Skip to main content
Advertisement
  • Loading metrics

Choices in vaccine trial design in epidemics of emerging infections

Summary points

  • The 2014–2016 Ebola epidemic highlighted variations in the design of randomized trials to evaluate investigational vaccines in an emergency setting. Here, we summarize scientific, ethical, and feasibility considerations relevant to different trial designs.
  • We focus on four fundamental choices in designing a trial of an experimental vaccine in the setting of an emerging infectious disease for which no proven vaccines exist: randomization unit, trial population, comparator intervention, and trial implementation. We also consider three ethical issues relevant to trial design: the social and scientific value of the trial, its risk–benefit profile, and the fairness of participant selection.
  • We believe that individual rather than cluster randomization is better suited for estimating the direct protective effect of a vaccine, a measure of great intrinsic interest. Individual randomization should therefore be the default strategy for evaluating investigational vaccines during epidemics.
  • Trial participants may be selected either from the general population or from a group at high risk of exposure to infection, depending on the characteristics of the infection together with statistical, fairness, and feasibility considerations.
  • Use of a placebo control, rather than an active control or delayed intervention, is likely to maximize the social and scientific value of the trial because it facilitates double-blinding and removes concerns that the comparison intervention may affect the incidence of the disease under study.
  • Starting the trial at approximately the same time for all participants should minimize the time required to obtain a result. Such a strategy will be facilitated when sufficient supplies of the investigational vaccine and control interventions (if any) are available at the start of a trial, when the geographic area for the trial is clearly identified (and anticipated to have continuing disease transmission throughout the trial), and when logistics permit rapid recruitment of the entire trial population. Otherwise, a stepped rollout may be necessary, in which recruitment to the trial is staggered over a period.

Introduction

In outbreaks of emerging infectious diseases for which no proven efficacious vaccines exist but investigational vaccines have been developed, it is important both to rapidly test the investigational vaccines and, if effective, to deploy them. Following the 2014–2016 Ebola epidemic, the World Health Organization (WHO), the Coalition for Epidemic Preparedness Innovations, and other bodies committed to developing investigational vaccines for emerging infectious diseases [1,2]. They aim to evaluate them for immunogenicity and safety, so that promising candidates will be available for efficacy testing and possible deployment when an epidemic occurs.

In the Ebola epidemic, various strategies were used for the design of efficacy (i.e., Phase 3) trials for investigational vaccines [3]. Some investigators argued for individually randomized controlled trials (iRCTs), while others argued for forms of cluster-randomized controlled trials (cRCTs) [4,5]. Later in the epidemic, rapidly declining disease incidence required changes to some trial designs. Ideally, principles and protocols based on scientific, ethical, and feasibility considerations should be drawn up in advance of an epidemic, facilitating expediency and trust for rapid, early implementation once an epidemic occurs.

Here, we summarize key scientific, ethical, and feasibility considerations relevant to the design of Phase 3 vaccine trials in epidemic situations. Trial design choices are discussed, highlighting the benefits and drawbacks of each in given contexts.

Scope

When designing and implementing randomized efficacy trials for investigational vaccines after safety and immunogenicity data have been collected (in Phase 1 and 2 trials), some key choices must be made. In the current regulatory system, randomized trials are considered the gold standard and, except in rare circumstances, have been required for vaccine licensure [6,7]. We restrict our scope to randomized trials of a single vaccine against an emerging infectious disease for which no effective vaccine exists. We assume that all participants, whether in the intervention or control group (if any), will have access to the best currently available other preventative measures (e.g., information on how to prevent infection).

We discuss four key elements of trial design: randomization unit, trial population, comparator intervention, and trial implementation. We weave into that discussion three important ethical considerations: the social and scientific value of the trial, its risk–benefit profile, and the fairness of participant selection [8,9]. These aspects are, in our views, key to trial design in these settings.

Randomized vaccine trial design choices during epidemics

Table 1 summarizes the major designs that have been used or proposed for vaccine trials. Some have not been employed in epidemic settings.

thumbnail
Table 1. Possible trial designs to evaluate the efficacy of investigational vaccines during epidemics of emerging infectious diseases.

The table does not provide an exhaustive list, and not all designs have been used for evaluating vaccines against emerging infectious diseases.

https://doi.org/10.1371/journal.pmed.1002632.t001

Randomization unit

Table 2 summarizes key features of iRCTs, in which vaccination is randomized between individuals in the same population, and cRCTs, in which groups of individuals are randomized. In iRCTs, individual-level protective effects are measured (also called direct effects, which can be thought of as the extent to which an individual’s risk of infection, or disease, would be reduced if he or she were the only person vaccinated). In cRCTs, population-level protective effects are measured (i.e., direct plus indirect effects, which take into account the reduction in the transmission of infection in a population if many are vaccinated) [20]. Either approach depends on the fact that, if the investigational vaccine is effective, the control group will be at greater risk of infection; the difference is simply how the membership of the control group is assigned [21].

thumbnail
Table 2. Key features of individually randomized and cluster-randomized trials.

https://doi.org/10.1371/journal.pmed.1002632.t002

When testing an investigational vaccine during an epidemic, it is important to establish an efficacy estimate as rapidly as possible so that, if efficacious, the vaccine may still be deployed in the same epidemic. Also, in a declining epidemic, cases may become so rare that a trial is no longer feasible. iRCTs generally yield results more rapidly and that are easier to interpret than cRCTs and, in that respect, enhance the social and scientific value of a trial.

The fact that an iRCT measures the direct effect of a vaccine, whereas a cRCT measures the combined direct plus indirect effect, may favor either design. By including indirect effects, cRCTs provide a measure of protection closer to what might be obtained in widespread rollout of a vaccine. The combined effect may thus be of specific interest to decision-makers. However, indirect effects are more difficult to extrapolate to other settings than direct effects, the former depending on setting, population, network structure, and vaccine coverage [2831]. A cRCT measures the protective effect that is highly relevant to the context in which the trial is conducted but may be less relevant at a later time in the same population, or in a different population. A cRCT’s direct and indirect effects cannot be easily separated, and it is possible that a vaccine’s performance would be different if another rollout strategy were used or in different settings within the same epidemic (e.g., urban versus rural) [32].

The direct vaccine effect, as measured in iRCTs, is likely to be less variable in different settings and, with assumptions about transmission dynamics, can be used to model indirect effects in different coverage and epidemic settings. Deriving an estimate of the direct effect from a cRCT is more complex and assumption dependent. Thus, we posit that the most valuable parameter to estimate in trials of unproven vaccines is the direct effect, as measured in an iRCT. Importantly, also, direct effects are generally the basis for regulatory decisions on the licensure of vaccines. For all these reasons, we believe that iRCTs should be the default design for evaluating investigational vaccines during epidemics.

Nonetheless, particular circumstances may weigh in favor of a cRCT. Recent work has shown that despite the larger sample size typically required in cRCTs compared to iRCTs because of the design effect (see glossary), in some settings, the difference in sample size may be modest, because the larger effect measured (indirect plus direct) in a cRCT partly offsets this effect [33]. Additionally, in some circumstances, an iRCT design may be logistically complex or may be unacceptable to the local population, which could threaten a trial’s successful completion—and thus its social and scientific value, essential to its justification [12,34]. In the Ebola epidemic, many considered a cRCT as the most feasible and acceptable design. However, with extensive community engagement, it was possible to launch an iRCT of an investigational Ebola vaccine in Liberia [12].

Trial population

Trial participants may be selected either from the general population or from a group at high risk of exposure to infection.

When a vaccine is intended for widespread use in the general population, conducting the trial in the general population will enhance the generalizability of trial results. However, such trials will be feasible only if the incidence of the disease under study is high enough for a trial of manageable size. A vaccine trial conducted in persons at high risk of exposure, such as serodiscordant couples for a sexually transmitted infection [13] or healthcare workers for a disease transmitted by direct contact [14], is likely to reduce the required sample size and have greater statistical efficiency.

Efforts to enhance the risk–benefit profile of a trial may lead to performing a trial in a group that is especially likely to benefit if the investigational vaccine proves effective, such as those with occupational, familial, or household exposure to infection. Similarly, efforts to enhance a trial’s risk–benefit profile may favor excluding those who are most at risk from possible adverse effects of the investigational vaccine; these may include children, pregnant women or the fetuses they carry, or individuals with particular medical conditions, such as immune deficiencies. However, if such individuals would be in the eventual target population for a vaccination program, there are compelling arguments for including them in a trial.

Complexities ensue when these considerations conflict. Consider, for example, pregnant women and investigational vaccine trials against Zika virus infection. Concern about adverse effects on the fetus might argue for excluding all pregnant women. However, pregnant women and their fetuses are likely to benefit the most if an investigational Zika vaccine proves effective. Excluding them can make a trial’s risk–benefit profile significantly less favorable, by not collecting data on a key target population for the vaccine. Importantly, these considerations are also relevant for judging whether a trial’s participant selection is fair, inasmuch as compelling reasons are required for excluding entire population subgroups. A systematic precautionary approach has led to the previous exclusion of pregnant women from vaccine trials, even when they are an important target population for the vaccine [35,36]. The default should therefore be to include pregnant women and other so-called vulnerable groups in investigational vaccine trials during epidemics, provided that the risks of participation are judged acceptable [8,37].

For naturally immunizing infections, investigators sometimes restrict enrollment in a trial to those who have not previously been infected to ensure that trial participants are truly at risk of becoming infected; this is especially relevant when selecting individuals thought to be at high risk for infection. However, selecting participants who both have risk factors for infection and are uninfected at enrollment may be problematic. First, it means that all potential participants must be tested for evidence of prior infection. Second, individuals who have remained uninfected despite many opportunities for exposure may be more resistant to infection (or have lower-risk exposures) than is typical in the general population [38]. Serodiscordant couples, for example, may tend to be those who practice safer sex or for whom the infected partner is less infectious than in other couples. Likewise, healthcare workers who remain uninfected despite apparent intense exposure may be ones who practice excellent personal protection. In the early stages of the recent Ebola epidemic, health workers were identified as a group with particularly high incidence (and a good population for a vaccine trial), but when the high incidence was recognized, personal protective measures against infection were rapidly implemented, leading to a dramatic decline in their risk of infection. Failure to account for such factors may lead to overestimating the likely infection rate during the trial [38]. Moreover, if the vaccine is differentially effective in different populations, such as because of the intensity of exposure, then the effect estimate from such a trial may be different than it would be for the general population [3941].

Targeting at-risk populations can promote fair participant selection, depending on how fairness is defined. When a population is at increased risk of infection because they are undertaking important work, such as burying the dead or caring for patients or family members, targeting this population serves fairness as reciprocity. However, focusing a trial on certain at-risk populations, such as frontline health workers, may invite charges of giving priority to those of relatively high social standing or undermining efforts to equalize access to the investigational vaccine. Ethical debate between these approaches to fairness is ongoing [42,43].

Practical or feasibility considerations may justify use of a high-risk group, for example, if there is a risk of obtaining an inconclusive result in a nontargeted trial, if the available supply of vaccine is limited so a smaller trial is necessary, or if there are substantial resource savings from conducting the study in a high-risk group.

Comparator intervention

A trial may compare participants randomized to receive an investigational vaccine with those randomized to receive placebo, an active control (most commonly a proven effective vaccine against another infection), or delayed administration of the investigational vaccine.

The rationale for considering an active control (a vaccine against another disease) is to provide some benefit to the control group, which may enhance a trial’s risk–benefit profile, albeit not with respect to the disease under study. This is especially the case when the active control can be administered in such a way as to allow for double-blinding, as in the RTS,S/AS01 malaria vaccine trial, in which rabies and meningococcal C vaccines were used as active controls for infants and children, respectively [44]. However, use of an active control can complicate the safety assessment for the investigational vaccine, as only comparative safety between the two interventions will be measured [45]. Additionally, there may be a possibility that the control vaccine has an effect on the outcome measures against which the investigational vaccine is targeted. Active controls should therefore be considered but used judiciously.

Compared to both an active control and delayed intervention, use of a placebo control is likely to maximize a trial’s social and scientific value because of the opportunity for a double-blind design (neither investigators nor participants are aware of who has received the vaccine) and absence of concern that the placebo may affect the incidence of the disease under study in the control group. Use of a placebo control also facilitates assessment of adverse effects. Perhaps the strongest objection to using a placebo arises when the investigational vaccine is thought likely to be highly effective against a lethal disease (e.g., Ebola). Then, investigators may be thought to have a “duty to rescue” by providing the vaccine to all participants in a trial. While vaccines are a form of prophylaxis, not “rescue,” some may see a vaccine against a highly lethal disease as a form of rescue. However, we agree with Millum and Wendler that, in nearly all circumstances, the duty to rescue does not apply to clinical trials [46], when the intervention under investigation is unlicensed and of unproven efficacy and there is no alternative effective intervention. Placebo-controlled trials are typically conducted when the intervention under test is not licensed and is of unproven efficacy. A placebo control group is often scientifically necessary for assessing the investigational vaccine. In such circumstances, the loss for future populations from foregoing a placebo-controlled trial is too high [46]. The placebo control itself, typically a saline injection, introduces hardly any risk. We therefore believe a placebo control is typically the preferred comparator.

During an epidemic, political leaders or community representatives may have a strong preference for ensuring that all trial participants have access to the investigational vaccine, especially if prior evidence strongly suggests effectiveness (e.g., high immunogenicity and protection shown in animal studies), the disease is serious, the disease burden is high, and available preventative or therapeutic measures are limited. If this preference makes use of a placebo control unfeasible, delayed administration may be an alternative, provided the social and scientific value of the study remains intact [47]. When the investigational vaccine is expected to be effective, delayed administration can enhance the trial’s risk–benefit profile compared to a placebo control. The major disadvantage is that individuals clearly know if they are in the vaccine or control group, and this may lead to differential behavior changes, affecting their risk of disease independently of any protective effect of the vaccine. In cRCTs, another disadvantage of assigning control clusters to delayed administration (rather than active or placebo control) is that it may be impossible to identify those in the control clusters who would have been vaccinated had they been offered the vaccine. As acceptors and refusers may be at differential risk of disease independently of vaccination, the only possible unbiased comparison is disease rates among all those in the vaccine clusters compared to those in the control clusters—an intention-to-treat analysis. If substantial numbers refuse vaccination, the vaccine’s effect may be underestimated. A final disadvantage of delayed administration is that it may compromise assessment of the longer-term efficacy of the vaccine. This concern may be moot, however, if the plan is to offer the vaccine to all participants after the trial [47], in which case such an evaluation would be impossible, anyway, regardless of the comparator used in the trial.

Trial implementation

In the simplest experimental trial design, known as a “parallel” design, all participants are enrolled into the trial at about the same time and followed for the same period of time. However, sometimes it is not feasible to enroll all participants in a short period and a “stepped” rollout is used, in which entry to the trial is phased over time. While some degree of stepped rollout occurs in almost all trials (not all participants can be vaccinated on the same day), stepped rollout has been used deliberately in cRCTs. In a so-called “stepped-wedge” cRCT, the order of introduction of the vaccine to the various clusters is randomized, and disease incidence is then compared in successive time intervals between those clusters in which the intervention has already been rolled out and those in which it has not yet been rolled out [17]. By the end of the study, all clusters will have received the investigational vaccine.

Deliberate rollout of the investigational vaccine over a period of time may be implemented because of limited supply of the investigational vaccine or limited capacity to implement the intervention in many locations simultaneously [48]. A stepped rollout can permit prompt commencement of the trial, without waiting until rollout everywhere is feasible. Low and variable disease incidence may also make parallel rollout unfeasible, because the trial can gain sufficient power only by targeting at-risk populations and randomizing individuals or clusters in the vicinity of known or predicted cases [15,19,49]. Such trials, following cases in real time, are necessarily rolled out in a stepped fashion.

Stepped rollout does not have this same advantage if the order of rollout is fixed in advance, as, for example, in a classic stepped-wedge cRCT design, in which clusters (e.g., villages) are offered an investigational vaccine in an order predetermined by randomization. This design was considered during the Ebola epidemic when supplies of investigational vaccines were limited. However, in this case the design was poorly suited to a setting with unpredictable and spatially variable incidence, because much variation would occur between clusters because of factors other than the vaccine, inflating the design effect and reducing the trial’s power. The solution proposed was an iRCT with parallel rollout, which would have greater power than a stepped-wedge design and thus greater social and scientific value [18]. However, an iRCT with a stepped rollout to areas of likely high incidence as supplies become available was also proposed, combining the advantages of stepped rollout with the greater power of an iRCT [15,21].

The Ebola experience shows that conditions favoring stepped rollout can readily occur in an emergency setting. This was an important part of the justification for the ring vaccination design of the “Ebola ça Suffit!” trial. Notably, some conditions (limited supplies and logistics) were particularly acute at the start of the epidemic, while others (variable disease incidence) were particularly pressing at the end. When sufficient supplies of the investigational vaccine and any control interventions are available at the trial’s start; when the trial’s geographic area is clearly identified and anticipated to have continuing transmission throughout the trial; and when logistics permit large-scale rollout simultaneously to the entire trial population, starting the trial at a similar time in all participants will minimize the time required to obtain a result. When these criteria are met, a parallel rollout should be used.

Conclusion

We argue that individually randomized trials with a placebo control should be the default strategy for evaluating investigational vaccines during epidemics. Placebo-controlled trials typically maximize the social and scientific value of the trial, and objections to using placebo, such as a duty to “rescue” individual participants—with an unlicensed investigational vaccine candidate of unproven efficacy—are rarely persuasive. Depending on the pathogen as well as statistical, fairness, and feasibility considerations, trial participants may be selected either from the general population or from a group at high risk of exposure to infection. Starting the trial at approximately the same time for all participants, in a parallel rollout, will minimize the time required to obtain a result and maximize social and scientific value. If resources are limited and/or incidence is spatiotemporally variable, a stepped rollout may be necessary, in which recruitment to the trial is staggered over time.

We have not discussed all aspects of vaccine trial design during epidemics of emerging infectious diseases. However, through a discussion of four fundamental choices and three ethical considerations, we hope to have highlighted the range of options that should be considered during future epidemics. A closely overlapping set of decisions has recently been incorporated into an online interactive decision tool as part of the WHO Research and Development Blueprint for Action to Prevent Epidemics (the InterVax-Tool: http://vaxeval.com/) [50].

In order to further prepare in advance for epidemics, future work should characterize choices for specific pathogen characteristics and contexts. Mathematical modeling and simulation are useful tools for addressing choices beyond the four discussed here, e.g., sample size, trial location, duration, or end point [33,5052]. Rigorous ethical analysis, as well as inclusive and transparent debate by different stakeholders, would help to illuminate the underlying moral questions. Ethical considerations not discussed here, such as informed consent and community acceptance, may also tip the balance between several possible designs. Both scientific and ethical questions would be best debated in advance of future epidemics.

Glossary

  • Blinded design: In a blinded design, the trial’s participants do not know whether they are receiving the investigational vaccine or the control. In a double-blinded design, neither the trial’s participants nor the investigators know who is receiving the investigational vaccine or the control. In non-blinded trials, bias can arise in intervention allocation if, for example, the investigators knowingly put the more or less vulnerable participants in the investigational vaccine arm, or if participants change their behavior if they know they did or did not receive the investigational vaccine.
  • Design effect: A statistical “penalty” incurred in the analysis of cRCTs to account for correlations in outcomes among members of the same cluster, leading to larger sample size requirements [23,24].
  • Direct effects: The extent by which the risk of disease is reduced when an individual is exposed to the infectious agent. The direct effect of a vaccine is mainly a characteristic of the vaccine itself and how it interacts with individuals rather than of the way the vaccine is deployed in a particular population. While direct vaccine effects are not always generalizable [29,40,41,53], they are often assumed to be more easily extrapolated to other settings because direct effects are measured at the individual level and not typically thought to depend much on the patterns of exposure and transmission in the population.
  • Effective/efficacy: We use effective and efficacy in this paper as shorthand for protection against becoming infected without prejudging various aspects of this protection, which are sometimes called efficacy and effectiveness.
  • Indirect effects: Indirect effects occur when the number of persons vaccinated in a community reduces the overall transmission rate of the infection in the community. Sometimes called herd effects, indirect effects benefit both vaccinated and unvaccinated individuals, and the size of the effect will depend both on the level of the direct effect and the proportion of persons vaccinated in the population [20]. The indirect effects of a vaccine depend not only on the direct effects on vaccinated individuals but on such factors as the incidence rate of the disease, the level of pre-existing immunity in the population, the contact network structure, the coverage of the vaccine, and the phase of the epidemic (growing or declining), among others. Each of these factors will vary across populations that may consider using the investigational vaccine if it proves effective, and some of them (e.g., epidemic phase, immunity) may vary over short time periods within the same population.

Acknowledgments

We thank the Ethox Centre at Oxford University for supporting the workshop that led to the writing of this paper, which draws content from some of the materials distributed in advance of that meeting by the organizers (RK NE AR ML) and from the presentations of AR and PS there. We also thank the participants of this workshop for their feedback on this material.

The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institute of General Medical Sciences or the National Institutes of Health.

References

  1. 1. Brende B, Farrar J, Gashumba D, Moedas C, Mundel T, Shiozaki Y, et al. CEPI—a new global R&D organisation for epidemic preparedness and response. Lancet. 2017;389: 233–235. pmid:28109539
  2. 2. WHO. Blueprint for R&D preparedness and response to public health emergencies due to highly infectious pathogens. WHO workshop on prioritization of pathogens report. 2015.
  3. 3. National Academy of Sciences Engineering and Medicine. Integrating clinical research into epidemic response: the Ebola experience. National Academies Press; 2017.
  4. 4. Cohen J, Kupferschmidt K. Tough choices ahead in Ebola vaccine trials. ScienceMag. 2014; 1–8.
  5. 5. Cohen J. Issues continue to dog the testing of Ebola drugs and vaccines. ScienceMag. 2014.
  6. 6. Lipsitch M, Jha A, Simonsen L. Observational studies and the difficult quest for causality: Lessons from vaccine effectiveness and impact studies. Int J Epidemiol. 2016;45: 2060–2074. pmid:27453361
  7. 7. Lipsitch M, Eyal N. Improving vaccine trials in infectious disease emergencies. Science. 2017;357: 153–156. pmid:28706038
  8. 8. WHO, Council for International Organizations of Medical Sciences. International ethical guidelines for health-related research involving humans. Geneva: Council for International Organizations of Medical Sciences; 2016.
  9. 9. Emanuel EJ, Wendler D, Grady C. What makes clinical research ethical? JAMA. 2000;283: 2701–11. pmid:10819955
  10. 10. Black S, Shinefield H, Fireman B, Lewis E, Ray P, Hansen JR, et al. Efficacy, safety and immunogenicity of heptavalent pneumococcal conjugate vaccine in children. Northern California Kaiser Permanente Vaccine Study Center Group. Pediatr Infect Dis J. 2000;19: 187–95. pmid:10749457
  11. 11. Isanaka S, Guindo O, Langendorf C, Matar Seck A, Plikaytis BD, Sayinzoga-Makombe N, et al. Efficacy of a Low-Cost, Heat-Stable Oral Rotavirus Vaccine in Niger. N Engl J Med. 2017;376: 1121–1130. pmid:28328346
  12. 12. Kennedy SB, Neaton JD, Lane HC, Kieh MWS, Massaquoi MBF, Touchette NA, et al. Implementation of an Ebola virus disease vaccine clinical trial during the Ebola epidemic in Liberia: Design, procedures, and challenges. Clin Trials. 2016;13: 49–56. pmid:26768572
  13. 13. Stanberry LR, Spruance SL, Cunningham AL, Bernstein DI, Mindel A, Sacks S, et al. Glycoprotein-D–Adjuvant Vaccine to Prevent Genital Herpes. N Engl J Med. 2002;347: 1652–1661. pmid:12444179
  14. 14. Widdowson M-A, Schrag SJ, Carter RJ, Carr W, Legardy-Williams J, Gibson L, et al. Implementing an Ebola Vaccine Study—Sierra Leone. MMWR Suppl. 2016;65: 98–106. pmid:27387395
  15. 15. Lipsitch M, Eyal N, Halloran ME, Hernán MA, Longini IM, Perencevich EN, et al. Obtaining Efficacy Data in an Environment of Variable and Uncertain Incidence: Ebola and Beyond. Science. 2015;348: 46–48. pmid:25838371
  16. 16. O’Brien KL, Moulton LH, Reid R, Weatherholtz R, Oski J, Brown L, et al. Efficacy and safety of seven-valent conjugate pneumococcal vaccine in American Indian children: Group randomised trial. Lancet. 2003;362: 355–361. pmid:12907008
  17. 17. The Gambia Hepatitis Intervention Study. Cancer Res. 1987;47: 5782–5787. pmid:2822233
  18. 18. Bellan SE, Pulliam JRC, Pearson CAB, Champredon D, Fox SJ, Skrip L, et al. Statistical power and validity of Ebola vaccine trials in Sierra Leone: a simulation study of trial design and analysis. Lancet Infect Dis. 2015;15: 703–710. pmid:25886798
  19. 19. Henao-Restrepo AM, Camacho A, Longini IM, Watson CH, Edmunds WJ, Egger M, et al. Efficacy and effectiveness of an rVSV-vectored vaccine in preventing Ebola virus disease: final results from the Guinea ring vaccination, open-label, cluster-randomised trial (Ebola Ça Suffit!). Lancet. 2017;389: 505–518. pmid:28017403
  20. 20. Halloran ME, Longini IM, Struchiner CJ. Design and Analysis of Vaccine Studies: Introduction. Springer; 2009. https://doi.org/10.1007/978-0-387-68636-3
  21. 21. Eyal N, Lipsitch M. Vaccine testing for emerging infections: the case for individual randomisation. J Med Ethics. 2017; pmid:28396558
  22. 22. Donner A, Birkett N, Buck C. Randomization by cluster: sample size requirements and analysis. Am J Epidemiol. 1981;114: 906–914. pmid:7315838
  23. 23. Donner A, Klar N. Methods for comparing event rates in intervention studies when the unit of allocation is a cluster. Am J Epidemiol. 1994;140: 279–89. pmid:8030631
  24. 24. Hayes RJ, Alexander ND, Bennett S, Cousens SN. Design and analysis issues in cluster-randomized trials of interventions against infectious diseases. Stat Methods Med Res. 2000;9: 95–116. pmid:10946429
  25. 25. Gaudart J, Rebaudet S, Barrais R, Boncy J, Faucher B, Piarroux M, et al. Spatio-Temporal Dynamics of Cholera during the First Year of the Epidemic in Haiti. PLoS Negl Trop Dis. 2013;7(4). pmid:23593516
  26. 26. Grad YH, Miller JC, Lipsitch M. Cholera modeling: challenges to quantitative analysis and predicting the impact of interventions. Epidemiology. 2012;23: 523–30. pmid:22659546
  27. 27. Zhang Q, Sun K, Chinazzi M, Pastore y Piontti A, Dean NE, Rojas DP, et al. Spread of Zika virus in the Americas. Proc Natl Acad Sci U S A. 2017;114: E4334–E4343. pmid:28442561
  28. 28. Staples PC, Ogburn EL, Onnela J-P. Incorporating Contact Network Structure in Cluster Randomized Trials. Sci Rep. 2015;5: 1–12. pmid:26631604
  29. 29. Ali M, Emch M, Von Seidlein L, Yunus M, Sack DA, Rao M, et al. Herd immunity conferred by killed oral cholera vaccines in Bangladesh: A reanalysis. Lancet. 2005;366: 44–49. pmid:15993232
  30. 30. Worby CJ, Chaves SS, Wallinga J, Lipsitch M, Finelli L, Goldstein E. On the relative role of different age groups in influenza epidemics. Epidemics. 2015;13: 10–16. pmid:26097505
  31. 31. Feikin DR, Kagucia EW, Loo JD, Link-Gelles R, Puhan MA, Cherian T, et al. Serotype-Specific Changes in Invasive Pneumococcal Disease after Pneumococcal Conjugate Vaccine Introduction: A Pooled Analysis of Multiple Surveillance Sites. PLoS Med. 2013;10(9). pmid:24086113
  32. 32. Hitchings MDT, Grais RF, Lipsitch M. Using simulation to aid trial design: Ring-vaccination trials. PLoS Negl Trop Dis. 2017;11(3). pmid:28328984
  33. 33. Hitchings MDT, Lipsitch M, Wang R, Bellan SE. Competing Effects Of Indirect Protection And Clustering On The Power Of Cluster-Randomized Controlled Vaccine Trials. Am J Epidemiol. 2018 Mar 7. pmid:29522080
  34. 34. WHO. Ethical issues related to study design for trials on therapeutics for Ebola Virus Disease. 2014.
  35. 35. Lyerly AD, Little MO, Faden R. The second wave: Toward responsible inclusion of pregnant women in research. Int J Fem Approaches Bioeth. 2008;1: 5–22. pmid:19774226
  36. 36. Dresser R. Wanted: single, white male for medical research. Hastings Cent Rep. 1992;22: 1–9.
  37. 37. Ethics Working Group on ZIKV Research & Pregnancy. Pregnant Women & the Zika Virus Vaccine Research Agenda: Ethics Guidance on Priorities, Inclusion, and Evidence Generation. Baltimore; 2017.
  38. 38. Tuite AR, Fisman DN. Spectrum Bias and Loss of Statistical Power in Discordant Couple Studies of Sexually Transmitted Infections. Sex Transm Dis. 2011;38: 50–56. pmid:20693935
  39. 39. Fiebelkorn AP, Rosen JB, Brown C, Zimmerman CM, Renshowitz H, D’Andrea C, et al. Environmental factors potentially associated with mumps transmission in Yeshivas during a mumps outbreak among highly vaccinated students Brooklyn, New York, 2009–2010. Hum Vaccines Immunother. 2013;9: 189–194. pmid:23442590
  40. 40. Lopman BA, Pitzer VE, Sarkar R, Gladstone B, Patel M, Glasser J, et al. Understanding reduced rotavirus vaccine efficacy in low socio-economic settings. PLoS ONE. 2012;7(8). pmid:22879893
  41. 41. Fine PEM. Variation in protection by BCG: implications of and for heterologous immunity. Lancet. 1995;346: 1339–1345. pmid:7475776
  42. 42. Folayan MO, Haire B. Prioritization of healthcare workers for experimental Ebola therapeutic would exacerbate existing inequalities. Developing World Bioethics. 2015. pp. 113–114. pmid:26193194
  43. 43. Satalkar P, Elger BE, Shaw DM. Prioritising healthcare workers for Ebola treatment: Treating those at greatest risk to confer greatest benefit. Dev World Bioeth. 2015;15: 59–67. pmid:25655050
  44. 44. The RTS SCTP (2014). Efficacy and Safety of the RTS,S/AS01 Malaria Vaccine during 18 Months after Vaccination: A Phase 3 Randomized, Controlled Trial in Children and Young Infants at 11 African Sites. PLoS Med. 2014;11(7): e1001685. pmid:25072396
  45. 45. Gessner BD, Knobel DL, Conan A, Finn A. Could the RTS,S/AS01 meningitis safety signal really be a protective effect of rabies vaccine? Vaccine. 2017;35: 716–721. pmid:28065475
  46. 46. Millum J, Wendler D. The Duty to Rescue and Randomized Controlled Trials Involving Serious Diseases. Brill; 2018.
  47. 47. Rid A, Miller FG. Ethical Rationale for the Ebola “Ring Vaccination” Trial Design. Am J Public Health. 2016;106: 432–5. pmid:26794172
  48. 48. King G, Gakidou E, Ravishankar N, Moore RT, Lakin J, Vargas M, et al. A “politically robust” experimental design for public policy evaluation, with application to the Mexican universal health insurance program. J Policy Anal Manage. 2007;26: 479–506. pmid:17633445
  49. 49. Harling G, Wang R, Onnela J, Degruttola V. Leveraging Contact Network Structure in the Design of Cluster Randomized Trials. Harvard Univ Biostat Work Pap Ser. 2016; pmid:27798376
  50. 50. Bellan SE, Eggo RM, Gsell P-S, Kucharski A, Dean N, Donohue R, et al. Guiding Vaccine Efficacy Trial Design During Public Health Emergencies: An interactive web-based decision support tool. bioRxiv. 2018; Available: https://www.biorxiv.org/content/biorxiv/early/2018/01/24/252783.full.pdf
  51. 51. Nason M. Statistics and logistics: Design of Ebola vaccine trials in West Africa. Clinical Trials. 2016;13: 87–91. pmid:26768560
  52. 52. Kahn R, Hitchings M, Wang R, Bellan S, Lipsitch M. Modeling Vaccine Trials in Epidemics with Mild and Asymptomatic Infection. bioRxiv. 2018; Available: https://www.biorxiv.org/content/early/2018/04/06/295337
  53. 53. Gomes MGM, Lipsitch M, Wargo AR, Kurath G, Rebelo C, Medley GF, et al. A Missing Dimension in Measures of Vaccination Impacts. PLoS Pathog. 2014;10(3). pmid:24603721