the alum adjuvant had activated particular genes related to innate immunity and that this was linked to protection against SIV challenge. However, these were post hoc findings because the experiment was not designed or statistically powered to compare the adjuvants, and a subsequent macaque study with an alum adjuvant (albeit not precisely the same) did not duplicate the results.114 Other researchers, including HVTN director Lawrence Corey, have pointed out that protection against SHIV infection has been reported in some macaque studies employing MF69 as a vaccine adjuvant,115 countering Franchini’s suggestion that it could have a negative effect.116 The debate has not altered the design of HVTN 702, but it is possible the issue could be revisited if no protection is observed; regular interim evaluations will be carried out by a data safety monitoring board, and the trial can be halted early if there is evidence the vaccines are failing or harmful.

An update on the status of HVTN 702 was provided on a webinar hosted by AVAC on May 8, 2017, by protocol co-chair Fatima Laher from Chris Hani Baragwanath Hospital in Soweto. The first immunizations began in October 2016 and, as of April 2017, 526 participants have been enrolled. Laher noted that the protocol has undergone a revision, with version 2 including additional details on the HIV prevention package offered to participants, including updated information related to obtaining access to PrEP. Collection of DBS samples from participants using PrEP has also been added in order to obtain data on Truvada drug levels.117

PrEP in Biomedical Prevention Trials

The efficacy of Truvada PrEP has raised difficult questions regarding how it should be integrated into trials of biomedical prevention interventions, whether vaccines, passive immunization, microbicides, or alternative forms of PrEP. Current UNAIDS/WHO guidelines118 recommend that clinical trials provide access to proven “state of the art” HIV prevention modalities for clinical trial participants, and an experimental intervention is tested to find out whether it can further reduce the risk of HIV acquisition when given in addition to these modalities. But Truvada PrEP is so efficacious that if all trial participants were to use it consistently as part of a background prevention package, evaluating whether a new experimental intervention has any significant effect on HIV risk would become extremely challenging—perhaps impossible. PrEP is not necessarily ideal for everyone, however, and this means that there remains a need to develop other user-friendly biomedical prevention technologies and also that trial participants who choose not to use PrEP (or for whom PrEP is not recommended) can ethically be included as participants in clinical trials. The HVTN 704/HPTN 085 AMP trial offers one example of how the issue of PrEP provision is currently being addressed: Truvada PrEP is being offered free of charge to all participants. Those participants based in the United States who choose to receive Truvada PrEP are referred to a program that integrates provision of the drug into their primary health care. Participants in Peru and Brazil, where Truvada is not yet licensed for PrEP, will be referred to demonstration projects.

In contrast, the HVTN 703/HPTN 081 AMP trial is offering information on Truvada PrEP and referrals to access programs where possible but is not providing the drug itself. The protocol explains that this approach is based on differing recommendations for PrEP use in women and the lack of local regulatory approvals, but it acknowledges HIV prevention standards are continually evolving and states “arrangements for provision of PrEP in this trial will take into account current evidence regarding PrEP efficacy in the populations to be enrolled in this trial, community consultation, guidance from international/regional/national/local and other regulatory authorities, and advice from persons/groups with bioethics and human subjects protection expertise.”

The differences between the protocols—both of which were reviewed and approved by multiple stakeholders, including community members and regulators—highlight the current gray areas regarding PrEP provision in biomedical prevention trials, which have been a topic of extensive discussion in the scientific literature.119,120,121 These discussions are likely to continue for the foreseeable future.

In addition to HVTN 702 and the work surrounding it, there is a second major thrust in HIV vaccine research being driven by Janssen Vaccines & Prevention B.V., part of the Janssen Pharmaceutical Companies of Johnson & Johnson. The company is sponsoring multiple studies involving combinations of two viral vectors—adenovirus serotype 26 (Ad26) and modified vaccinia Ankara strain (MVA)—and clade C gp140 Env protein boosts, with the goal of launching a first proof-of-concept efficacy trial in the near future. A key element of the program is the use of mosaic HIV antigens designed to induce immune responses capable of recognizing diverse viral variants. The research is being carried out incollaboration with Beth Israel Deaconess Medical Center/Harvard, the Bill & Melinda Gates Foundation, HVTN, IAVI, the U.S. Military HIV Research Program, the National Institute of Allergy and Infectious Diseases, and the Ragon Institute.

Several of the HIV vaccine trials that have begun over the past year are related to the Janssen program. Ad26 vectors are being administered as the priming immunizations in trivalent and tetravalent mixtures: the former includes two mosaic Gag-Pol antigens and a mosaic Env, and the latter adds a second mosaic Env. Booster immunizations comprise the same Ad26 mixtures or MVA vectors encoding two mosaic Gag- Pol-Env antigens and/or a soluble gp140 Env trimer protein (the trimeric form of Env more closely mimics the natural HIV Env protein). In some cases a second mosaic version of the gp140 Env protein122 is also included.

The groundwork for the effort was laid by experiments in the macaque model demonstrating significant protective efficacy against both SIVmac251 and SHIV-SF162P3 challenges.123 The highest degree of protection has been observed in recipients of Ad26 prime followed by Ad26 plus gp140 protein boost; the regimen was associated with a 94% reduction in per-exposure risk of infection, and eight out of a group of 12 macaques (66%) remained uninfected after six SHIV-SF162P3 challenges.124 Correlates of protection included binding antibodies against the Env protein, Env-specific T-cell responses, and functional antibodies capable of inducing antibody-dependent cellular phagocytosis, a process in which antibodies promote the killing of virus-infected cells.125

If all goes according to plan and immune response targets are met in the preparatory studies, a placebo- controlled efficacy trial (HPX2008/HVTN 705) will be launched in late 2017 or early 2018. The aim is to enroll 2,600 sexually active women aged between 18 and 35 at sites in South Africa, Zambia, Zimbabwe, Malawi, and Mozambique. The likely regimen would be the tetravalent Ad26 vector mix administered at months 0, 3, 6, and 12, with soluble gp140 Env trimer protein boosts added at months 6 and 12. The Env protein will be delivered in an alum adjuvant, so the trial may be able to contribute information to the discussion regarding the importance of alum to the protection documented in RV144.

The fate of the diverse collection of other experimental HIV vaccine candidates in the pipeline will almost certainly be significantly influenced by the outcomes of HVTN 702 and the Janssen program. No extant candidate is capable of inducing bNAbs, which remains the holy grail for the vaccine field, and so more information is required regarding the protective potential of non-neutralizing immune responses in order to rationally assess the relative promise of the current crop of contenders. That does not diminish the importance of continuing to develop vaccine candidates in order to have options for future efficacy trials as the science advances. Over the past year, updates have been offered on a variety of approaches, including intranasally administered Sendai virus vectors,126 DNA/MVA regimens127,128,129 (including constructs developed by Geovax designed to encode virus-like particles130), and a NYVAC plus Env protein combination.131 Planning is also underway to conduct a first-in-human trial of a CMV vector,132 which has generated considerable interest due to evidence that it led to clearance of a highly pathogenic SIV when administered prophylactically to macaques.133,134

CONCLUSION

Despite encouraging signs that available prevention options may be diminishing HIV incidence in some areas, the need for increased global access and additional, more user-friendly biomedical prevention tools—particularly an effective vaccine—remains dire. The current pipeline is diverse but heavily dependent on increasingly constrained public and philanthropic funding.

The political climate in the United States, which is by far the largest financer of scientific research, is extremely concerning—the Trump administration has demonstrated a distinct antiscience bent, exemplified by its budget proposals that slash support for the NIH and CDC. The instability of the administration and the countervailing views of many congressional leaders may lessen the likelihood that these cuts will manifest, or at least reduce their severity, but vigilance is essential regarding the potential impact on biomedical HIV prevention research.

RECOMMENDATIONS

  • Research sponsor and investigator adherence to Good Participatory Practice (GPP) guidelines135 is essential in all biomedical prevention trials, particularly in the post-iPrEx era. Gilead ran into extensive pushback after developing the study protocol for the DISCOVER trial without sufficiently engaging community advocates. The trial initially required a 30-day washout period for any interested participant already taking Truvada as PrEP, which raised several ethical red flags for community advocates. Had Gilead worked with an existing trial network with more experience in working with the community, or had they initially engaged the community in a way that was in line with GPP guidelines, several complications could have been avoided.
  • There is an urgent need for researchers, key stakeholders, and community advocates to establish basic ethical standards for the provision of Truvada as PrEP in HIV prevention trials. All parties involved have an obligation to determine the best way to ethically offer PrEP to participants in a way that doesn’t lead to impossibly large clinical efficacy trials for new technologies.
  • Additionally, ethical recruitment guidelines for clinical trials are needed for the post-PrEP era. There are a number of potential recruitment pitfalls that need to be considered; explicitly advertising the possibility of PrEP access in recruitment materials for a randomized controlled trial testing the efficacy of an unproven technology or misrepresenting the trial as a PrEP access study are just a few potentially unethical scenarios that arose with the launch of the DISCOVER trial.
  • Clinical trials continue to underrepresent a number of priority populations, including youth and transgender men and women. In the United States, underrepresentation of people of color is a chronic problem in research. Researchers and funding entities should consistently require plans for recruitment of these key priority populations as part of study protocol or be required to explain why they do not find that specific recruitment is necessary or feasible. Studies should include individuals from priority populations at numbers that allow for the possibility of statistically significant outcomes. Recruiting only a handful of transgender women and then including that population in the title of the study is misleading and inadequate.
  • In anticipation of long-acting injectable technologies, a recent NIH-funded review article looked at what would be necessary to fully implement these new modalities and bring them to scale. This should be standard practice for any prevention technology that seems likely to be approved for broader use; addressing implementation as an afterthought leads to significant delays in access, particularly for marginalized communities that are most in need of new options.
  • As new technologies come closer to market, prices set for novel preventive technologies should be judged not only in terms of potential out-of-pocket costs for key populations, but also by the likely system-wide costs and the anticipated burden on the health care system. Pricing products solely based upon what the market will bear—as Gilead did when it set the price of its hepatitis C cure at $96,000 for a standard course of treatment—forces private and public payers to either explicitly or implicitly ration access via arbitrary restrictions or create unnecessary hurdles. When bringing a product to market, companies should be required to provide a plan for ensuring easy, unfettered universal access, particularly when public funding has gone into any portion of the foundational research.
  • Despite a moderately improved safety profile of F/TAF compared with TDF/FTC, health care providers and community members should be wary of paying higher prices for Descovy as PrEP and of discouraging uptake of potential generic PrEP options. Should Descovy prove to be noninferior as PrEP, it will be of enormous benefit for potential PrEP users with compromised renal function but will not be worth the additional cost for the majority of individuals.

REFERENCES

  1. Song R, Hall IH, Green TA, et al. Using CD4 data to estimate HIV incidence, prevalence, and percent of undiagnosed infections in the United States. J Acquir Immune Defic Syndr. 2017 Jan 1;74(1):3–9.
  2. Okano JT, Robbins D, Palk L, Gerstoft J, Obel N, Blower S. Testing the hypothesis that treatment can eliminate HIV: a nationwide, population-based study of the Danish HIV epidemic in men who have sex with men. Lancet Infect Dis. 2016 Jul;16(7):789–96. doi: 10.1016/S1473-3099(16)30022-6.
  3. Nwokolo N, Whitlock G, McOwan A. Not just PrEP: other reasons for London’s HIV decline. Lancet HIV. 2017 Apr;4(4):e153. doi: 10.1016/S2352-3018(17)30044-9.
  4. Johnson J. Toward health equity. TAGline. 2016 Fall [cited 2017 May 5]. https://www.treatmentactiongroup.org/tagline/2016/fall/toward-health-equity.
  5. Indiana State Department of Health. HIV outbreak in Southeastern Indiana [Internet]. Indianapolis (IN); July 5, 2017 [cited 2016 May 5]. http://www.in.gov/isdh/26649.htm.
  6. UNAIDS. UNAIDS warns that after significant reductions, declines in new HIV infections among adults have stalled and are rising in some regions [Internet]. [updated 2016 Jul 12] [cited 2017 May 5]. http://www.unaids.org/en/resources/presscentre/pressreleaseandstatementarchive/2016/july/20160712_prevention-gap.
  7. Nel A, Kapiga S, Bekker LG, et al. Safety and efficacy of dapivirine vaginal ring for HIV-1 prevention in African women (Abstract 110LB). Paper presented at: 2016 Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA http://www.croiconference.org/sessions/safety-and-efficacy-dapivirine-vaginal-ring-hiv-1-prevention-african-women.
  8. McCoy LE, Burton DR. Identification and specificity of broadly neutralizing antibodies against HIV. Immunol Rev. 2017 Jan;275(1):11–20. doi: 10.1111/imr.12484.
  9. The National Institute of Allergy and Infectious Diseases (NIAID) (Press Release). NIH launches large clinical trials of antibody-based HIV prevention. 2016 April 7. https://www.niaid.nih.gov/news-events/nih-launches-large-clinical-trials-antibody-based-hiv-prevention.
  10. Scheid JF, Mouquet H, Feldhahn N, et al. A method for identification of HIV gp140 binding memory B cells in human blood. J Immunol Methods. 2009 Apr 15;343(2):65–7. doi: 10.1016/j.jim.2008.11.012.
  11. Scheid JF, Mouquet H, Feldhahn N, et al. Broad diversity of neutralizing antibodies isolated from memory B cells in HIV- infected individuals. Nature. 2009 Apr 2;458(7238):636–40. doi: 10.1038/nature07930.
  12. Tiller T, Meffre E, Yurasov S, Tsuiji M, Nussenzweig MC, Wardemann H. Efficient generation of monoclonal antibodies from single human B cells by single cell RT-PCR and expression vector cloning. J Immunol Methods. 2008 Jan 1;329(1- 2):112–24. Epub 2007 Oct 31.
  13. Anderson DJ, Politch JA, Zeitlin L, et al. Systemic and topical use of monoclonal antibodies to prevent the sexual transmission of HIV. AIDS. 2017 May 1. doi: 10.1097/QAD.0000000000001521. [Epub ahead of print]
  14. Derdeyn CA, Moore PL, Morris L. Development of broadly neutralizing antibodies from autologous neutralizing antibody responses in HIV infection. Curr Opin HIV AIDS. 2014 May;9(3):210–6. doi: 10.1097/COH.0000000000000057.
  15. Schief WR. Immunogen design to induce HIV neutralizing antibodies (Abstract 143). Paper presented at: 2017 Conference on Retroviruses and Opportunistic Infections; 2017 February 13–16; Seattle, WA.
  16. 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 Dec 3;361(23):2209–20. doi: 10.1056/NEJMoa0908492.
  17. The National Institute of Allergy and Infectious Diseases (NIAID) (Press Release). First new HIV vaccine efficacy study in seven years has begun. 2016 November 27. https://www.niaid.nih.gov/news-events/first-new-hiv-vaccine-efficacy-study-seven-years-has-begun.
  18. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02842086, Safety and efficacy of emtricitabine and tenofovir alafenamide (F/TAF) fixed-dose combination once daily for pre-exposure prophylaxis in men and transgender women who have sex with men and are at risk of HIV-1 infection (DISCOVER); 2016 July 20 (cited 2017 May 5).  https://clinicaltrials.gov/ct2/show/NCT02842086?term=discover+taf&rank=1.
  19. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02985996, Body Compartment PK for New HIV Pre-exposure Prophylaxis Modalities; 2016 December 5 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT02985996?term=discover+taf&rank=1.
  20. Mugwanya K, Baeten J, Celum C, et al. Low risk of proximal tubular dysfunction associated with emtricitabine-tenofovir disoproxil fumarate pre-exposure prophylaxis in men and women. J Infect Dis. 2016 Mar 29. doi: 10.1093/infdis/ jiw125. [Epub ahead of print]
  21. Gandhi M, Glidden DV, Liu AY, et al. Higher cumulative TFV/FTC levels in PrEP associated with decline in renal function (Abstract 866). Paper presented at: 2016 Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA http://www.croiconference.org/sessions/higher-cumulative-tfvftc-levels-prep-associated-decline-renal-function-0.
  22. Liu AY, Vittinghoff E, Anderson PL, et al. Changes in renal function associated with TDF/FTC PrEP use in the US Demo Project (Abstract 867). Paper presented at: 2016 Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA. http://www.croiconference.org/sessions/changes-renal-function-associated-tdfftc-prep-use-us-demo-project-0.
  23. Liu AY, Vittinghoff E, Sellmeyer DE, et al. Bone mineral density in HIV-negative men participating in a tenofovir pre- exposure prophylaxis randomized clinical trial in San Francisco. PLoS One. 2011;6(8):e23688. doi: 10.1371/journal. pone.0023688.
  24. Mulligan K, Glidden DV, Anderson PL, et al. Preexposure prophylaxis initiative study team. effects of emtricitabine/ tenofovir on bone mineral density in HIV-negative persons in a randomized, double-blind, placebo-controlled trial. Clin Infect Dis. 2015 Aug 15;61(4):572–80. doi: 10.1093/cid/civ324.
  25. Grant R, Mulligan K, McMahan V, et al. Recovery of bone mineral density after stopping oral HIV preexposure prophylaxis (Abstract 48LB). Paper presented at: 2016 Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA. http://www.croiconference.org/sessions/recovery-bone-mineral-density-after-stopping-oral-hiv-preexposure-prophylaxis.
  26. Centers for Disease Control and Prevention (U.S.). Updated guidelines for antiretroviral postexposure prophylaxis after sexual, injection drug use, or other nonoccupational exposure to HIV— United States, 2016. Atlanta: Department of Health and Human Services (U.S.), Centers for Disease Control and Prevention. https://www.cdc.gov/hiv/pdf/programresources/cdc-hiv-npep-guidelines.pdf.
  27. Markowitz M, Zolopa A, Squares K, et al. Phase I/II study of the pharmacokinetics, safety and antiretroviral activity of tenofovir alafenamide, a new prodrug of the HIV reverse transcriptase inhibitor tenofovir, in HIV-infected adults. J Antimicrob Chemother. 2014 May;69(5):1362–9. doi: 10.1093/jac/dkt53.
  28. Ruane P, DeJesus E, Berger D, et al. Antiviral activity, safety, and pharmacokinetics/pharmacodynamics of tenofovir alafenamide as 10-day monotherapy in HIV-1-positive adults. J Acquir Immune Defic Syndr. 2013 Aug 1;63(4):449–55. doi: 10.1097/QAI.0b013e3182965d45.
  29. ClinicalTrials.gov [Internet]. Identifier NCT02842086.
  30. European AIDS Treatment Group. Community Demand for temporary halt to Gilead DISCOVER study [Internet]. Brussels, Belgium. [updated 2016 Nov 16] [cited 2017 May 5]. http://www.eatg.org/news/community-demand-for-temporary-halt-to-gilead-discover-study/.
  31. Gilead Sciences. Statement on DISCOVER Study of F/TAF for PrEP [Internet]. Foster City (CA); [updated 2016 Nov 11] [cited 2017 May 5]. http://www.gilead.com/news/statement%20on%20discover%20study.
  32. POZ. Lawsuit claims Gilead delayed the release of a less toxic version of tenofovir. POZ [Internet]. New York; [updated 2016 May 31] [cited 2017 May 5]. https://www.poz.com/article/lawsuit-claims-gilead-delayed-release-less-toxic-version-hiv-med-tenofovir.
  33. Massud I, Mitchell J, Babusis D, Deyounks F, Ray AS, Rooney JF, Heneine W, Miller MD, García-Lerma JG. Chemoprophylaxis with oral emtricitabine and tenofovir alafenamide combination protects macaques from rectal simian/ human immunodeficiency virus infection. J Infect Dis. 2016 Oct 1;214(7):1058–62. doi: 10.1093/infdis/jiw312.
  34. Garrett KL, Cottrell ML, Prince HM, et al. Concentrations of TFV and TFVdp in female mucosal tissues after a single dose of TAF (Abstract 102LB). Paper presented at: 2016 Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA. http://www.croiconference.org/sessions/concentrations-tfv-and-tfvdp-female-mucosal-tissues-after-single-dose-taf.
  35. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02904369, PK and PD study of Oral F/TAF for HIV prevention; 2016 September 1 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT02904369?term=PK+PD+study+oral+F%2FTAF&rank=1.
  36. Ford N, Irvine C, Shubber Z, et al. Adherence to HIV postexposure prophylaxis: a systematic review and meta-analysis. AIDS. 2014 Nov 28;28(18):2721–7. doi: 10.1097/QAD.0000000000000505.
  37. Hoffman RM, Jaycocks A, Vardavas R, Wagner G, Lake JE, Mindry D, et al. Benefits of PrEP as an Adjunctive Method of Centers for Disease Control and Prevention (U.S.). Updated guidelines for antiretroviral postexposure prophylaxis.
  38. Mayer KH, Mimiaga MJ, Gelman M, Grasso C. Raltegravir, tenofovir DF, and emtricitabine for postexposure prophylaxis to prevent the sexual transmission of HIV: safety, tolerability, and adherence. J Acquir Immune Defic Syndr. 2012 Apr 1;59(4):354–9. doi: 10.1097/QAI.0b013e31824a03b8.
  39. Mcallister J, Towns JM, Mcnulty A, Pierce AB, Foster R, Richardson R, Carr A. Dolutegravir with tenofovir disoproxil fumarate – emtricitabine as HIV post-exposure prophylaxis in gay and bisexual men. AIDS. 2017 Mar 15. doi: 10.1097/ QAD.0000000000001447. [Epub ahead of print]
  40. Valin N, Fonquernie L, Daguenel A, et al. Evaluation of tolerability with the co-formulation elvitegravir, cobicistat, emtricitabine, and tenofovir disoproxil fumarate for post-HIV exposure prophylaxis. BMC Infect Dis. 2016 Nov 29;16(1):718.
  41. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02998320, Evaluation of compliance with treatment by Genvoya in HIV post-exposure prophylaxis; 2016 December 14 (cited 2017 May 5).  https://clinicaltrials.gov/ct2/show/NCT02998320?term=genvoya+post&rank=1.
  42. Drake AL, Wagner A, Richardson B, John-Stewart G. Incident HIV during pregnancy and postpartum and risk of mother- to-child HIV transmission: a systematic review and meta-analysis. PLoS Med. 2014 Feb 25;11(2):e1001608. doi: 10.1371/journal.pmed.1001608.
  43. Hoffman RM, Jaycocks A, Vardavas R, Wagner G, Lake JE, Mindry D, et al. Benefits of PrEP as an Adjunctive Method of HIV Prevention During Attempted Conception Between HIV-uninfected Women and HIV-infected Male Partners. J Infect Dis. 2015 Nov 15;212(10):1534-43. doi: 10.1093/infdis/jiv305.
  44. National Institutes of Health (U.S.). Recommendations for use of antiretroviral drugs in pregnant HIV-1-infected women for maternal health and interventions to reduce perinatal HIV transmission in the United States. Bethesda, MD: Department of Health and Human Services (U.S.), National Institutes of Health. https://aidsinfo.nih.gov/guidelines/htmltables/3/4868.
  45. Mugo NR, Hong T, Celum C, Donnell D, Bukusi EA, John-Stewart G, et al; Partners PrEP Study Team. Pregnancy incidence and outcomes among women receiving preexposure prophylaxis for HIV prevention: a randomized clinical trial. JAMA. 2014 Jul 23-30;312(4):362-71. doi: 10.1001/jama.2014.8735.
  46. Seidman D, Weber S, Oza K, Mullins E, Timoney MT, Wright R (Weber S presenting). Use of HIV pre-exposure prophylaxis during pregnancy and lactation at 2 U.S. centers (Abstract WEPEC195). Paper presented at: 21st International AIDS Conference; 2016 July 18–22; Durban, South Africa. http://programme.aids2016.org/Abstract/Abstract/1957.
  47. Siberry GK, Williams PL, Mendez H, et al. Safety of tenofovir use during pregnancy: early growth outcomes in HIV- exposed uninfected infants. AIDS. 2012 Jun 1;26(9):1151–9. doi: 10.1097/QAD.0b013e328352d135.
  48. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT01209754, EMBRACE (Evaluation of maternal and baby outcome registry after chemoprophylactic exposure); 2010 September 23 (cited 2017 May 5).  https://clinicaltrials.gov/ct2/show/NCT01209754?term=embrace+hiv&rank=1
  49. Pintye J, Drake AL, Kinuthia J, et al. A risk assessment tool for identifying pregnant and postpartum women who may benefit from preexposure prophylaxis. Clin Infect Dis. 2017 Mar 15;64(6):751–8. doi: 10.1093/cid/ciw850.
  50. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT03049176, the impact and cost-effectiveness of safer conception strategies for HIV-discordant couples (SAFER); 2017 February 7 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT03049176?term=impact+cost+safer+conception+discordant&rank=1.
  51. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02998320, Evaluation of compliance with treatment by Genvoya in HIV post-exposure prophylaxis; 2016 December 14 (cited 2017 May 5).  https://clinicaltrials.gov/ct2/show/NCT02998320?term=genvoya+post&rank=1.
  52. Ward H, Rönn M. Contribution of sexually transmitted infections to the sexual transmission of HIV. Curr Opin HIV AIDS. 2010 Jul;5(4):305–10. doi: 10.1097/COH.0b013e32833a8844.
  53. Abara WE, Hess KL, Neblett Fanfair R, Bernstein KT, Paz-Bailey G. Syphilis trends among men who have sex with men in the United States and Western Europe: a systematic review of trend studies published between 2004 and 2015. PLoS One. 2016 Jul 22;11(7):e0159309. doi: 10.1371/journal.pone.0159309.
  54. Bolan RK, Beymer MR, Weiss RE, Flynn RP, Leibowitz AA, Klausner JD. Doxycycline prophylaxis to reduce incident syphilis among HIV-infected men who have sex with men who continue to engage in high-risk sex: a randomized, controlled pilot study. Sex Transm Dis. 2015 Feb;42(2):98–103. doi: 10.1097/OLQ.0000000000000216.
  55. Molina J-M, Charreau I, Chidiac C, et al. On demand post exposure prophylaxis with doxycycline for MSM enrolled in a PrEP trial (Abstract 91LB). Paper presented at: 2017 Conference on Retroviruses and Opportunistic Infections; 2017 February 13–16; Seattle, WA. http://www.croiconference.org/sessions/demand-post-exposure-prophylaxis-doxycycline-msm-enrolled-prep-trial.
  56. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02864550, Oral doxycycline for the prevention of syphilis in men who have sex with men; 2016 August 9 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT02864550?term=oral+doxycycline+prevention+syphilis&rank=1.
  57. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02844634, Tenofovir/emtricitabine with doxycycline for combination HIV and syphilis pre-exposure prophylaxis in HIV-negative MSM (DuDHS); 2016 July 22 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT02844634?term=tenofovir+doxycycline+syphilis&rank=1.
  58. Knox DC, Tan DH, Harrigan PR, Anderson PL. HIV-1 infection with multiclass resistance despite pre-exposure prophylaxis (PrEP) (Abstract 169aLB). Paper presented at: Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA. http://www.croiconference.org/sessions/hiv-1-infection-multiclass-resistance-despite-preexposure-prophylaxis-prep.
  59. Grossman H, Anderson P, Grant R, et al. Newly acquired HIV-1 infection with multi-drug resistant (MDR) HIV-1 in a patient on TDF/FTC-based PrEP (Abstract OA03.06LB). Paper presented at: HIV Research for Prevention (HIVR4P) 2016 Conference; 2016 October 17–21; Chicago, IL.
  60. Hoornenborg E, de Bree GJ. Acute infection with a wild-type HIV-1 virus in a PrEP user with high TDF levels (Abstract 953). Paper presented at: Conference on Retroviruses and Opportunistic Infections; 2017 February 13–16; Seattle, WA. http://www.croiconference.org/sessions/acute-infection-wild-type-hiv-1-virus-prep-user-high-tdf-levels.
  61. Alcorn K. Implants and injectables: PrEP in the future. Aidsmap [Internet]. London; [updated 2017 Jan 13] [cited 2017 May 5]. http://www.aidsmap.com/Implants-and-injectables-PrEP-in-the-future/page/3110737/.
  62. Meyers K, Golub SA. Planning ahead for implementation of long-acting HIV prevention: challenges and opportunities. Curr Opin HIV AIDS. 2015 Jul;10(4):290–5. doi: 10.1097/COH.0000000000000159.
  63. Markowitz M, Frank I, Grant R, et al. ECLAIR: Phase 2A safety and PK study of cabotegravir LA in HIV-uninfected men (Abstract 106). Paper presented at: Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA. http://www.croiconference.org/sessions/%C3%A9clair-phase-2a-safety-and-pk-study-cabotegravir-la-hiv-uninfected-men.
  64. Ford S. ECLAIR study of cabotegravir LA injections: characterization of safety and PK during the “PK tail” phase (Abstract OA12.06LB). Paper presented at: HIV Research for Prevention (HIVR4P) Conference; October 17–21. Chicago, IL.
  65. HIV Prevention Trials Network. HPTN 077: A phase IIa study to evaluate the safety, tolerability and PK of the investigational injectable HIV integrase inhibitor, GSK1265744, in HIV-uninfected men and women version 3.0. [Internet]. 2016 November 7 [cited 2017 May 5]. https://www.hptn.org/sites/default/files/2016-11/HPTN%20077%20LoA%203%20to%20V%203_FINAL_07Nov2016.pdf.
  66. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02720094, Safety and efficacy study of injectable cabotegravir compared to daily oral tenofovir disoproxil fumarate/emtricitabine (TDF/ FTC), for pre-exposure prophylaxis in HIV-uninfected cisgender men and transgender women who have sex with men; 2016 March 21 (cited 2017 May 5).  https://clinicaltrials.gov/ct2/show/NCT02720094?term=hptn+083&rank=1.
  67. HIV Prevention Trials Network. HPTN 084: A phase 3 double blind safety and efficacy study of long-acting injectable cabotegravir compared to daily oral TDF/FTC for pre-exposure prophylaxis in HIV-uninfected women [Internet]. 2017 March 2 (cited 2017 May 5). https://hptn.org/sites/default/files/2017-04/HPTN%20084%20Protocol%20V1.0.pdf.
  68. Jackson AGA, Else LJ, Mesquita PMM, et al. A compartmental pharmacokinetic evaluation of long-acting rilpivirine in HIV-negative volunteers for pre-exposure prophylaxis. Clin Pharmacol Ther. 2014 Sep;96(3):314–23. doi: 10.1038/ clpt.2014.118.
  69. McGowan I, Siegel A, Duffil K, et al. A phase 1 open label safety, acceptability, pharmacokinetic, and pharmacodynamic study of intramuscular TMC278 LA (the MWRI-01 study) (Abstract OA27.06 LB). Paper presented at: HIV Research for Prevention (HIV R4P); 2014 October 28–31; Cape Town, South Africa. http://online.liebertpub.com/doi/full/10.1089/aid.2014.5131a.abstract.
  70. Dezzutti CS, Else LJ, Yandura SE, et al. Distinct pharmacodynamics activity of rilpivirine in mucosal explant tissue (Abstract 874). Paper presented at: 2016 Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA. http://www.croiconference.org/sessions/distinct-pharmacodynamic-activity-rilpivirine-mucosal-explant-tissue.
  71. Bekker, LG, Shuying SL, Tolley B, et al.; HPTN 076. TMC278 LA safe, tolerable, and acceptable for HIV preexposure prophylaxis (Abstract 421LB). Paper presented at: 2017 Conference on Retroviruses and Opportunistic Infections; 2017 February 13–16; Seattle, WA. http://www.croiconference.org/sessions/hptn-076-tmc278-la-safe-tolerable-and-acceptable-hiv-preexposure-prophylaxis.
  72. Alcorn K. Implants and injectables: PrEP in the future [Internet]. London.
  73. Gunawardana M, Remedios-Chan M, Miller CS, et al. Pharmacokinetics of long-acting tenofovir alafenamide (GS- 7340) subdermal implant for HIV prophylaxis. Antimicrob Agents Chemother. 2015 Jul;59(7):3913–9. doi: 10.1128/ AAC.00656-15.
  74. Ibid.
  75. Dunne N. Northwestern receive $17 million grant for HIV prevention research [Internet]. Chicago: Northwestern University, Feinberg School of Medicine; 2015 August 3 (cited 2017 May 5). http://news.feinberg.northwestern.edu/2015/08/17-million-grant-for-hiv-prevention-research/.
  76. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02722343, Exploratory pharmacodynamic study of tenofovir-based products; 2016 March 9 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT02722343?term=exploratory+tenofovir+products&rank=1.
  77. Nel A, Kapiga S, Bekker LG, et al. Safety and efficacy of a dapivirine vaginal ring for HIV prevention in women. N Engl J Med. 2016 Dec 1;375(22):2133–2143.
  78. Baeten JM, Palanee-Phillips T, Brown ER, et al. A phase III trial of the dapivirine vaginal ring for HIV-1 prevention in women (Abstract 109LB). Paper presented at: 2016 Conference on Retroviruses and Opportunistic Infections; 2016 February 22–26; Boston, MA. http://www.croiconference.org/sessions/phase-iii-trial-dapivirine-vaginal-ring-hiv-1-prevention-women.
  79. Baeten JM, Palanee-Phillips T, Brown ER, et al. Use of a vaginal ring containing dapivirine for HIV-1 prevention in women. N Engl J Med. 2016 Feb 22. doi: 10.1056/NEJMoa1506110. [Epub ahead of print]
  80. Brown E, Palanee-Phillips T, Marzinke M, et al. Residual dapivirine ring levels indicate higher adherence to vaginal ring is associated with HIV-1 protection (Abstract TUAC0105LB). Paper presented at: 21st International AIDS Conference; 2016 July 18–22; Durban, South Africa. http://programme.aids2016.org/Abstract/Abstract/10376.
  81. Montgomery ET, van der Straten A, Chitukuta M, et al. Acceptability and use of a dapivirine vaginal ring in a phase III trial. AIDS. 2017 May 15;31(8):1159–67. doi: 10.1097/QAD.0000000000001452.
  82. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02858037, Trial to assess the continued safety of and adherence to a vaginal ring containing dapivirine in women; 2016 July 18 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT02858037?term=trial+assess+continued+adherence+vaginal+ring+dapivirine&rank=2.
  83. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02862171, To assess continued safety of and adherence to the dapivirine (25 mg) vaginal ring-004 in healthy, HIV-negative women; 2016 July 25 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT02862171?term=trial+assess+continued+adherence+vaginal+ring+dapivirine&rank=1.
  84. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02847286, To assess the drug-drug interaction potential between dapivirine vaginal ring-004, containing 25 mg of dapivirine, and clotrimazole 10 mg/g (1%); 2016 July 25 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT02847286?term=clotrimazole+dapivirine&rank=1.
  85. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02808949, Pharmacokinetic study of the dapivirine vaginal ring in lactating women; 2015 Dec 18 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT02808949.
  86. Hillier SL, Meyn LA, Bunge K, et al. Impact of vaginal microbiota on genital tissue and plasma concentrations of tenofovir (Abstract 86LB). Paper presented at: 2017 Conference on Retroviruses and Opportunistic Infections; 2017 February 13–16; Seattle, WA. http://www.croiconference.org/sessions/impact-vaginal-microbiota-genital-tissue-and-plasma-concentrations-tenofovir.
  87. Studies [Internet]. Microbicide Trials Network. [cited 5 May 2017]. http://www.mtnstopshiv.org/studies.
  88. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT03082690, ImQuest (IQP) DuoGel phase 1 pharmacokinetic study; 2017 March 5 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT03082690.
  89. Barton C, Kouokam JC, Hurst H, Palmer KE. Pharmacokinetics of the antiviral lectin griffithsin administered by different routes indicates multiple potential  uses. Viruses. 2016 Dec 17;8(12). pii: E331. doi: 10.3390/v8120331.
  90. P Xiao, S Gumber, M Marzinke, Villinger F, et al. Hypo-osmolar formulation of TFV enemas promotes uptake and transformation of TFV to TFV-DP in tissues and prevents SHIV/SIV infection (Abstract OA04.03). Paper presented at: HIV Research for Prevention (HIVR4P) 2016 Conference; 2016 October 17–21; Chicago, IL.
  91. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (U.S.). 2017. Identifier NCT02750540, Optimization of a tenofovir enema for HIV prevention (DREAM-01); 2016 April 11 (cited 2017 May 5). https://clinicaltrials.gov/ct2/show/NCT02750540?term=dream+enema&rank=1.
  92. Rees H, Delany-Moretlwe S, Lombard C, et al. FACTS 001 phase III trial of pericoital tenofovir 1% gel for HIV prevention in women (Abstract 26LB). Paper presented at: 2015 Conference on Retroviruses and Opportunistic Infections; 2015 February 23–26; Seattle, WA. http://www.croiconference.org/sessions/facts-001-phase-iii-trial-pericoital-tenofovir-1-gel-hiv-prevention-women.
  93. Marrazzo JM, Ramjee G, Richardson BA, et al. Tenofovir-based preexposure prophylaxis for HIV infection among African women. N Engl J Med. 2015 Feb 5;372(6):509–18. doi:10.1056/NEJMoa1402269.
  94. Friedland BA, Hoosley C, Gehret Plagianos M, et al. A first-in-human trial of PC-1005 (MIV-150 and zinc acetate in a carrageenan gel (Abstract 875). Paper presented at: 2016 Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA. http://www.croiconference.org/sessions/first-human-trial-pc-1005-miv-150-and-zinc-acetate-carrageenan-gel.
  95. Villegas G, Zhang S, Mizenina O, et al. CVLs from women vaginally dosed with PC-1005 inhibit mucosal HIV-1 and HSV-2 ex vivo (Abstract 876). Paper presented at: 2016 Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA. http://www.croiconference.org/sessions/cvls-women-vaginally-dosed-pc-1005-inhibit-mucosal-hiv-1-and-hsv-2-ex-vivo.
  96. Friedland BA, Hoesley CJ, Plagianos M, et al. First-in-human trial of MIV-150 and zinc acetate coformulated in a carrageenan gel: safety, pharmacokinetics, acceptability, adherence, and pharmacodynamics. J Acquir Immune Defic Syndr. 2016 Dec 15;73(5):489–96.
  97. Scheid JF, Mouquet H, Ueberheide B, et al. Sequence and structural convergence of broad and potent HIV antibodies that mimic CD4 binding. Science. 2011 Sep 16;333(6049):1633–7. doi: 10.1126/science.1207227.
  98. Mouquet H, Scharf L, Euler Z, et al. Complex-type N-glycan recognition by potent broadly neutralizing HIV antibodies. Proc Natl Acad Sci U S A. 2012 Nov 20;109(47):E3268–77. doi: 10.1073/pnas.1217207109.
  99. Walker LM, Huber M, Doores KJ, et al. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature. 2011 Sep 22;477(7365):466–70. doi: 10.1038/nature10373.
  100. Rudicell RS, Kwon YD, Ko SY, et al. Enhanced potency of a broadly neutralizing HIV-1 antibody in vitro improves protection against lentiviral infection in vivo. J Virol. 2014 Nov;88(21):12669–82. doi: 10.1128/JVI.02213-14.
  101. Schnepp BC, Johnson PR. Adeno-associated virus delivery of broadly neutralizing antibodies. Curr Opin HIV AIDS. 2014 May;9(3):250–6. doi: 10.1097/COH.0000000000000056.
  102. Johnson PR, Schnepp BC, Zhang J, et al. Vector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeys. Nat Med. 2009 Aug;15(8):901–6. doi: 10.1038/nm.1967.
  103. Saunders KO, Wang L, Joyce MG, et al. Broadly neutralizing human immunodeficiency virus type 1 antibody gene transfer protects nonhuman primates from mucosal simian-human immunodeficiency virus infection. J Virol. 2015 Aug;89(16):8334–45. doi: 10.1128/JVI.00908-15.
  104. Martinez-Navio JM, Fuchs SP, Pedreño-López S, Rakasz EG, Gao G, Desrosiers RC. Host anti-antibody responses following adeno-associated virus-mediated delivery of antibodies against HIV and SIV in rhesus monkeys. Mol Ther. 2016 Feb;24(1):76–86. doi: 10.1038/mt.2015.191.
  105. Morris GC, Wiggins RC, Woodhall SC, et al. MABGEL 1: first phase 1 trial of the anti-HIV-1 monoclonal antibodies 2F5, 4E10 and 2G12 as a vaginal microbicide. PLoS One. 2014 Dec 29;9(12):e116153. doi: 10.1371/journal. pone.0116153.
  106. De Logu A, Williamson RA, Rozenshteyn R, et al. Characterization of a type-common human recombinant monoclonal antibody to herpes simplex virus with high therapeutic potential. J Clin Microbiol. 1998 Nov;36(11):3198–204.
  107. Teh AY, Maresch D, Klein K, Ma JK.Characterization of VRC01, a potent and broadly neutralizing anti-HIV mAb, produced in transiently and stably transformed tobacco. Plant Biotechnol J. 2014 Apr;12(3):300–11. doi: 10.1111/ pbi.12137.
  108. Zhao C, Gunawardana M, Villinger F, et al. Pharmacokinetics and preliminary safety of pod-intravaginal rings delivering the monoclonal antibody VRC01-N for HIV prophylaxis in a macaque model. Antimicrob Agents Chemother. 2017 Apr 17. pii: AAC.02465-16. doi: 10.1128/AAC.02465-16. [Epub ahead of print]
  109. Ma JK, Drossard J, Lewis D, et al. Regulatory approval and a first-in-human phase I clinical trial of a monoclonal antibody produced in transgenic tobacco plants. Plant Biotechnol J. 2015 Oct;13(8):1106–20. doi: 10.1111/pbi.12416.
  110. Bekker LG, Gray GE. Hope for HIV control in southern Africa: The continued quest for a vaccine. PLoS Med. 2017 Feb 28;14(2):e1002241. doi: 10.1371/journal.pmed.1002241.
  111. 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 Dec 3;361(23):2209–20. doi: 10.1056/NEJMoa0908492.
  112. Bekker L-G, Laher F, Moodie Z, et al. Meeting the “Go” criteria: immunogenicity from HVTN100, a phase 1–2 randomized, double-blind, placebo-controlled trial of clade C ALVAC-® (vCP2438) and bivalent subtype C gp120/ MF59®in HIV-uninfected South African adults (Abstract TUAX0102LB). Paper presented at: 21st International AIDS Conference; 2016 July 18–22; Durban, South Africa. http://programme.aids2016.org/Abstract/Abstract/10652.
  113. Vaccari M, Gordon SN, Fourati S, et al. Adjuvant-dependent innate and adaptive immune signatures of risk of SIVmac251 acquisition. Nat Med. 2016 Jul;22(7):762–70. doi: 10.1038/nm.4105.
  114. Schifanella L, Venzon D, Barnett S, et al. Immunogenicity and efficacy of ALVAC-HIV/gp120-clade C in alum regimen in macaques (Abstract 317). Paper presented at: 2016 Conference on Retroviruses and Opportunistic Infections; 2016 February 22–25; Boston, MA. http://www.croiconference.org/sessions/immunogenicity-and-efficacy-alvac-hivgp120-clade-c-alum-regimen-macaques.
  115. Barnett SW, Burke B, Sun Y, et al. Antibody-mediated protection against mucosal simian-human immunodeficiency virus challenge of macaques immunized with alphavirus replicon particles and boosted with trimeric envelope glycoprotein in MF59 adjuvant. J Virol. 2010 Jun;84(12):5975–85. doi: 10.1128/JVI.02533-09.
  116. Cohen J. Controversial HIV vaccine strategy gets a second chance. Science. 2016 Nov 4;354(6312):535.
  117. Laher F. Building on (and building!) Success—status of HVTN 702. Presented at: AVAC HIV Vaccine Awareness Day 2017 Webinar Series. 2017 May 8. http://www.avac.org/event/hvad-2017-webinar-series.
  118. Joint United Nations Programme on HIV/AIDS (UNAIDS), World Health Organization (WHO) Geneva: UNAIDS and World Health Organization. 2012 (English original 2007, additional guidance point added 2012). UNAIDS/07.28E/ JC1349E Ethical Considerations in Biomedical HIV Prevention Trials. UNAIDS/WHO Guidance Document. http://www.unaids.org/en/media/unaids/contentassets/documents/unaidspublication/2012/jc1399_ethical_considerations_en.pdf.
  119. Haire B, Folayan MO, Hankins C, et al. Ethical considerations in determining standard of prevention packages for HIV prevention trials: examining PrEP. Dev World Bioeth. 2013 Aug;13(2):87–94. doi: 10.1111/dewb.12032.
  120. Dawson L, Zwerski S. Clinical trial design for HIV prevention research: determining standards of prevention. Bioethics. 2015 Jun;29(5):316–23. doi: 10.1111/bioe.12113.
  121. Sugarman J. Ethical considerations regarding oral preexposure prophylaxis in HIV prevention trials. Curr Opin HIV AIDS. 2016 Jan;11(1):109–15. doi: 10.1097/COH.0000000000000214.
  122. Nkolola JP, Bricault CA, Cheung A, et al. Characterization and immunogenicity of a novel mosaic M HIV-1 gp140 trimer. J Virol. 2014 Sep 1;88(17):9538–52. doi: 10.1128/JVI.01739-14.
  123. Barouch DH, Alter G, Broge T, et al. Protective efficacy of adenovirus/protein vaccines against SIV challenges in rhesus monkeys. Science. 2015 Jul 17;349(6245):320–4. doi: 10.1126/science.aab3886.
  124. Wegmann F, Alter G, Nkolola JP, et al. Protective efficacy of candidate clinical HIV-1 vaccine regimens against SHIV- SF162P3 challenges in rhesus monkeys (Abstract OA11.05). Paper presented at: HIV Research for Prevention (HIVR4P) 2016 Conference; 2016 October 17–21; Chicago, IL.
  125. Pau M. HIV prophylactic vaccine development program. Presented at: AVAC HIV Vaccine Awareness Day 2017 Webinar Series. 2017 April 28. http://www.avac.org/event/hvad-2017-webinar-series.
  126. Nyombayire J, Anzala O, Gazzard B, et al. Recombinant Sendai vaccine delivered mucosally induces gag-specific functional T-cells or antibody responses in prime-boost regimens in humans (Abstract PD01.01). Paper presented at: HIV Research for Prevention (HIVR4P) 2016 Conference; 2016 October 17–21; Chicago, IL.
  127. Joachim A, Nilsson C, Onkar S, et al. Frequent and durable VIV2 antibody responses induced by HIV-1 DNA priming followed by HIV-MVA boosting in healthy Tanzanian volunteers (Abstract PD01.04). Paper presented at: HIV Research for Prevention (HIVR4P) 2016 Conference; 2016 October 17–21; Chicago, IL.
  128. Viegas EO, Missange MT, Nilsson C, et al. Intradermal electroporation of HIV-DNA vaccine followed by HIV-MVA boost with or without addition of GLA adjuvanted gp140 (Abstract PD01.03). Paper presented at: HIV Research for Prevention (HIVR4P) 2016 Conference; 2016 October 17–21; Chicago, IL.
  129. Joseph S, Quinn K, Greenwood A, et al. A comparative phase I study of combination, homologous subtype-C DNA, MVA, and Env gp140 protein/adjuvant HIV vaccines in two immunization regimes. Front Immunol. 2017 Feb 22;8:149. doi: 10.3389/fimmu.2017.00149.
  130. Buchbinder S, Grunenberg N, Sanchez B, et al. DNA/MVA HIV vaccine producing viruslike (VLPs) Is well-tolerated and induces durable functional antibodies (Abs) in HVTN 094 (Abstract OA22.02). Paper presented at: HIV Research for Prevention (HIVR4P) 2016 Conference; 2016 October 17–21; Chicago, IL.
  131. Pantaleo G, Janes H, Tomaras G, et al. Comparing different priming strategies to optimize HIV vaccine antibody responses: results from HVTN 096/EV04 (NCT01799954) (Abstract OA11.06LB). Paper presented at: HIV Research for Prevention (HIVR4P) 2016 Conference; 2016 October 17–21; Chicago, IL.
  132. Terry L. OHSU’s HIV vaccine moves toward clinical trial. The Oregonian [Internet]. 2016 June 1 [cited 2017 May 22]. http://www.oregonlive.com/health/index.ssf/2016/06/ohsus_hiv_vaccine_moves_toward.html.
  133. Hansen SG, Ford JC, Lewis MS, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature. 2011 May 26;473(7348):523–7. doi: 10.1038/nature10003.
  134. Hansen SG, Piatak M Jr, Ventura AB, et al. Immune clearance of highly pathogenic SIV infection. Nature. 2013 Oct 3;502(7469):100–4. doi: 10.1038/nature12519. Erratum in: Nature. 2014 Oct 30;514(7524):654.
  135. Good Participatory Practice (GPP) Guidelines [Internet]. New York: AVAC [cited 2017 May 5]. http://www.avac.org/good-participatory-practice.
Back To Top