National Academies Press: OpenBook
« Previous: Front Matter
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 1
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 2
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 3
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 4
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 5
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 6
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 7
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 8
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 9
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 10
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 11
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 12
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 13
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 14
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 15
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 16
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 17
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 18
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 19
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 20
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 21
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 22
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 23
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 24
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 25
Suggested Citation:"Summary." Institute of Medicine. 2008. Methodological Challenges in Biomedical HIV Prevention Trials. Washington, DC: The National Academies Press. doi: 10.17226/12056.
×
Page 26

Below is the uncorrected machine-read text of this chapter, intended to provide our own search engines and external engines with highly rich, chapter-representative searchable text of each book. Because it is UNCORRECTED material, please consider the following text as a useful but insufficient proxy for the authoritative book pages.

Summary ABSTRACT: This IOM committee was formed at the request of the Bill & Melinda Gates Foundation and charged with addressing methodological challenges in late-stage nonvaccine biomedical HIV prevention trials with a specific focus on microbicide and pre-exposure prophylaxis trials. A near-perfect biomedical intervention for preventing HIV infection is unlikely to be available in the near future. This underscores the need for late-stage clinical trials of biomedical interventions that can detect and quantify modest intervention effects on HIV infection, and adequately evaluate product safety. The committee’s key recommendations for pretrial research and plan- ning include the following. Estimating HIV incidence is critical to determin- ing the size and duration of a late-stage trial and should be based on direct longitudinal follow-up of individuals in the planned trial site(s), and cor- roborated by at least one other source. Investigators should also undertake pretrial assessments of a product’s potential effects on pregnant women and their fetuses to determine circumstances in which women who become pregnant during a trial might continue to use the study product. Investi- gators should place a high priority on developing effective strategies to achieve accrual targets, retain participants, and improve adherence to study products. The committee underscores the need for sponsors to adequately invest in trial site capacity (human, physical, and regulatory) and develop sustainability plans so that a trial site can continue to contribute to the community and other research studies after the trial is completed. Late-stage trials designed to evaluate biomedical interventions should incorporate randomized comparisons of behavioral interventions when pos- 

 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS sible. Other important design recommendations include: using endpoint- driven trials; considering inclusion of both blinded and unblinded control arms in future trials; collecting information for evaluating the effects of biomedical interventions on women who become pregnant during a trial and their fetuses; and selecting methods for evaluating product adherence and risk-taking behavior. Key recommendations for conducting late-stage HIV prevention tri- als include monitoring the evolving results of a trial to ensure that it is maintaining the best interests of participants, adjusting the trial to improve adherence or other aspects of the study protocol, and using safety informa- tion that may become available from external sources. Recommendations for analyzing trial results include using participant adherence in evaluating the relationship between interventions and HIV risk; the practice of excluding results from participants judged to have been already infected at the time of enrollment, and accounting for prod- uct discontinuation due to pregnancy in the analysis of the risk of HIV infection. Finally, in order to enable more efficient evaluations of biomedical interventions, the committee recommends that researchers give priority to developing biomarkers of recent HIV infection which can be used in cross-sectional samples to estimate HIV incidence rates, identifying surro- gate markers for HIV infection and product activity that investigators can reliably use as intermediate trial endpoints, and exploring alternative trial designs that might answer important research questions more efficiently than the traditional two-arm superiority design. I n the more than 25 years of the human immunodeficiency (HIV) epi- demic, significant strides have been made in identifying effective HIV prevention interventions. Early successes included biomedical interven- tions, most notably those that led to dramatic increases in safety of the blood supply (IOM, 1995) and the prevention of mother-to-child trans- mission (The International Perinatal HIV Group, 1999; Bulterys et al., 2004). Some behavioral interventions and voluntary testing and counseling interventions have also been shown to reduce reported sexual and inject- ing risk behaviors and non-HIV sexually transmitted infections (STIs), although none has been shown to reduce HIV infection (Auerbach et al., 2006). Treatment for injecting drug users and programs providing access to sterile injecting equipment can also decrease the risk of HIV infection in drug users (IOM, 2007). Condoms remain a vital prevention technology. When used correctly and consistently, condoms can reduce HIV infection risk by 80–90 percent (Weller and Davis, 2002; Kajubi et al., 2005), though in practice they are often not used to their potential. Most recently, three

SUMMARY  Box S-1 Biomedical Approaches to HIV Prevention Tested in Late-Stage Efficacy Trials Male circumcision, or removal of the penile foreskin, has been shown to reduce the risk of HIV infection in men. Microbicides are topical substances applied to the vagina or rectum that can potentially prevent HIV. Pre-exposure prophylaxis (PrEP), employing antiretroviral drugs used for HIV treatment, may help prevent HIV infection. Cervical barriers were hypothesized to protect women from HIV by covering the cervix and blocking the upper genital tract, which is more vulnerable to HIV infection. Suppression of HSV-2, the primary cause of genital herpes, may help reduce sexual acquisition and transmission of HIV. Vaccines may enhance the body’s immune defenses to prevent HIV infection. Source: Global HIV Prevention Working Group, 2006. randomized, controlled trials found that male circumcision reduced the risk of heterosexually acquired HIV infection among men (Auvert et al., 2005; Bailey et al., 2007; Gray et al., 2007). Yet the epidemic continues to take a terrible toll. With an estimated 2.5 million new HIV infections occurring globally each year (UNAIDS, 2007), efforts are urgently needed to better utilize existing effective HIV prevention strategies and to identify new ones. Because of the large number of women who become infected under circumstances not under their control, the need for additional women-controlled methods of prevention is vital. Researchers are currently testing or have recently evaluated a variety of biomedical HIV prevention interventions in late-stage clinical trials. These interventions include vaginal microbicides, pre-exposure prophylaxis (PrEP) using antiretroviral drugs, suppression of genital herpes (HSV-2) with acyclovir, cervical barriers, male circumcision, and vaccines (see Box S-1). These strategies primarily target sexual transmission of HIV, which accounts for the vast majority of new infections, although PrEP and vac- cines may also help prevent HIV infection in populations at risk through other avenues, such as injecting drug use. Yet many recent trials have had disappointing results. Late-stage trials Late stage trials of behavioral interventions are also underway but are not the focus of this report.   accine trials are not covered in this report. V

 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS have failed to demonstrate a benefit in reducing HIV infection risk, includ- ing a phase 3 trial of the diaphragm and Replens gel (Padian, 2007) and two phase 3 trials of HSV-2 suppression with acyclovir (Watson-Jones et al., 2007; Celum et al., 2008). Two vaginal microbicide trials (of N-9 and cellulose sulfate [CS]) were halted because of evidence that they may have a harmful effect (Van Damme et al., 2002; Van Damme, 2007), and a sepa- rate trial of CS was stopped as a precautionary measure based on evidence from the other CS trial (Cates, 2007). Several PrEP trials were prematurely closed or canceled because of ethical concerns raised by advocates, govern- ments, and community members. One PrEP trial and two microbicide trials in (Savvy in Ghana and Nigeria) did not have sufficient power to determine efficacy of the intervention (Peterson et al., 2007a,b). In addition, two trials of an HIV vaccine were recently stopped based on a lack of evidence of ben- efit and concerns that the vaccine might also increase HIV infection risk. It was in this context that the Bill & Melinda Gates Foundation asked the Institute of Medicine (IOM) in November 2006 to convene a commit- tee to examine the methodological challenges facing late-stage biomedical HIV prevention trials (see Box S-2 for the Statement of Task). The sponsor clarified that the committee’s review should cover late-stage nonvaccine biomedical HIV prevention trials, with a focus on microbicide and PrEP trials. The committee did not review vaccine or mother-to-child transmis- sion trials. However, the committee did consider the role of risk reduction counseling in biomedical prevention trials (see Chapter 3). This report recommends a number of ways to improve the design, monitoring, and analysis of late-stage randomized clinical trials that evalu- ate nonvaccine biomedical interventions to prevent HIV infection. The goals are to increase the chances that these trials will detect a beneficial intervention effect and better quantify the effect size, to more fully assess the effects of using an intervention on behavior and how this and product adherence might influence effectiveness in preventing HIV infection, and to reduce biases that can lead to false positive trial results. Another goal is to allow early termination of these trials, if warranted by their interim results or external information. Below the committee highlights its key recommendations (see Box S-3 at the end of the chapter for a complete list). Where possible, the com- mittee recommends investigators consider alternative trial designs which   he T STEP study was discontinued based on recommendations made by a Data and Safety Monitoring Board, which concluded that the vaccine neither prevented HIV infection nor reduced the amount of virus in those who became infected with HIV (http://www.avac.org/ pdf/STEP_data_release.7Nov.pdf), and possibly might have increased the risk of HIV infec- tion. Based on review of the STEP data, the Phambili study in South Africa was also stopped (http://www.hvtn.org/media/pr/PhambiliSAAVIstatement.pdf).

SUMMARY  Box S-2 Statement of Task The Institute of Medicine (IOM) will convene a committee to examine meth- odological challenges in HIV prevention trials. The committee will prepare a report to improve the methodology, design, and conduct of HIV prevention trials, focusing on microbicide and pre-exposure prophylaxis (PrEP) trials, in order to increase their likelihood of success and to enable donors to optimally invest resources. The committee will undertake a study with the following tasks: 1. The committee will review select phase 2 and 3 HIV prevention trials in order to provide an assessment of best practices for site preparedness and esti- mation of incidence. 2. The committee will make recommendations regarding methodological best practices for microbicide and PrEP efficacy trials. Issues to be addressed include but are not limited to: loss of study power through lower-than-expected incidence and high pregnancy rates; other design considerations such as choice of endpoints and control groups; methods for monitoring the interim results of trials (including adjustments to trial size/duration); pooling of data from trials testing the same product; methods for improving adherence to study regimens and the quality of self-reported behavioral data; and optimizing retention of trial participants. The committee will also consider the ethical issues directly related to methodological issues under study, such as those that might arise during interim monitoring of trials. This study will not address broader ethical issues such as adequacy of informed consent, compensation for trial-related adverse events, access to HIV treatment for seroconverters, and best practices for engaging community members. could offer potential advantages over the traditional two-arm superiority design. Efficacy versus Effectiveness Trials and Lack of a Reliable Surrogate Marker An initial consideration when designing a clinical trial is whether the goal is to assess efficacy—whether a product works in a tightly controlled setting—or effectiveness: whether a product works in the real world. For HIV prevention trials, in which investigators can only partially control participants’ adherence to the product regimen and risk-taking behav- ior, this distinction between efficacy and effectiveness can be substantial (Chapter 2).

 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS Effectiveness trials have historically measured disease outcomes, such as clinical improvement or survival. In contrast, efficacy trials often use intermediate, or “surrogate,” endpoints rather than clinical outcomes—if those surrogates are sufficiently predictive of the clinical endpoint, and if the effect of the interventions on the surrogate predicts its effect on the clinical response (see, for example, Prentice, 1989). This allows investiga- tors to assess interventions in much less time and/or with fewer subjects. For example, HIV treatment trials use viral suppression as a surrogate marker for clinical progression. Both efficacy and effectiveness trials for biomedical prevention inter- ventions must use HIV infection as the primary endpoint, as no reliable marker is available to serve as a surrogate endpoint. This slows research considerably. Moreover, because HIV infection is a relatively uncommon event (compared with other disease outcomes), even in areas with high HIV incidence rates, short-term HIV prevention efficacy trials often need to enroll large numbers of subjects, just as longer-term effective- ness trials do. Late-stage effectiveness trials that evaluate HIV infection offer the opportunity to evaluate potential surrogate markers for HIV infection, and the committee believes that this is a worthwhile secondary goal for these studies. The choice of candidate surrogates must be securely anchored in the knowledge of the pathophysiology of infection, and how the surrogate marker relates biologically to HIV infection. In addition to increasing the time and resources needed to evaluate a new nonvaccine HIV intervention, the lack of a surrogate marker raises another complication. If an efficacy trial demonstrates a reduction in the short-term risk of becoming HIV infected, it may be difficult to ethically justify conducting a subsequent longer-term effectiveness trial that uses a placebo group, even if there remain uncertainties about the ability of the intervention to confer a longer-term protective effect. Similar ethical con- cerns may affect Phase 2B trials, which follow subjects for similar durations as phase 3 effectiveness trials, but aim to save funds by enrolling fewer subjects, and then conducting a longer trial only if the Phase 2B trial results are sufficiently promising. This situation has led the committee to recommend the following: • Although such research is challenging, priority should be given to identifying and validating surrogate endpoints for HIV infection for use in late-stage trials of nonvaccine biomedical interventions. • Until a surrogate endpoint is identified, modified trial designs should be used to provide information on both the short- and longer-term benefits of an intervention. In particular, investigators should consider greater use

SUMMARY  of two modified designs—an efficacy study with extended follow-up and a phase 3 trial with stopping rules for futility. The first modified trial design can allow investigators to obtain some information on longer-term effectiveness in an efficacy trial whose main goal is to assess short-term efficacy. The second modified trial design can allow investigators to terminate a longer-term effectiveness trial if an interim anal- ysis shows insufficient evidence of short-term efficacy. The rationale behind these designs is that an HIV prevention product that has efficacy might not be effective in a real-world setting, and that a product that does not have efficacy would likely not be effective in a real-world setting (Chapter 2). Estimating Power and Sample Size Several factors can adversely affect a trial’s power if investigators do not adequately account for them when calculating a trial’s sample size and duration before the study starts. These include HIV incidence, participant attrition, and the number of participants who discontinue using the study product because of pregnancy or other reasons. Although the committee emphasizes the need for accurate a priori estimates of these factors in cal- culating the required sample size for a late-stage trial, the committee also realizes that such estimates can be imprecise for a variety of reasons, includ- ing both random sampling error and systematic sources of bias. As a guard against inaccurate estimates, the committee recommends the use of “events- driven” trial designs, which follow participants until a prespecified number of subjects become HIV infected, rather than for a prespecified period of time (Chapter 2). Choice of Control Group In most randomized trials that test whether a new intervention is supe- rior to the current standard, a double-blind design is highly desirable to help ensure an unbiased evaluation of the relative effect of the intervention. However, the use of a blinded control group in late-stage HIV prevention trials of a biomedical intervention can be disadvantageous if a participant’s knowledge of his/her intervention would affect that person’s risk-taking behavior. In that case, the relative effectiveness observed in the trial might not reflect that seen when the intervention is introduced into the commu- nity. Trials that include unblinded arms that more closely mimic the real world could potentially provide more useful results. The committee believes that both blinded and unblinded control arms can provide useful informa- tion on the effects of an intervention on risk-taking behavior and the risk of

 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS HIV infection. Investigators and donors should consider conducting trials that include both control groups (Chapter 2). In some instances, there can be value in identifying alternative interven- tions that are believed to have similar effectiveness to more standard inter- ventions (such as risk reduction counseling and condom use), but which have other advantages. These advantages could include reduced cost, fewer side effects, or the personal preference of the user. For example, women who find negotiating condom use difficult might prefer microbicides or PrEP. For these settings, noninferiority (or “equivalence”) designs, which aim to identify interventions of similar efficacy rather than aiming to show that one is superior, could be useful (Chapter 10). Evaluating and Integrating Behavioral RISK-REDUCTION Strategies INTO BIOMEDICAL PREVENTION TRIALS In discussions over the past decade about the ethics of vaccine trials, researchers, community representatives, human rights advocates, and ethi- cists reached broad agreement—based on several ethical principles, includ- ing beneficence, autonomy, and justice—that participants in clinical trials of HIV prevention interventions should receive risk-reduction counseling, and access to condoms and other means to reduce their risk of becoming infected with HIV (UNAIDS, 2000). Despite this widespread agreement, considerable uncertainty remains about what the nature and intensity of such interventions should be. Uncer- tainty about the appropriate prevention standard in biomedical HIV pre- vention trials stems in part from ethical considerations. For example, should the standard risk-reduction intervention be the one shown to be most effective, regardless of cost or sustainability? The ethical uncertainties are compounded by the lack of definitive findings on the effectiveness of behav- ioral risk-reduction interventions in many of the resource-poor settings where biomedical HIV prevention trials are conducted. That knowledge gap reflects the fact that studies of behavioral risk-reduction interventions have largely been conducted in the United States, and from the difficulty of extrapolating behavioral risk-reduction interventions shown to be effica- cious in one setting and population to settings with different populations, risk behaviors, and sociocultural norms. Finally, although some behavioral risk-reduction interventions have been shown to decrease self-reported risk behaviors, and a few have shown decreases in STIs, none to date have been shown to significantly reduce HIV infection rates. Effective behavioral interventions increase the effectiveness of bio- medical interventions and are valuable in their own right. Thus, in light of the uncertainties about the effectiveness of behavioral risk reduction

SUMMARY  interventions in settings where many biomedical trials are being planned, the committee believes that investigators designing biomedical interven- tion trials should also incorporate randomized comparisons of behavioral interventions into the trials whenever possible (Chapter 3). While doing so would increase the logistical complexity of a site’s responsibilities, finding improved behavioral interventions for reducing HIV risk would provide lasting benefits to the community. One methodological approach to achiev- ing this is to use a partially blinded factorial design (see Chapter 10). Such a design can assess both the relative efficacy of a new biomedical interven- tion and the comparative effectiveness of different behavioral interventions without an increase in sample size. Other types of trial designs can also make an important impact on the HIV epidemic by attempting to identify ways of using a variety of partially effective interventions more efficiently. These include noninferiority trials, trials utilizing HIV discordant couples, and dynamic designs (which aim to evaluate strategies for using and modifying different combinations of behavioral and biomedical interventions over time) (see Chapter 10). In addition, investigators should involve behavioral and social scien- tists, the community, and other stakeholders, in the early planning stages of a trial, to identify the most appropriate and sustainable behavioral risk- reduction interventions for use in that community, and to most efficiently plan their implementation. If a trial will adapt specific interventions shown to be effective in other settings, investigators should field-test the strategies during the planning of the trial, to ensure that they can be implemented as envisioned. PREGNANCY Many late-stage biomedical HIV prevention trials are conducted among sexually active women of reproductive age in areas with high fertility rates. Despite intensive counseling on family planning, and provision of or access to contraceptives, a large percentage of women enrolled in biomedical HIV prevention trials become pregnant. Trials testing new products and devices (or new indications of existing drugs) typically restrict pregnant women from enrolling and take women who become pregnant during the trial off the product, either permanently or for the duration of their pregnancy, based on concerns about its potential effect on the pregnant woman and the fetus. If a study discontinues product use among participants who become pregnant and no longer follows them for HIV infection, it can lose statisti- cal power. That is, it will be less able to detect any effect of the biomedical intervention, because of the reduced number of women-years of observa- tion. Failing to follow a woman who becomes pregnant for HIV infection can also bias the analyses of a trial’s results. Thus the committee emphasizes

10 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS the importance of continuing to follow pregnant women for HIV infection regardless of whether they discontinue use of the product, and using this information in the analysis of trial results. This will minimize bias, yet such discontinuations will reduce study power. Thus, the committee also recommends ways of calculating the required sample size and duration of a trial to adjust for the anticipated loss of power when an intervention will be discontinued upon pregnancy. An even greater concern is that trials that discontinue product use in women who become pregnant typically do not provide any information about the safety and efficacy of the product for pregnant women and their fetuses. This is important because if the intervention were introduced into the community, many women would continue to use it after becoming pregnant, despite any cautions about its unknown effects on pregnancy. Assessing safety and efficacy in pregnant women after completing a trial is challenging. Because pregnancy is a common occurrence among women who would use a biomedical HIV intervention, it is critical that an overall product evaluation plan include specific and realistic plans for assessing the intervention’s impact on pregnant women and their fetuses. Because of the difficulty of obtaining such information after a success- ful trial, the committee finds that the current “one size fits all” policy of discontinuing product use upon pregnancy is unnecessary and potentially counterproductive. The committee suggests specific circumstances in which it might be ethical to allow trial participants who become pregnant to con- tinue to use the study product. The committee also recommends that trials collect and analyze infor- mation on pregnancy outcomes on all women who become pregnant during a trial, regardless of their study arm or whether they discontinue product use, as this will provide preliminary information on the possible effects of the product on the fetus. The committee further recommends that investigators specify in advance of a late-stage clinical trial how they will establish product safety and effi- cacy for pregnant women and their fetuses, based on information collected before, during, and after the trial. Investigators should complete reproduc- tive toxicity and pharmacokinetic studies in animals—ideally before the start of phase 2 clinical trials, but no later than the start of phase 2B/3 trials. The study protocol should specify how investigators will collect and monitor information on pregnancy outcomes during the trial, and indicate activities that they will undertake if the trial demonstrates that the product is effective in preventing HIV infection (Chapter 4).

SUMMARY 11 PRODUCT ADHERENCE AND RISK-TAKING BEHAVIOR The ultimate effectiveness of a biomedical intervention is mediated by how well participants adhere to the regimen for using it, and by their risk-taking behavior during the trial. For example, if a trial shows that a product provides an overall benefit, being able to relate the level of protec- tion to the level of adherence could be very useful in interpreting the results. Similarly, if a trial fails to show a protective effect, it would be valuable to distinguish the extent to which the product was not biologically effica- cious, participants did not use it as directed, or they engaged in more risky behavior because they thought the product was protecting them. Although researchers agree on the importance of product adherence in both research and real-world settings, there is less agreement on how to define, measure, improve, and analyze it. Clinical trials often report adher- ence by a single number, such as the percentage of coital acts in which participants use a gel, or the percentage of pills they take over a given time period (Chesney, 2006). While simple, use of such measures to define adherence may mask crucial insights into adherence problems, product acceptability, and potential areas for intervention (Kerr et al., 2005; Berg and Arnsten, 2006). Because understanding these patterns can be critical to identifying and ameliorating problems with product use, investigators should develop, evaluate, and use adherence measures that can capture dif- ferent adherence patterns over time. Investigators can gather information on product adherence and risk behavior through a variety of measures. Indirect measures include self- reports, pill counts, electronic product monitoring, pharmacy refills, and biomarkers of product exposure and risk behavior. Direct measures of prod- uct adherence include pharmacokinetic studies (which measure drug levels or metabolites in subjects’ blood or bodily fluids), and directly observed therapy. These measures vary substantially in expense, the effort required of participants and their partners, their perceived invasiveness, and their accuracy and reliability (Berg and Arnsten, 2006). Several studies have found that using multiple measures to “triangu- late” adherence levels and risk behaviors is helpful in reducing the error introduced by any particular method (Liu et al., 2001; Pool, 2006). The committee endorses this approach. Rather than collecting detailed informa- tion on all participants, investigators could collect such information on a well-chosen random sample, and collect less detailed information on other participants. While directly observed therapy (DOT) or modified DOT could be very useful for proof-of-concept trials, investigators should not use these approaches in effectiveness trials if that approach will not work in real-world practice because the trial results may be poor predictors of the effectiveness of an intervention.

12 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS Little empirical evidence exists on the effectiveness of strategies to improve adherence to nonvaccine biomedical HIV prevention interventions. Methods to improve the adherence of HIV-infected patients to antiretrovi- ral therapy (ART) can inform efforts to enhance adherence in biomedical HIV prevention trials. Experience with ART may have particular relevance for medication-based HIV prevention strategies, such as PrEP or acyclo- vir for HSV-2 suppression. Recent meta-analyses of strategies to improve ART adherence found that such interventions can have a positive impact on adherence, but these analyses offer few prescriptive guidelines about the specific intervention components that are most effective for which populations in which circumstances. Thus the committee recommends that investigators undertake empirical evaluations of strategies to increase adherence during and after HIV prevention trials. These evaluations should be adequately powered, methodologically rigorous, socially and culturally relevant, and grounded in behavioral and social theories. Additional evi- dence is needed in particular about the effectiveness of adherence strategies in resource-poor areas where many biomedical HIV prevention trials are conducted. Investigators should analyze adherence and behavior as both outcomes in an HIV prevention trial and modifiers of the effect of the bio- medical intervention on HIV infection risk. Investigators should also specify in the protocols how they plan to measure, monitor, improve, and analyze adherence (Chapter 5). The availability of reliable information on product adherence and sex- ual behavior is critical to developing “dynamic” HIV intervention strategies consisting of decision rules for how to vary interventions for individuals over time according to measurements of adherence and behavior for each individual (Chapter 10). The committee believes that collecting reliable information on adherence and risk-taking behavior is critical to under- standing how best to utilize biomedical HIV interventions. RECRUITMENT AND RETENTION Late-stage biomedical HIV prevention trials typically require investi- gators to enroll 1,000–4,000 participants at one or multiple sites, and to follow them for several years. A lower-than-expected rate of enrollment can result in an underpowered trial that fails to reveal an effective intervention, or delay the public health impact of a positive trial. Retention is equally critical because loss of trial participants to follow-up reduces study power and can lead to biased results. Despite the critical threat that inadequate recruitment and retention pose to trial validity, there is very little empirical evidence about the effectiveness of alternative recruitment and retention strategies (Lovato et al., 1997; Robinson et al., 2007; Villacorta et al.,

SUMMARY 13 2007). Most of the strategies identified in the literature are based on practi- cal “lessons learned” from investigators in the field. Investigators and sponsors should anticipate that maintaining timely accrual and high retention rates will be labor intensive and costly through- out a trial. However, these investments are necessary to maintain internal and external validity. Successful recruitment and retention will require the use of multiple strategies and incentives, the ability to rapidly change procedures when they are not working, and persistence and innovation among staff. Because of the loss in study power that can result from inadequate accrual and because the potential biases resulting from losses to follow-up cannot be avoided simply by increasing sample size, investigators should place a high priority on developing effective strategies to achieve accrual rate goals and to minimize losses to follow-up. To this end, the commit- tee recommends that investigators conduct pretrial research to assess the community’s and individuals’ interest in the trial (including motivations and barriers to participating), to pilot test recruitment and retention strat- egies and procedures, and to set realistic timeline and resource needs for the enrollment period and retention. The committee also recommends that investigators develop a detailed and multifaceted plan for retaining enrolled participants before beginning a study, for monitoring retention during the conduct of the trial if retention rates are lower than anticipated. Investiga- tors should collect detailed tracking information on all participants early in the process and should develop strategies to engage, train, and reward staff for building trust and accountability with participants and within the community and for meeting recruitment and retention targets. SITE PREPAREDNESS The HIV prevention research agenda requires access to large study populations in settings with the capacity to conduct a wide range of clini- cal trials. The regions with the greatest need for HIV prevention efforts are often those with limited medical and research infrastructure. When developing the research protocol for a late-stage trial, and when selecting and preparing a trial site, external investigators should develop equal part- nerships with local investigators and to involve community representatives in developing the protocol and throughout the trial. Investigators also may need to conduct extensive pretrial research at the study sites, with popu- lations similar to those that the main trial will enroll, to ensure that the interventions are culturally relevant, and to estimate the anticipated rates of HIV infection, attrition, pregnancy, and adherence for the trial site. Donors need to invest adequate resources to developing the infrastructure for trial sites, especially in terms of human resources (Chapter 7).

14 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS The time and resources required to build a relationship with local investigators and local communities, conduct pretrial research, and estab- lish the infrastructure needed to conduct a late-stage trial are considerable. However, such efforts are critical to ensuring high-quality research and the ultimate value to the community of trial results. Investigators and donors establishing new research sites should work with the community to create a sustainability plan that will allow a site to continue to perform research after a trial closes, to enable the community to reap long-term benefit. ESTIMATING HIV INCIDENCE One of the most important aspects of planning a late-stage HIV preven- tion trial is accurately estimating the HIV incidence rate in the trial popula- tion. Because studies are powered based on the number of HIV infections investigators expect to occur among participants during the trial, modest overestimates of HIV incidence can substantially reduce the power of a trial to detect an important intervention effect. Overestimates of HIV incidence have led to premature closure of several recent trials. Three general approaches are available for estimating HIV incidence, each with its strengths and weaknesses: longitudinal cohort studies, labo- ratory biomarkers to identify recent infections, and mathematical model- ing of serial prevalence data (Chapter 8). The committee finds that direct longitudinal follow-up of individuals, through cohort studies, provides the most unbiased estimate of HIV incidence, compared with the indirect approaches currently available. However, longitudinal cohort studies have several drawbacks including the time and expense to conduct them, and the lack of precision in the resulting estimates of HIV incidence rates. Thus the committee recommends that investigators rely on at least one direct longitudinal follow-up study of individuals in the trial setting to estimate HIV incidence, and that they use at least one other independent source to corroborate that estimate. The committee further emphasizes that the development of a reliable, accurate biomarker-based test for recent HIV infection that can be run with blood from a single draw would be a major advance in estimating incidence. Donors and appropriate U.S. and international agencies should make such research a high priority. While current approaches, based on the Serologic Testing Algorithm for Determin- ing HIV Seroconversion (STAHRS) or BED capture enzyme immunoassay (BED-CEIA), are promising, further validation studies are needed to address concerns that these tests may produce biased estimates of HIV incidence. As a result, the committee recommends that investigators should not now rely solely on these biomarker assays of recent infection to estimate HIV incidence for the purpose of designing a prevention trial.

SUMMARY 15 INTERIM MONITORING AND ANALYSIS OF TRIAL RESULTS Because late-stage HIV prevention trials can require several years to complete, it is ethically important and scientifically valuable to monitor a trial’s evolving results. An independent data monitoring committee (DMC) typically performs such monitoring. The DMC evaluates (1) whether the key assumptions underlying the trial’s size and duration are consistent with the evolving data, (2) whether interim results on the efficacy of the interven- tion warrant early termination of the trial, (3) whether any unanticipated safety concerns have arisen, and (4) whether the emergence of any external information requires modifying or terminating the trial. The committee makes several recommendations regarding the role of DMCs in HIV prevention trials (Chapter 5). First, because of differing social and cultural norms, it is important that the composition of DMCs include appropriate expertise and representation from participating coun- tries and regions, and that they meet often enough to ensure that the trial is maintaining the best interests of the participants. Second, DMCs should always have the option of unblinding the interim results of a trial if, for any reason, the DMC believes that doing so is in the best interests of the trial participants. Third, in addition to criteria for a DMC recommendation to terminate a trial based on safety concerns or early indications that the inter- vention is effective, late-stage HIV prevention trials should include criteria for termination owing to futility—that is, on the grounds that, if continued, there would be a very small chance of demonstrating benefit. Finally, the committee believes that researchers should develop method- ologies to enable DMCs of simultaneous HIV prevention trials with common intervention arms—that is, those that evaluate the same intervention—to share information on safety. Although concurrent trials provide a valuable opportunity to learn more about a product’s safety, the valid and effective use of such information poses a number of challenges. The committee discusses methods used to analyze the results of preven- tion trials using HIV infection as the primary efficacy endpoint, along with their limitations. A complication in such trials results from the fact that HIV infection is a silent event: at best, investigators can determine only a time interval in which an individual’s infection occurred. The tests used to diagnose HIV infection are also imperfect, leading to the possibility that trials might unknowingly enroll individuals already infected and might not detect enrollees who become infected during the trial. These features have implications for the analysis of trial results. These include the post hoc exclusion of enrollees who are suspected of having been HIV infected at enrollment, the importance of designing follow-up so infected subjects in each study arm have equal likelihood of being detected at each clinic visit, and the potential biases from censoring analyses of time

16 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS to HIV infection upon product discontinuation owing to pregnancy or other reasons (Chapter 9). In sum, the committee concludes that alternative trial designs, more extensive site preparation, and careful monitoring and analysis of trial results are key to evaluating prevention interventions and determining which of them can exert the greatest possible long-term impact on the HIV epidemic.

SUMMARY 17 Box S-3 Findings and Recommendations Chapter 2 Basic Design Features: Size, Duration, and Type of Trials, and Choice of Control Group Recommendation 2-1: Investigators should take steps to develop accurate a priori estimates of rates of participant accrual, HIV incidence, product discon- tinuation, and participant retention, and incorporate those into the sample size calculations. As a guard against inaccurate estimates, investigators should consider using an “events-driven” approach, by analyzing study results when the prespecified number of enrolled subjects has become HIV infected, rather than at prespecified calendar times. Recommendation 2-2: Until validated surrogate endpoint(s) for HIV infection or product activity is (are) identified, investigators should use modified trial designs that can provide information on both the short- and long-term benefits of an intervention. Recommendation 2-3: Sponsors, investigators, and regulatory agencies should consider using both blinded and unblinded control groups in future trials to more fully understand the effects of the intervention on HIV infection risk and behavior. Chapter 3 Design Considerations: Risk-Reduction Counseling Recommendation 3-1: Given the lack of evidence on the effectiveness of behav- ioral risk-reduction interventions in settings where many HIV biomedical trials are planned, investigators planning such trials should incorporate randomized comparisons of behavioral risk-reduction interventions into their designs whenever possible. Recommendation 3-2: Donors and investigators should involve behavioral and so- cial scientists in the early planning stages of a trial, to identify the most appropriate behavioral risk reduction interventions, and to efficiently plan their implementation during the trial. Recommendation 3-3: Investigators planning to test behavioral risk-reduction in- terventions as part of a late-stage biomedical HIV prevention trial should consult with the community, governments, donors, and other stakeholders about the cost and sustainability of those interventions in the community. Recommendation 3-4: If a trial will adapt specific behavioral interventions shown to be effective in other settings, investigators should field-test the strategies during the planning of the trial, to ensure that they can be implemented as envisioned. Continued

18 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS Box S-3  Continued Chapter 4 Design Considerations: Pregnancy Recommendation 4-1: Investigators should take several steps to minimize the loss of study power and potential biases in results that can occur when women become pregnant during a trial: • Before the start of the trial, investigators should attempt to accurately es- timate the rate of pregnancy that will occur during participant follow-up, and use these estimates in calculating sample size and trial duration. • Data monitoring committees should monitor actual pregnancy rates during the trial, and recommend appropriate adjustments to sample size and trial duration if these rates exceed expectations. • Investigators should continue to follow all women who become pregnant for HIV infection, regardless of whether they discontinue the study product. Recommendation 4-2: Although the current policy of excluding pregnant women from biomedical HIV prevention and other trials stems from an historically protec- tionist orientation adopted by regulators, the principles of research ethics neither mandate nor preclude use of the product by pregnant women. Because any ap- proved product subsequently would likely be used by many women who become pregnant, sponsors and investigators of a biomedical intervention should specify in advance of any late-stage trial how they will establish its safety and efficacy for pregnant women and their fetuses, based on information collected both during and after clinical trials. At a minimum, investigators should take the following steps to collect such information. • Investigators should conduct appropriate preclinical tests in animals, includ- ing reproductive toxicity and pharmacokinetic studies, to allow a more informed decision on whether to continue product use in pregnant women participating in late-stage trials. These tests would ideally be completed before the product or device enters phase 2 testing, but should be completed no later than phase 3 testing. • Investigators should routinely collect and analyze information about birth outcomes from women who become pregnant during a trial, regardless of whether a product is discontinued upon detection of pregnancy. • In trials that discontinue the use of a product by women who become preg- nant, investigators should allow women who are no longer pregnant to have the choice of resuming the study medication. • Investigators should conduct observational or randomized studies in preg- nant women in the postapproval, premarketing, and posttrial periods, to provide additional information on the safety and efficacy of biomedical HIV prevention interventions for pregnant women. Recommendation 4-3: Regulators, sponsors, and investigators should evaluate the strength of the evidence on the beneficial and harmful effects to both a preg-

SUMMARY 19 Box S-3  Continued nant woman and her fetus on a product-by-product basis, and evaluate whether there are circumstances in which women who become pregnant can continue to receive the study product, based on what is known about its benefits and risks. Recommendation 4-4: Trials using products with favorable risk-benefit profiles, but which are nonetheless discontinued upon pregnancy, should monitor pregnancy outcomes during the interim analysis of trial results, as this information might alter the risk-benefit profile to allow continuation of the product during pregnancy. Such trials might be modified to thereafter allow women who become pregnant to remain on product or offer them the opportunity to be randomized to remain on product versus to discontinue product. Recommendation 4-5: Regulatory agencies and institutional review boards (IRBs) should receive periodic safety updates during a trial that include experience with the product during pregnancy. When interim analyses provide evidence of fetal safety and potential benefit to women, regulators and IRBs should consider al- lowing women to stay on product while pregnant. Chapter 5 Design Considerations: Adherence Recommendation 5-1: Because simple measures of adherence can mask sub- stantially different underlying adherence problems, investigators should develop and use adherence measures that can capture different adherence patterns over time. Recommendation 5-2: In light of the uncertainty about the accuracy of various methods for collecting data on adherence and risk behavior, investigators of bio- medical HIV prevention trials should strive to use multiple types of measures to triangulate adherence estimates. Rather than collecting detailed information on all participants, investigators could collect more detailed information on a well-chosen random sample, and collect less detailed information on all participants. Recommendation 5-3: Although directly observed therapy (DOT) or modified DOT could be very useful in proof-of-concept trials, investigators should not use these methods in effectiveness trials if that approach will not be used in real-world practice, because the trial results may then be poor predictors of the effectiveness of the interventions. Recommendation 5-4: Donors should fund and investigators should undertake empirical evaluations of strategies to increase adherence to biomedical HIV pre- vention products during and after a clinical trial. These evaluations should be adequately powered, methodologically rigorous, socially and culturally relevant, grounded in behavioral and social science theories, and conducted in the regions where the strategies will be utilized. Continued

20 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS Box S-3  Continued Recommendation 5-5: Investigators should specify in the study protocol detailed plans for monitoring, measuring, and analyzing adherence data, and steps they will take to improve adherence if it is poorer than anticipated. Recommendation 5-6: Investigators should provide data on product adherence and risk behavior results to the data monitoring committee, as this information may influence the committee’s views of the relative efficacy and safety of the study arms, and the feasibility of the study. Recommendation 5-7: Investigators should analyze adherence and behavior as both outcomes in an HIV prevention trial and modifiers of the effect of the biomedi- cal intervention on HIV infection risk. Recommendation 5-8: Investigators should analyze the potential impact of adher- ence by doing the following: • Perform a stratified analysis when adherence appears similar between study arms. Such analyses aim to provide unbiased comparisons of subpopula- tions across study arms. • Postulate causal models and performing randomization-based analyses. • Perform matched case-control adherence analyses involving subjects who become HIV infected. Chapter 6 Design Considerations: Recruitment and Retention Recommendation 6-1: Investigators should conduct pretrial research to assess the community and individuals’ interest in the trial, to pilot test recruitment and retention strategies, and to set a realistic timeline and resource needs for the enrollment period and for retention. Recommendation 6-2: Because of the loss in study power that can result from inad- equate accrual and because the potential biases resulting from losses to follow-up cannot be avoided simply by increasing sample size, investigators should place a high priority on developing effective strategies to achieve accrual rate goals and to minimize losses to follow-up. Specifically, investigators should do the following: • Develop a detailed and multifaceted plan for retaining enrolled participants before beginning a study for systematically and frequently monitoring the results, and for modifying the plan if strategies are not working. • Collect as much detailed tracking information as possible on participants. • Develop systems to engage, train, and reward staff for building trust and accountability with participants and within the community, and for meeting recruit- ment and retention targets. Recommendation 6-3: Funders and investigators should include evaluations of the effectiveness of recruitment and retention strategies in future research plans.

SUMMARY 21 Box S-3  Continued Chapter 7 Site Preparedness Recommendation 7-1: Donors and investigators should invest in the human capac- ity and physical infrastructure needed to ensure successful HIV prevention trials in resource-poor settings. These efforts should include a comprehensive and realistic assessment of how to prepare a site, a training plan for staff, and a mentoring plan for inexperienced investigators. Recommendation 7-2: If the regulatory infrastructure of a planned study site is insufficient, study sponsors, funding agencies, research organizations, and other stakeholders should assist local IRBs in developing the ability to provide comprehensive and timely oversight of clinical trials according to international standards. Recommendation 7-3: Sponsors and investigators from outside the trial region should solicit meaningful input from local investigators and community representa- tives as they develop the study protocol, and throughout the trial. The trial should itself promote equal partnerships between outside and local investigators. Recommendation 7-4: Donors should fund and investigators should undertake extensive pretrial research to develop accurate estimates of HIV incidence, par- ticipant accrual, retention, and pregnancy rates, and to develop and evaluate logistical and regulatory processes to be used during the trial. Recommendation 7-5: When considering a new trial site that requires extensive preparation, investigators, sponsors, and community leaders should discuss and carefully consider how the site could be sustained after completion of the trial. Recommendation 7-6: Given limited funding and the extensive investment required to prepare research sites, donors and investigators should explore creative and flexible collaborations with HIV and non-HIV trial networks, health organizations, and local research units that have access to suitable study populations or existing research infrastructure, with cost sharing benefiting both partners. Chapter 8 Estimating HIV Incidence Recommendation 8-1: Investigators should base their estimate of HIV incidence on at least one source of data from the direct longitudinal follow-up of individuals in the trial setting. Given the importance of accurate estimates and the inherent uncertainties of any single approach, the direct estimate of HIV incidence should be corroborated by at least one other source. Recommendation 8-2: Donors and appropriate U.S. and international agencies should make development of a reliable, accurate biomarker-based test for recent Continued

22 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS Box S-3  Continued HIV infection that can be run with blood from a single draw a high priority. They should provide the necessary funding and laboratory resources to conduct a substantial cross-sectional screening program. This will require recruiting subjects from countries with low-level, concentrated, and generalized epidemics during the preseroconversion period and following them for several years. Recommendation 8-3: Although further validation studies are being conducted to examine concerns that the STAHRS and BED tests may produce biased estimates of HIV incidence, investigators should not rely solely at this time on these or other biomarker assays of recent infection to estimate HIV incidence for the specific purpose of designing a prevention trial. Chapter 9 Performing Interim Monitoring and Analyzing Trial Results Recommendation 9-1: The data monitoring committees of trials with sponsors and scientific leaders from outside the host countries should include multiple repre- sentatives from those countries. These members—who should compose at least one-third of the committee—should include scientists, ethicists, and lay people familiar with the community and local norms. Recommendation 9-2: The data monitoring committees for HIV prevention trials should always have the option of unblinding interim results if they believe that doing so might lead them to recommend that the trial be modified or terminated, or lead to other actions that are in the best interests of the trial participants. In particular, when the efficacy data show nonsignificant trends favoring one of the blinded arms, a DMC should unblind itself as this might reflect an intervention that may be harming patients. Recommendation 9-3: Investigators should clearly describe in the study protocol the basis and criteria for any recommendation by the data monitoring committee to modify a trial’s size or duration. If such changes are implemented, the protocol should also specify how investigators should evaluate the trial results. Recommendation 9-4: For effectiveness trials, guidelines for stopping HIV preven- tion trials based on positive interim results should require evidence of a sustained impact on cumulative HIV incidence. Recommendation 9-5: Investigators, donors, and regulatory agencies should en- courage research on how to combine safety information from concurrent trials of similar products, including the scientific advantages and disadvantages of sharing information, the timing and logistics of doing so, ethical concerns (such as how such information might affect the informed-consent process), and how to report the results from such trials.

SUMMARY 23 Box S-3  Continued Recommendation 9-6: Investigators should base their primary analysis of the effi- cacy of an intervention on all randomized subjects. Secondary sensitivity analyses that exclude subjects believed to have been HIV infected when they were random- ized can be useful. However, investigators should not substitute such analyses for the primary analysis, unless such exclusions (and nonexclusions) can confidently be made without error. Recommendation 9-7: Investigators of trials evaluating an intervention that is be- lieved to have a delayed impact may find it efficient to exclude people found to be HIV infected after randomization but before a given follow-up time. If so, the trial protocol should specify and justify such an approach, and investigators should use it only if follow-up of subjects and assessment and confirmation of HIV infection during this period is identical in all study arms. Investigators should undertake secondary analyses based on all randomized subjects. Recommendation 9-8: In all trials, investigators should continue to follow women who become pregnant for HIV infection, regardless of whether they discontinue their study intervention. In addition, intention-to-treat analyses should be the pri- mary basis for comparing intervention groups with respect to HIV infection and other efficacy endpoints. Investigators can include as-treated analyses as second- ary analyses, but should interpret them cautiously, because of the possibility that such discontinuations represent a type of informative censoring. Chapter 10 Alternative Trial Designs Recommendation 10-1: Investigators planning late-stage randomized trials of biomedical interventions are encouraged to utilize partially blinded factorial de- signs in order to also evaluate the relative effectiveness of different behavioral intervention strategies. Factorial designs can provide valuable information about both types of interventions with the same sample size as a trial evaluating only the biomedical intervention. Recommendation 10-2: When feasible and consistent with the scientific goals of a late-stage HIV prevention trial, investigators are encouraged to consider discor- dant couple designs because of their advantages over designs in which the actual HIV exposures of participants are unknown. Recommendation 10-3: Investigators should consider the potential merits of using noninferiority, cluster randomization, and dynamic designs in future biomedical HIV prevention trials.

24 METHODOLOGICAL CHALLENGES IN HIV PREVENTION TRIALS REFERENCES Auerbach, J. D., R. J. Hayes, and S. M. Kandathil. 2006. Overview of effective and promising interventions to prevent HIV infection. World Health Organization Technical Report Series 938:43-78; discussion 317-341. Auvert, B., D. Taljaard, E. Lagarde, J. Sobngwi-Tambekou, R. Sitta, and A. Puren. 2005. Randomized, controlled intervention trial of male circumcision for reduction of HIV infection risk: The ANRS1265 trial. PLoS Medicine 2(11):e298. Bailey, R. C., S. Moses, C. B. Parker, K. Agot, I. Maclean, J. N. Krieger, C. F. Williams, R. T. Campbell, and J. O. Ndinya-Achola. 2007. Male circumcision for HIV pre- vention in young men in Kisumu, Kenya: A randomised controlled trial. Lancet 369(9562):643-656. Berg, K. M., and J. H. Arnsten. 2006. Practical and conceptual challenges in measuring an- tiretroviral adherence. Journal of Acquired Immune Deficiency Syndromes 43(Suppl 1): S79-S87. Bulterys, M., M. G. Fowler, K. K. Van Rompay, and A. P. Kourtis. 2004. Prevention of mother- to-child transmission of HIV-1 through breast-feeding: Past, present, and future. Journal of Infectious Diseases 189(12):2149-2153. Cates, W. 2007. HIV prevention research: The optimist’s view. Paper presented at the Fourth International AIDS Society Conference on HIV Pathogenesis, Treatment, and Prevention, Sydney, Australia. Celum, C., A. Wald, J. Hughes, J. Sanchez, S. Reid, S. Delaney-Moretlwe, F. M. Cowan, J. Fuchs, B. Koblin, L. Corey, and HPTN-039. 2008. HSV-2 suppressive therapy for prevention of HIV acquisition: Results of HPTN 039. Paper presented at Conference on Retroviruses and Opportunistic Infections, Boston, Massachusetts. Chesney, M. A. 2006. The elusive gold standard. Future perspectives for HIV adherence as- sessment and intervention. Journal of Acquired Immune Deficiency Syndromes 43(Suppl 1):S149-S155. Chesney, M. A., D. B. Chambers, and J. O. Kahn. 1997. Risk behavior for HIV infection in participants in preventive HIV vaccine trials: A cautionary note. Journal of Acquired Immune Deficiency Syndromes and Human Retrovirology 16(4):266-271. Global HIV Prevention Working Group. 2006. New approaches to HIV prevention: Accelerat- ing research and ensuring future access. Prepared for the Bill & Melinda Gates Founda- tion and the Kaiser Family Foundation. Gray, R. H., G. Kigozi, D. Serwadda, F. Makumbi, S. Watya, F. Nalugoda, N. Kiwanuka, L. H. Moulton, M. A. Chaudhary, M. Z. Chen, N. K. Sewankambo, F. Wabwire-Mangen, M. C. Bacon, C. F. Williams, P. Opendi, S. J. Reynolds, O. Laeyendecker, T. C. Quinn, and M. J. Wawer. 2007. Male circumcision for HIV pre- vention in men in Rakai, Uganda: A randomised trial. Lancet 369(9562):657-666. International Perinatal HIV Group. 1999. The mode of delivery and the risk of vertical trans- mission of human immunodeficiency virus type 1—a meta-analysis of 15 prospective cohort studies. New England Journal of Medicine 340(13):977-987. IOM (Institute of Medicine). 1995. HIV and the blood supply. Washington, DC: National Academy Press. IOM (Institute of Medicine). 2007. Preventing HIV infection among injecting drug users in high-risk countries: An assessment of the evidence. Washington, DC: The National Academies Press. Kajubi, P., M. R. Kamya, S. Kamya, S. Chen, W. McFarland, and N. Hearst. 2005. Increas- ing condom use without reducing HIV risk: Results of a controlled community trial in Uganda. Journal of Acquired Immune Deficiency Syndromes 40(1):77-82.

SUMMARY 25 Kerr, T., J. Walsh, E. Lloyd-Smith, and E. Wood. 2005. Measuring adherence to highly active antiretroviral therapy: Implications for research and practice. Current HIV/AIDS Reports 2(4):200-205. Liu, H., C. E. Golin, L. G. Miller, R. D. Hays, C. K. Beck, S. Sanandaji, J. Christian, T. Maldonado, D. Duran, A. H. Kaplan, and N. S. Wenger. 2001. A comparison study of multiple measures of adherence to HIV protease inhibitors. Annals of Internal Medicine 134(10):968-977. Lovato, L. C., K. Hill, S. Hertert, D. B. Hunninghake, and J. L. Probstfield. 1997. Recruitment for controlled clinical trials: Literature summary and annotated bibliography. Controlled Clinical Trials 18(4):328-352. McKenzie, M., J. P. Tulsky, H. L. Long, M. Chesney and A. Moss. 1999. Tracking and fol- low-up of marginalized populations: A review. Journal of Health Care for the Poor and Underserved 10(4):409-429. Peterson, L., D. Taylor, R. Roddy, G. Belai, P. Phillips, K. Nanda, R. Grant, E. E. K. Clarke, A. S. Doh, R. Ridzon, H. S. Jaffe, and W. Cates. 2007a. Tenofovir disoproxil fumarate for prevention of HIV infection in women: A phase 2, double-blind, randomized, placebo- controlled trial. PLoS Clinical Trials 2(5). Peterson, L., K. Nanda, B. K. Opoku, W. K. Ampofo, M. Owusu-Amoako, A. Y. Boakye, W. Rountree, A. Troxler, R. Dominik, R. Roddy, and L. Dorflinger. 2007b. Savvy(R) (C31g) gel for prevention of HIV infection in women: A phase 3, double-blind, random- ized, placebo-controlled trial in Ghana. PLoS ONE 2(12):e1312. Pool, R. 2006. PC64 Innovative uses of multi-method triangulation to increase the accuracy of adherence and sexual behaviour data in the MDP301 trial. Paper presented at the Microbicides 2006 conference, Cape Town, South Africa. Prentice, R. L. 1989. Surrogate endpoints in clinical trials: Definition and operational criteria. Statistics in Medicine 8(4):431-440. Robinson, K. A., C. R. Dennison, D. M. Wayman, P. J. Pronovost, and D. M. Needham. 2007. Systematic review identifies number of strategies important for retaining study participants. Journal of Clinical Epidemiology 60(8):757-765. UNAIDS. 2000. Ethical considerations in HIV preventive vaccine research. Geneva, Switzer- land: UNAIDS. UNAIDS. 2007. 2007 AIDS Epidemic Update. Geneva, Switzerland: UNAIDS. Van Damme, L. 2007. Phase 3 trial of 6% cellulose sulfate (CS) gel for the prevention of HIV transmission. Paper read at the Fourth International AIDS Society Conference on HIV Pathogenesis, Treatment and Prevention, Sydney, Australia. Van Damme, L., G. Ramjee, M. Alary, B. Vuylsteke, V. Chandeying, H. Rees, P. Sirivongrangson, L. Mukenge-Tshibaka, V. Ettiegne-Traore, C. Uaheowitchai, S. S. Karim, B. Masse, J. Perriens, and M. Laga. 2002. Effectiveness of col-1492, a nonoxynol-9 vaginal gel, on HIV-1 transmission in female sex workers: A randomised controlled trial. Lancet 360(9338):971-977. Villacorta, V., S. Kegeles, J. Galea, K. A. Konda, J. P. Cuba, C. F. Palacios, and T. J. Coates. 2007. Innovative approaches to cohort retention in a community-based HIV/STI preven- tion trial for socially marginalized Peruvian young adults. Clinical Trials 4(1):32-41. Watson-Jones, D., M. Rusizoka, H. Weiss, K. Mugeye, K. Baisley, J. Changalucha, D. Everett, C. Tanton, T. Clayton, D. Ross, and R. Hayes. 2007. Impact of HSV-2 suppressive therapy on HIV incidence in HSV-2 seropositive women: A randomised controlled trial in Tanzania. Paper read at Fourth International AIDS Society Conference on HIV Patho- genesis, Treatment and Prevention, Sydney, Australia. Weller, S., and K. Davis. 2002. Condom effectiveness in reducing heterosexual HIV transmis- sion. Cochrane Database System Review (1):CD003255.

Next: Introduction »
Methodological Challenges in Biomedical HIV Prevention Trials Get This Book
×
Buy Paperback | $66.00 Buy Ebook | $54.99
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

The number of people infected with HIV or living with AIDS is increasing at unprecedented rates as various scientists, organizations, and institutions search for innovative solutions to combating and preventing the disease. At the request of the Bill & Melinda Gates Foundation, Methodological Challenges in Biomedical HIV Prevention Trials addresses methodological challenges in late-stage nonvaccine biomedical HIV prevention trials with a specific focus on microbicide and pre-exposure prophylaxis trials. This book recommends a number of ways to improve the design, monitoring, and analysis of late-stage clinical trials that evaluate nonvaccine biomedical interventions. The objectives include identifying a beneficial method of intervention, enhancing quantification of the impact, properly assessing the effects of using such an intervention, and reducing biases that can lead to false positive trial results.

According to Methodological Challenges in Biomedical HIV Prevention Trials, the need to identify a range of effective, practical, and affordable preventive strategies is critical. Although a large number of promising new HIV prevention strategies and products are currently being tested in late-stage clinical trials, these trials face a myriad of methodological challenges that slow the pace of research and limit the ability to identify and fully evaluate effective biomedical interventions.

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  6. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  7. ×

    View our suggested citation for this chapter.

    « Back Next »
  8. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!