Skip to main content
Biostatistics (Oxford, England) logoLink to Biostatistics (Oxford, England)
. 2022 Jul 15;24(4):850–865. doi: 10.1093/biostatistics/kxac024

A controlled effects approach to assessing immune correlates of protection

Peter B Gilbert 1,, Youyi Fong 2, Avi Kenny 3, Marco Carone 4
PMCID: PMC10583729  PMID: 37850938

Summary

An immune correlate of risk (CoR) is an immunologic biomarker in vaccine recipients associated with an infectious disease clinical endpoint. An immune correlate of protection (CoP) is a CoR that can be used to reliably predict vaccine efficacy (VE) against the clinical endpoint and hence is accepted as a surrogate endpoint that can be used for accelerated approval or guide use of vaccines. In randomized, placebo-controlled trials, CoR analysis is limited by not assessing a causal vaccine effect. To address this limitation, we construct the controlled risk curve of a biomarker, which provides the causal risk of an endpoint if all participants are assigned vaccine and the biomarker is set to different levels. Furthermore, we propose a causal CoP analysis based on controlled effects, where for the important special case that the biomarker is constant in the placebo arm, we study the controlled vaccine efficacy curve that contrasts the controlled risk curve with placebo arm risk. We provide identification conditions and formulae that account for right censoring of the clinical endpoint and two-phase sampling of the biomarker, and consider G-computation estimation and inference under a semiparametric model such as the Cox model. We add modular approaches to sensitivity analysis that quantify robustness of CoP evidence to unmeasured confounding. We provide an application to two phase 3 trials of a dengue vaccine indicating that controlled risk of dengue strongly varies with 50Inline graphic neutralizing antibody titer. Our work introduces controlled effects causal mediation analysis to immune CoP evaluation.

Keywords: Controlled direct effects, COVID-19 vaccine, Dengue vaccine efficacy, E-value, Immune correlate of protection, Sensitivity analysis

1. Introduction

Safe and effective vaccines to prevent SARS-CoV-2 acquisition and coronavirus disease 2019 (COVID-19) disease are needed to curtail the COVID-19 pandemic. Approval of a vaccine requires demonstration that the vaccine confers a favorable benefit-to-risk profile in reducing clinically significant endpoints, usually established through phase 3 randomized, placebo-controlled, vaccine efficacy (VE) trials (e.g., Baden and others, 2021). Where SARS-CoV-2 vaccines are approved and widely locally available, placebo arms in future efficacy trials will likely be infeasible. Thus, there is a need for alternative approaches to approving SARS-CoV-2 vaccines, such as surrogate endpoint trials that use as primary endpoint an antibody response biomarker measured postvaccination. Effectiveness of this approach would require that the biomarker be measured using a validated assay and has been “scientifically well established to reliably predict clinical benefit” (traditional approval) or be “reasonably likely to predict clinical benefit” (accelerated approval) (Fleming and Powers, 2012; US Food and Drug Administration, 1999).

Immunologic surrogate endpoints based on binding or functional antibody assays have been accepted by regulatory agencies for many licensed vaccines (Plotkin, 2010; Plotkin and Gilbert, 2018). Acceptance has been based on evidence from a variety of sources, including statistical analysis of phase 3 VE trials, natural history studies, vaccine challenge studies in animals and in humans, and passive monoclonal antibody transfer studies (Plotkin, 2010). Phase 3 trials constitute one of the most important sources of evidence, because they rigorously characterize the level of VE, and biomarker levels from breakthrough infection or disease case samples can be analyzed and contrasted with biomarker levels from noncase samples to infer immune correlates of risk (CoRs) and immune correlates of protection (CoPs) (Qin and others, 2007). By CoR, we mean an immunologic biomarker measured from vaccine recipients associated with subsequent occurrence of a clinical endpoint of interest (Qin and others, 2007). In contrast, by CoP, we mean an immunologic biomarker that has been established to provide reliable predictions of VE and hence has been accepted for use in either accelerated or traditional approval (Plotkin and Gilbert, 2018). Establishing a CoP typically requires multiple phase 3 placebo-controlled trials, with the placebo arm needed to enable assessment of criteria for a valid CoP within various statistical frameworks, including surrogate endpoint evaluation (Prentice, 1989; Molenberghs and others, 2008), principal stratification VE moderation evaluation (Follmann, 2006; Moodie and others, 2018), natural effects mediation evaluation (Cowling and others, 2019), stochastic interventional VE evaluation (Hejazi and others, 2021), and meta-analysis (Molenberghs and others, 2008).

This manuscript has two objectives. The first is to propose a new causal inference approach to CoP evaluation based on randomized, placebo-controlled trials—controlled risk and controlled vaccine efficacy (CVE) analysis. Closely linked to the first objective, the second is to propose a causal inference approach to CoP evaluation based on analysis of the vaccine arm only, which is especially relevant for studies without a placebo arm. As a case in point, several ongoing SARS-CoV-2 placebo-controlled VE trials have crossed over placebo recipients to the vaccine arm (Baden and others, 2021), precluding the study of CoPs against longer-term endpoints via methods requiring a contemporaneous placebo arm. These trials follow large numbers of vaccine recipients for SARS-CoV-2 acquisition and disease outcomes, providing the requisite data for CoP analysis based on the vaccine arm only.

Based on analysis of a vaccinated group in a phase 3 or postapproval trial, regression methods (e.g., Cox regression accounting for case-cohort biomarker sampling (Moodie and others, 2018)) may be used to identify CoRs. However, a CoR may fail to be a CoP, because the association parameter (e.g., hazard ratio) may not reflect a causal relationship (VanderWeele, 2013). Consequently, identification of a CoR—while an important intermediate step—is insufficient for validating a CoP (Prentice, 1989; Fleming and Powers, 2012). We address this challenge in two steps. First, building on ideas from Joffe and Greene (2009) applied to the VE trial setting, we define two causal effect parameters whose interpretations directly address criteria for a CoP, and derive formal inferential procedures. Second, acknowledging that estimation of these two parameters requires the absence of unmeasured confounders, we propose a sensitivity analysis and conservative estimation strategy that accounts for potential violations of this assumption.

2. Controlled risk and VE CoPs

2.1. Notation and counterfactual definitions

Let Inline graphic indicate assignment to vaccine, and, if the study included a placebo, let Inline graphic indicate assignment to placebo. Let Inline graphic be a vector of baseline covariates chosen with purpose to render the identifiability assumptions of Sections 2.2 and 3.1 as plausible as possible. Inline graphic may also include additional baseline prognostic factors that are not strictly needed for identification but may increase the precision of resulting inferences. Let Inline graphic be an immunologic biomarker measured at a given postvaccination visit occurring at some fixed time Inline graphic, for example, the Day 57 visit in the Moderna COVE phase 3 trial (Baden and others, 2021). Let Inline graphic be the time from baseline until the clinical endpoint of interest. We will focus on studying the occurrence of the endpoint by a given fixed time Inline graphic of interest after Inline graphic. We assume that Inline graphic for each participant but discuss a relaxation of this assumption in Section A of the Supplementary Material available at Biostatistics online.

For each Inline graphic, we denote by Inline graphic and Inline graphic the potential values of Inline graphic and Inline graphic, respectively, under an intervention that sets Inline graphic, and by Inline graphic the potential value of Inline graphic under an intervention that sets both Inline graphic and Inline graphic. The controlled risk

graphic file with name Equation1.gif (2.1)

is the probability of endpoint occurrence by Inline graphic in the counterfactual scenario in which all trial participants are assigned to Inline graphic and have their biomarker level Inline graphic set to Inline graphic. For Inline graphic, this counterfactual estimand is defined even in the absence of a placebo arm. We refer to the function Inline graphic corresponding to vaccine recipients as the controlled risk curve. For a placebo-controlled trial, we define the CVE surface as

graphic file with name Equation2.gif (2.2)

In the literature on controlled effects causal mediation (Robins and Greenland, 1992; Pearl, 2001; Joffe and Greene, 2009), for any value Inline graphic, Inline graphic represents an average controlled direct effect at Inline graphic based on a multiplicative (rather than the traditional additive) contrast.

For many VE trials, including COVID-19 and HIV-1 studies, the primary study population is naive to the pathogen under study. Consequently, because Inline graphic measures an adaptive immune response specific to the pathogen, Inline graphic is known to be “negative” (i.e., below the assay limit Inline graphic of detection) for all placebo recipients. By convention, we set such negative values to an arbitrary number Inline graphic. In this scenario in which Inline graphic is constant, Inline graphic is well-defined for Inline graphic but undefined for all Inline graphic; see Gilbert and others (2011) for further discussion of this point. Therefore, in this scenario, the CVE surface can be summarized by the CVE curve Inline graphic, defined over possible values that Inline graphic can take. Because in this scenario Inline graphic almost surely, the CVE curve at biomarker level Inline graphic can be expressed as

graphic file with name Equation3.gif (2.3)

For other VE trials, the primary study population may instead include individuals previously infected with the pathogen, so that Inline graphic varies also in placebo recipients. In this case, the surface Inline graphic can be considered over scientifically relevant values of Inline graphic.

In either scenario, the vaccine arm provides the most relevant information for adjudicating a CoP, and we define Inline graphic to be a controlled risk CoP if Inline graphic varies in Inline graphic. The curve Inline graphic describes the strength and nature of the CoP, with a most useful CoP having a large decrease in Inline graphic as Inline graphic moves from small to large values. For a placebo-controlled trial with Inline graphic constant, we define Inline graphic to be a CVE CoP if Inline graphic varies in Inline graphic. An ideal CoP has Inline graphic—that is, no average controlled direct effect at level Inline graphic—indicating that Inline graphic fully mediates the effect of the vaccine. For Inline graphic variable, we define Inline graphic to be a CVE CoP if Inline graphic varies in Inline graphic for Inline graphic, and an ideal CoP has no average direct effects: Inline graphic for all Inline graphic.

In the constant Inline graphic scenario, because Inline graphic depends on Inline graphic entirely through Inline graphic, Inline graphic is a controlled risk CoP if and only if it is a CVE CoP. In this case, the distinction for applications is whether a placebo arm is available. Where available, a CVE CoP has a preferred interpretation, namely facilitating the prediction of VE in new settings (immunobridging). Advantageously, in the constant Inline graphic scenario, Inline graphic has a special connection to the natural effects mediation literature (Cowling and others, 2019), where the average controlled direct effect at Inline graphic equals the natural direct effect, and VE is completely mediated through Inline graphic if and only if Inline graphic.

2.2. Basic identification formulas with fully observed data

We first consider a simplified trial setting in which the data unit available on each study participant consists of Inline graphic, where Inline graphic is independent of Inline graphic for each Inline graphic by randomization. Define the marginalized risk at level Inline graphic to be

graphic file with name Equation4.gif (2.4)

with Inline graphic and where the outer expectation is over the marginal distribution of Inline graphic in the study population. The expression for Inline graphic is the single time-point G-computation formula, equivalent to direct standardization using the marginal distribution of Inline graphic as standard. We focus on the vaccine arm parameter Inline graphic. The placebo arm parameter Inline graphic can be studied similarly in the variable Inline graphic scenario. In the constant Inline graphic scenario, Inline graphic is only defined for Inline graphic, with Inline graphic

The parameter Inline graphic is a CoR association parameter, which averages the biomarker-conditional risk over the marginal distribution of Inline graphic. In contrast, Inline graphic and Inline graphic are causal parameters, which means they convey more useful information for whether and how a biomarker Inline graphic meets the “provisional approval” goalpost that an immunologic biomarker is reasonably likely to predict VE. To illustrate this, we note that if Inline graphic for Inline graphic, then by definition, risk decreases (and VE increases) if the antibody biomarker is increased from Inline graphic to Inline graphic. In contrast, this implication does not necessarily follow if Inline graphic for Inline graphic, because an unmeasured confounder not included in Inline graphic could create a reversal wherein Inline graphic even though Inline graphic.

Suppose that, for a given biomarker level Inline graphic: (A1) the Stable Unit Treatment Value assumption (stated in Section B.1.1 of the Supplementary Material available at Biostatistics online) holds; (A2) the covariate vector Inline graphic is sufficient to deconfound the relationship between Inline graphic and Inline graphic (stated in two parts as (A2.1) and (A2.2) below), and (A3) there is sufficient overlap, in that the conditional density function of Inline graphic evaluated at Inline graphic given Inline graphic and Inline graphic is positive almost surely (positivity assumption). Then, the controlled risk is identified by the marginalized risk:

graphic file with name Equation5.gif (2.5)

For identifying the Inline graphic surface in the variable Inline graphic scenario, conditions (A1) and (A3) are required for both the vaccine and placebo arms. Using the terminology of Joffe and Greene (2009), (A2) can be expressed as requiring both (A2.1) [initial randomization] Inline graphic and Inline graphic are independent given Inline graphic for each Inline graphic, and (A2.2) [strong sequential ignorability] Inline graphic and Inline graphic are independent given Inline graphic for each Inline graphic. In the constant Inline graphic scenario, (A2.1) and (A2.2) are only needed for Inline graphic, and the assumption that Inline graphic and Inline graphic are independent given Inline graphic is added to initial randomization. Figure 1 shows the directed acyclic graph (DAG) expressing (A2.1) and (A2.2) (Robins and Greenland, 1992) as well as a DAG relaxing (A2.2) to allow unmeasured confounding Inline graphic (addressed in Sections 4 and 5).

Fig. 1.

Fig. 1.

(A) DAG expressing the causal assumptions (A2.1) (initial randomization) and (A2.2) (strong sequential ignorability); (B) DAG relaxing (A2.2) to a more realistic causal model that allows an unmeasured confounder Inline graphic of the effect of Inline graphic on Inline graphic. The variables in light gray are unobserved.

3. Statistical inference under a two-phase sampling study design

3.1. Identification formulas based on two-phase sampling data with incomplete follow-up

In practice, the time-to-event outcome Inline graphic is observed subject to right-censoring by a random variable Inline graphic, which we can consider to only take values no greater than Inline graphic. Since measuring Inline graphic is resource-intensive, a two-phase sampling design may be employed, where Inline graphic is only measured for a subset of trial participants, possibly selected on the basis of all available data at time Inline graphic. The observed data unit for a participant can thus be written as Inline graphic, where Inline graphic is the observed follow-up time, Inline graphic is the event indicator, and Inline graphic is the indicator that Inline graphic is recorded. The notation Inline graphic emphasizes that Inline graphic is only available if Inline graphic.

In view of the missingness induced by two-phase sampling and right-censoring, conditions (A1)–(A3) alone do not identify Inline graphic. As argued above, under these conditions, Inline graphic can be expressed as Inline graphic, a summary of the distribution of the ideal observed data unit (i.e., with neither missingness nor loss to follow-up). Additional conditions are needed to express Inline graphic itself as a summary of the distribution of the actually observed data unit, namely: (A4) Inline graphic is missing at random given Inline graphic and Inline graphic, in that Inline graphic with Inline graphic; and (A5) Inline graphic is independent of Inline graphic given Inline graphic and Inline graphic. In the variable Inline graphic scenario, the placebo arm versions of (A4)–(A5) identify Inline graphic; without loss of generality, we focus on Inline graphic based on the vaccine arm Inline graphic.

3.2. Inference on controlled risk and CVE

Various approaches can be used, many of which are based on positing a model for Inline graphic, estimating the unknown parameters of this model, and obtaining predicted values Inline graphic for each vaccine recipient Inline graphic. Consistent estimation of Inline graphic requires accounting for the biased sampling of the two-phase sampling design, typically by incorporating estimated inverse-probability-of-sampling weights (Prentice, 1986; Breslow and Holubkov, 1997). Consistent estimation of Inline graphic can usually be guaranteed in VE trials because the investigator controls which participants are sampled for measurement of Inline graphic. Given the resulting estimate Inline graphic for each of the Inline graphic participants assigned Inline graphic, Inline graphic is then estimated by

graphic file with name Equation6.gif (3.6)

If there is a placebo arm, the average can be taken over vaccine and placebo recipients combined.

For prospective cohort studies or VE trials with two-phase sampling designs for measuring Inline graphic, semiparametric failure time models (e.g., the semiparametric linear transformation model) have been widely used to estimate the conditional risk Inline graphic. For example, Self and Prentice (1988) showed how Inline graphic and hence Inline graphic can be consistently estimated under (A1)–(A5) through correct specification of a proportional hazards model (Section C of the Supplementary Material available at Biostatistics online). A flexible parametric approach tailored to vaccine studies may also be used (e.g., Son and Fong, 2021). For parametric or simple semiparametric approaches, valid confidence intervals (CIs) for Inline graphic can be constructed either via the bootstrap or influence functions.

To reduce the risk of bias due to model misspecification, flexible nonparametric or machine learning-based regression approaches could be used to estimate Inline graphic (Price and others, 2018; Westling and others, 2021). However, to our knowledge, no nonparametric method currently exists to provide inference on Inline graphic while handling all the complexities present in VE trials, including a continuous “exposure” Inline graphic measured via two-phase sampling and a continuous failure time subject to right-censoring. Nonparametric methods for this problem are theoretically complex since they admit nonstandard asymptotics (Westling and Carone, 2020) and the nonparametric bootstrap typically fails. Accordingly, in this manuscript, we only consider traditional semiparametric modeling approaches to estimating Inline graphic.

4. Sensitivity analysis: conservative bound estimation of the controlled risk ratio and controlled risk curve

4.1. E-values and conservative upper bounds

As CoR analysis is based on observational data—the biomarker value is not randomly assigned—a central concern is that unmeasured or uncontrolled confounding of the association between Inline graphic and Inline graphic could render Inline graphic. Therefore, for any two fixed Inline graphic values Inline graphic and Inline graphic, the estimate of the marginalized risk ratio Inline graphic may be biased for the controlled risk ratio Inline graphic.

Sensitivity analysis is useful for evaluating how strong unmeasured confounding would have to be to explain away an inferred causal association, that is, inference on Inline graphic indicates an association yet Inline graphic is flat in Inline graphic. We supplement results on estimation and inference on Inline graphic through the equation Inline graphic (under the identifiability assumptions) with the reporting of E-values, which summarize the evidence of a causal effect accounting for potential unmeasured confounding. The E-value is the minimum strength of association, on the risk ratio scale, that an unmeasured confounder would need to have with both exposure Inline graphic and outcome Inline graphic to fully explain away a specific observed exposure–outcome association, conditional on the measured covariates (VanderWeele and Ding, 2017; VanderWeele and Mathur, 2020). We use the E-value defined in terms of marginalized means, such that “explained away” means that under the E-value-level of unmeasured confounding the causal effect would be nullified (Inline graphic). Because Inline graphic, evidence for Inline graphic is equivalently evidence for Inline graphic. Thus, in a placebo-controlled trial, Inline graphic can be interpreted as the multiplicative degree of superior VE caused by marker level Inline graphic compared to level Inline graphic.

Consider two marker-level subgroups Inline graphic and Inline graphic with Inline graphic and Inline graphic. We focus on two prespecified values Inline graphic and do not consider simultaneous inference over a set of values. The E-value for the point estimate Inline graphic is

graphic file with name Equation7.gif (4.7)

The E-value Inline graphic for the upper 95Inline graphic confidence limit Inline graphic for Inline graphic is

graphic file with name Equation8.gif

These two E-values quantify confidence in an immunologic biomarker as a controlled risk CoP—and as a CVE CoP in the constant Inline graphic scenario if there is a placebo arm—with E-values near one suggesting weak support and evidence increasing with greater E-values.

It is also useful to provide conservative estimates of controlled risk ratios and of the controlled risk curve, accounting for unmeasured confounding. We approach these tasks based on the sensitivity/bias analysis approach of Ding and VanderWeele (2016). Define two context-specific sensitivity parameters for any given Inline graphic: Inline graphic is the maximum risk ratio for the outcome Inline graphic comparing any two categories of the unmeasured confounder Inline graphic, within either exposure group Inline graphic or Inline graphic, conditional on the vector Inline graphic of observed covariates; and Inline graphic is the maximum risk ratio for any specific level of the unmeasured confounder Inline graphic comparing individuals with Inline graphic to those with Inline graphic, with adjustment already made for the measured covariate vector Inline graphic. Thus, Inline graphic quantifies the importance of the unmeasured confounder Inline graphic for the outcome, and Inline graphic quantifies how imbalanced the exposure/marker subgroups Inline graphic and Inline graphic are in the unmeasured confounder Inline graphic. The two sensitivity parameters are defined mathematically in Section D of the Supplementary Material available at Biostatistics online. We suppose that Inline graphic for the values Inline graphic used in a data analysis—this is the case of interest for immune correlates assessment.

Define the bias factor

graphic file with name Equation9.gif

for Inline graphic, and define Inline graphic the same way as Inline graphic except marginalizing over the joint distribution of Inline graphic and Inline graphic. Then, it follows that Inline graphic, where Inline graphic with Inline graphic and Inline graphic (Ding and VanderWeele, 2016). Translating this result into our context, under (A1) and (A3), we have that Inline graphic, so that

graphic file with name Equation10.gif (4.8)

It follows that a conservative (upper bound) estimate of Inline graphic is Inline graphic, and a conservative 95Inline graphic CI is obtained by multiplying each confidence limit for Inline graphic by Inline graphic. These estimates account for the presumed-maximum plausible amount of deviation from the no unmeasured confounders assumption specified by user-supplied values Inline graphic and Inline graphic. The bound (4.8) holds without making any assumption about the observed potential confounder Inline graphic or unmeasured confounder Inline graphic (Ding and VanderWeele, 2016).

To provide conservative inference for Inline graphic, we next select a central value Inline graphic of Inline graphic such that Inline graphic matches the observed overall risk, Inline graphic. At value Inline graphic the observed marginalized risk equals the observed overall risk. Next, we “anchor” the analysis by assuming Inline graphic where picking the central value Inline graphic makes this plausible to be at least approximately true. Under this assumption, the bound (4.8) implies the bounds

graphic file with name Equation11.gif (4.9)
graphic file with name Equation12.gif (4.10)

Therefore, after specifying Inline graphic and Inline graphic for all Inline graphic, we conservatively estimate Inline graphic by plugging Inline graphic into the formulas (4.9) and (4.10). Because Inline graphic is always greater than 1 for Inline graphic, formula (4.9) pulls the observed risk Inline graphic upwards for subgroups with high biomarker values, and formula (4.10) pulls the observed risk Inline graphic downwards for subgroups with low biomarker values. This makes the estimate of the controlled risk curve flatter, closer to the null curve, as desired for a sensitivity/robustness analysis.

To specify Inline graphic, we note that it should have greater magnitude for a greater distance of Inline graphic from Inline graphic, as determined by specifying Inline graphic and Inline graphic increasing with Inline graphic for Inline graphic. We consider one specific approach, which sets Inline graphic to the common value Inline graphic specified log-linearly as Inline graphic for Inline graphic. Then, for a user-selected pair of fixed values Inline graphic and Inline graphic with Inline graphic, we set a sensitivity parameter Inline graphic to some value above 1. It follows that, for Inline graphic,

graphic file with name Equation13.gif

Figure 2 illustrates the surfaces determined by Inline graphic and Inline graphic.

Fig. 2.

Fig. 2.

Inline graphic and Inline graphic surfaces for Inline graphic with user-supplied sensitivity parameter Inline graphic with Inline graphic the median of Inline graphic and Inline graphic the 95th percentile of Inline graphic (the specified degree of unmeasured confounding) where Inline graphic

4.2. Use of marginalized risk and generality of the approach

Our approach focuses on marginalized covariate-adjusted association parameters. This choice provides a simple conversion of the association parameters to causal parameters through the equation Inline graphic. Moreover, a very common approach to covariate-adjusted CoR analysis consists of fitting a two-phase sampling regression (e.g., logistic, Cox) model to the immunologic biomarker and observed confounders, and reporting estimates of the association parameter (e.g., odds ratio, hazard ratio) corresponding to the immunologic biomarker covariate. Because odds ratios and hazard ratios are not collapsible, the conditional odds ratio and conditional hazard ratio do not in general equal the marginalized odds ratio and marginalized hazard ratio, respectively (Robins and Greenland, 1992; Loux and others, 2017). The dependency of the conditional odds/hazard ratio on the set of confounders conditioned upon may make these parameters less useful for generalizability of inferences than the marginalized parameters. Yet, the same approach applies for alternative marginalized parameters, for example, the marginalized odds ratio Inline graphic. The same specification of Inline graphic and Inline graphic can be used, as the outcome in VE trials is generally rare (Ding and VanderWeele, 2016; VanderWeele and Ding, 2017).

5. Estimation of CVE with sensitivity analysis

5.1. Constant Inline graphic scenario

As evidence for a CVE CoP should be robust to potential bias from unmeasured confounding, we propose reporting conservative estimates and CIs for Inline graphic that include a margin for potential unmeasured confounding making the estimated CVE curve flatter. To do this, we write Inline graphic and estimate the bounds (4.9) and (4.10) rather than Inline graphic itself. The denominator is the placebo arm marginalized risk, which equals Inline graphic in view of randomization; there is thus no concern about unmeasured confounding. Robust and efficient approaches for estimation of this probability accounting for right-censoring include one-step debiasing (Westling and others, 2021) and targeted minimum loss-based estimation (TMLE) (Benkeser and others, 2019); additional details are provided in Section C.2 of the Supplementary Material available at Biostatistics online. We apply the latter for the dengue example that follows. While the bootstrap could be used whenever parametric or certain semiparametric regression approaches are used to estimate Inline graphic, we instead consider Wald intervals with influence function-based estimators of the standard error; additional details are provided in Section C.3 of the Supplementary Material available at Biostatistics online.

5.2. Variable Inline graphic scenario

If Inline graphic is variable, estimation and inference for Inline graphic does not change, whereas estimation and inference for Inline graphic proceeds similarly as Inline graphic but reversing the role of the vaccine and placebo arms. Again, influence function-based inference can be conducted for Inline graphic using a delta method argument. Given that vaccination can generally only increase immune responses compared to nonvaccination/placebo so that Inline graphic almost certainly, it is only relevant to study Inline graphic for values Inline graphic. One strategy for sensitivity analysis is based on the approach described for the constant Inline graphic scenario, first applied to inject unmeasured confounding that makes Inline graphic flatter, and secondly applied to inject unmeasured confounding that alters Inline graphic to further flatten Inline graphic. In the dengue example, we apply a simpler approach that only injects unmeasured confounding that flattens the estimated curve Inline graphic, assuming no unmeasured confounding for estimation of Inline graphic.

6. Analysis of two dengue VE trials

In the CYD14 (NCT01373281) (Capeding and others, 2014) and CYD15 (NCT01374516) (Villar and others, 2015) trials, participants were randomized 2:1 to receive CYD-TDV dengue vaccine or placebo, administered at Months 0, 6, and 12. The primary analyses assessed VE against symptomatic, virologically confirmed dengue (VCD) occurring at least 28 days after the third immunization through to the Month 25 visit. The Benkeser and others (2019) TMLE method (unbounded version) implemented with the survtmle R package estimated VE to be 57.0Inline graphic (95Inline graphic CI 44.5–65.9) in CYD14 and 61.7Inline graphic (95Inline graphic CI 51.9–68.4) in CYD15. Sanofi Pasteur conducted the CYD14 and CYD15 trials and provided access to the data.

Month 13 (M13) neutralizing antibody (nAb) titers to each serotype were measured through case-cohort Bernoulli random sampling of all randomized participants at enrollment and from all participants who experienced the VCD endpoint after M13 and by Month 25 (cases). A participant’s average M13 logInline graphic-transformed geometric mean titer across serotypes (“M13 average titer”) has been studied as a CoR of VCD (Vigne and others, 2017; Moodie and others, 2018); we study this biomarker as a controlled risk CoP and as a CVE CoP.

With Inline graphic the number of days from M13 to VCD studied through Inline graphic the M25 visit, we estimate Inline graphic using the case-cohort sampling Cox partial likelihood regression method used in Moodie and others (2018) (Section C of the Supplementary Material available at Biostatistics online). The baseline covariate vector Inline graphic includes protocol-specified age categories, sex, and country. Participants without VCD through the M13 visit and with M13 average titer measured are included in analyses.

We first investigate how the marginalized risk of VCD compares between vaccine recipients with highest versus lowest tertile values of M13 average titer, coded Inline graphic and Inline graphic. The tasks of CoR analysis are to estimate Inline graphic, Inline graphic and Inline graphic. Given the sampling design, Inline graphic is 1.0 for all observed VCD cases Inline graphic and is 0.195 (0.096) for observed noncases Inline graphic sampled into the CYD14 (CYD15) subcohort. We estimate each Inline graphic based on equation (3.6). We use the bootstrap to obtain 95Inline graphic pointwise CIs for each marginalized risk and marginalized risk ratio, which directly provide CIs for each controlled risk and controlled risk ratio assuming no unmeasured confounding (A2.1) and (A2.2). Plugging the results into the E-value formulas quantify robustness to unmeasured confounding. The results in Table 1 show E-values for Inline graphic much larger than 1.0 with 95Inline graphic CIs lying below 1.0 for both trials, supporting a controlled risk CoP robust to unmeasured confounding. For instance, the CYD15 results with 95Inline graphic CI 0.04–0.20 about Inline graphic that builds in margin for unmeasured confounding can be interpreted as M13 average titer in the third vs. first tertile causing at least five times (5 = 1/0.20) greater VE, accounting for uncertainty both due to sampling variability and unmeasured confounding.

Table 1.

Analysis of M13 average titer (upper vs. lower tertile) as a CoR and a controlled risk CoP: CYD14 and CYD15 dengue VE trials

  Marginalized risk ratio Inline graphic Controlled risk ratio Inline graphic e(0,1)Inline graphic
Trial Point Est. 95Inline graphic CI Point Est. 95Inline graphic CI Point Est. 95Inline graphic CI UL
CYD14 0.17 0.08–0.29 0.38 0.18–0.66 11.6 6.3
CYD15 0.05 0.02–0.09 0.10 0.04–0.20 43.7 22.0

Inline graphic Conservative (upper bound) estimate assuming unmeasured confounding at level Inline graphic and thus Inline graphic.

Inline graphic E-values are computed for upper tertile Inline graphic vs. lower tertile Inline graphic biomarker subgroups after controlling for age, sex, and country; UL = upper limit of 95Inline graphic confidence interval.

Next, we repeat the analysis treating Inline graphic as a quantitative variable, where Inline graphic is again estimated by two-phase Cox partial likelihood regression. Let Inline graphic and Inline graphic be the 15th and 85th percentile of M13 average titer in vaccine recipients. The results for Inline graphic are Inline graphic = 0.20 (95Inline graphic CI 0.12–0.31) for CYD14 and Inline graphic = 0.13 (95Inline graphic CI 0.09–0.18) for CYD15. For CYD14, the E-value Inline graphic is 9.2, with E-value Inline graphic for the upper limit equal to 5.8. For CYD15, the E-value Inline graphic is 14.6, with E-value Inline graphic for the upper limit equal to 10.5. Next, we set Inline graphic, such that Inline graphic. The resulting upper bound estimates of the controlled risk ratio are Inline graphic = 0.47 (95Inline graphic CI 0.28–0.72) for CYD14 and Inline graphic = 0.30 (95Inline graphic CI 0.19–0.42) for CYD15. Figure 3 shows the sensitivity analysis technique described in Section 4. After building in the margin for unmeasured confounding, for CYD14, estimated VCD risk decreases from 0.032 at low M13 average titer value Inline graphic to 0.01 at high titer value Inline graphic; for CYD15, these estimates are 0.033 and 0.008, respectively. Supplementary Figure S2 of the Supplementary Material available at Biostatistics online repeats the analysis under (A2.1) and (A2.2) using Wald CIs constructed using analytic variance estimation based on the influence function of Inline graphic under the Cox model (Section C.3 of the Supplementary Material available at Biostatistics online); the intervals are similar to the bootstrap intervals.

Fig. 3.

Fig. 3.

Analysis of M13 average titer Inline graphic (quantitative with anti-logInline graphic x-axis label) as a CoR and a controlled risk CoP: CYD14 and CYD15 dengue VE trials. Solid lines are point estimates and dashed lines are 95Inline graphic CIs. The bands that start higher on the left with a steeper shape are for marginalized risk Inline graphic and the bands with a shallower shape are conservative estimates of controlled risk Inline graphic, with Inline graphic and hence Inline graphic, where Inline graphic and Inline graphic are 15th and 85th percentiles of Inline graphic.

Figure 4 shows results for the Inline graphic surface for Inline graphic with Inline graphic fixed at the median of Inline graphic (Inline graphicInline graphic for CYD14 (CYD15)). The results show that after accounting for potential unmeasured confounding the point estimate of Inline graphic in CYD14 monotonically increases from 30.4Inline graphic (95Inline graphic CI 3.4–48.1) at Inline graphic (an average controlled direct effect) to 78.2Inline graphic (95Inline graphic CI 67.4–86.7) at Inline graphic. The degree of increase is similar for CYD15: from 37.5Inline graphic (95Inline graphic CI 23.9–48.2) at Inline graphic to 77.2Inline graphic (95Inline graphic CI 70.1–84.0) at value Inline graphic. This supports a robust controlled VE CoP.

Fig. 4.

Fig. 4.

Analysis of M13 average titer Inline graphic (quantitative with anti-logInline graphicx-axis label) as a CVE CoP: CYD14 and CYD15 dengue VE trials. Solid lines are point estimates of Inline graphic for Inline graphic with Inline graphic fixed at the median of the distribution of Inline graphic [Inline graphicInline graphic for CYD14 (CYD15)] and dashed lines are 95Inline graphic CIs. The faint lines are estimates of Inline graphic assuming no unmeasured confounding and the darker lines are conservative estimates of Inline graphic accounting for potential unmeasured confounding, using the same sensitivity parameters as for Figure 3.

For validity, the analyses require positivity (A3). Price and others (2018) showed that M13 average titer in vaccine recipients varied over its whole range at each level of Inline graphic in CYD14 and in CYD15, supporting this assumption. In conclusion, the evidence for M13 nAb titer as a controlled risk CoP and as a controlled VE CoP is robust to unmeasured confounding. As some vaccine recipients with high titer experienced VCD, the CoP is a relative CoP, not an absolute CoP, in the vaccine field nomenclature (Plotkin, 2010; Plotkin and Gilbert, 2018).

The same methods were applied to the Moderna phase 3 trial (Gilbert and others, 2022).

7. Discussion

In virtually all immune correlates analyses of VE trials or prospective cohort studies, immunologic biomarkers are studied as correlates of risk in vaccine recipients. Given the goal to establish a biomarker as an immune correlate of protection/surrogate endpoint, it is generally of interest to evaluate the extent to which the correlates of risk results—which are based on associational parameters—can be interpreted in terms of causal effects, making them constitute a more reliable basis for decision-making for various vaccine research applications. We proposed a general approach to augment correlates of risk analysis with a conservative analysis of the controlled risk curve Inline graphic, and also, if there was a randomized placebo arm, of the controlled vaccine efficacy curve Inline graphic (if Inline graphic is constant) and controlled vaccine efficacy surface Inline graphic (if Inline graphic is variable). The approach is general in that it allows the use of any semiparametric modeling method for estimation of the underlying conditional risk curve. It is conservative since it provides estimates and CIs incorporating a specified amount of unmeasured confounding, thus making it more difficult to conclude a controlled risk or controlled vaccine efficacy CoP. For the controlled risk ratio, the conservative estimation and inference is achieved by reporting E-values for the point estimate and upper 95Inline graphic confidence limit, whereas for the controlled risk and controlled vaccine efficacy curve/surface it is quantified through a parametrization of the bias function that makes the controlled risk estimate flatter/closer to the null.

In line with VanderWeele and Ding (2017), we do not think a particular E-value magnitude indicating a truly robust result should be prespecified, as the E-value interpretation depends on the problem context, including the study endpoint, the vaccine, the set of potential confounders that are adjusted for, and the plausible magnitude of unmeasured confounders. The assessment of controlled CoPs should include the study of the strength of observed confounding as context for interpreting potential unmeasured confounding (see Section E of the Supplementary Material available at Biostatistics online).

Our sensitivity analysis only addresses unmeasured confounding; violation of the positivity assumption, selection bias, and missing data could also make the controlled CoP results misleading. For prospective cohort studies with investigator control over which blood samples are selected for biomarker measurement, selection bias, and missing data should not be a problem, unless a large percentage of participants have missing blood samples at the biomarker sampling time point. The positivity assumption is required for identification of our causal parameters, and so, we recommend that the approach only be applied if diagnostics support that the biomarker in vaccine recipients tends to vary over its full range within each level of the potential confounders adjusted for. Alternatively, specialized methods designed to be more robust to positivity violations (e.g., van der Laan and Gruber, 2010) could be incorporated into our proposed framework.

Additionally, a premise of the proposed approach is that it is conceivable to assign every vaccine recipient to have an immunologic biomarker set to a given value Inline graphic. Again, diagnostics may be helpful to examine this premise. For example, if immunocompromised participants tend to have low immune responses, it may be difficult to conceive of assigning their biomarker to the highest levels of Inline graphic. One way to address this problem would be to base CoP evaluation on the stochastic interventional risk curve (Hejazi and others, 2021), which sets each vaccine recipient’s biomarker to some random draw from a specified distribution that may be more plausible.

In our application, we relied upon the two-phase Cox regression, a commonly used method, as the basis of the analysis of the marginalized risk curve Inline graphic. Yet, given the need to control for confounding and minimize systematic bias, it is appealing to alternatively employ nonparametric doubly robust methods for estimating Inline graphic under minimal assumptions, along the lines of Westling and others (2021). However, additional research is needed to achieve this goal.

Prior to sensitivity analysis, our methods make the initial randomization and strong sequential ignorability assumptions (Figure 1), which adjust for baseline confounders Inline graphic of the effect of Inline graphic on Inline graphic. In some VE trials, postrandomization confounders Inline graphic are collected before Inline graphic is measured at Inline graphic. Section B of the Supplementary Material available at Biostatistics online describes how our methods can also be applied adjusting for both Inline graphic and Inline graphic using longitudinal G-computation.

Among surrogate endpoint evaluation approaches, the principal stratification approach may most closely resemble the controlled vaccine efficacy approach, because it estimates a VE curve across principal strata defined by the level Inline graphic of the immunologic biomarker if assigned vaccine, or a VE surface across principal strata defined by Inline graphic and Inline graphic if assigned vaccine or placebo (Gilbert and Hudgens, 2008), and these analyses produce seemingly similar outputs. However, the two approaches address different scientific objectives, with principal stratification assessing VE across “natural” subgroups without assignment of the marker, whereas controlled effects assign/control the marker level. Thus, an advantage of principal stratification is obviating the need to conceive assignment of a marker that was not randomly assigned, yet this implies the limitation of not fully addressing causal mediation of VE (e.g., VanderWeele, 2008). Consequently, while it is more challenging to conceptualize controlled vaccine efficacy, if this challenge is overcome, then inferences are more relevant for core applications such as bridging, which predicts efficacy for a new population or vaccine based on the marker.

Lastly, the science of the biological assays used to define immunologic biomarkers should be highlighted as fundamental to the establishment and utility of a CoP. Prespecified validation criteria are typically required for an immunologic biomarker to be accepted as a surrogate endpoint (FDA, 2018). Postacceptance, effective use of a surrogate endpoint requires that the biomarker be measured by the same lab that established the CoP, or that a new lab conducting the immunoassay have validated concordance of its assay compared to the original assay conducted by the original lab. Thus, standardization and validation of the immunoassay used to measure the CoP is a basic requirement for use of a CoP for approving/bridging vaccines.

Supplementary Material

kxac024_Supplementary_Data

Acknowledgments

We thank the participants, investigators, and sponsor (Sanofi Pasteur) of the CYD14 and CYD15 trials, and Lindsay Carpp for technical document contributions.

Conflict of Interest: Peter Gilbert and Youyi Fong have received salary support from Sanofi Pasteur contracts to their institution.

Contributor Information

Peter B Gilbert, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA and Department of Biostatistics, University of Washington, Seattle, WA, USA.

Youyi Fong, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA and Department of Biostatistics, University of Washington, Seattle, WA, USA.

Avi Kenny, Department of Biostatistics, University of Washington, Seattle, WA, USA.

Marco Carone, Fred Hutchinson Cancer Center, Vaccine and Infectious Disease Division, 1100 Fairview Ave N, PO Box 19024 Seattle, WA 98109, USA and University of Washington, Department of Biostatistics, Hans Rosling Center for Population Health, 3980 15th Avenue NE, Box 351617 Seattle, WA 98195-1617, USA.

Data and software

The CYD14 and CYD15 data are available upon request to Sanofi Pasteur. The computing code is available at the Github repository https:/github.com/youyifong/CoPcontrolledVE.

Supplementary Material

Supplementary material is available at http://biostatistics.oxfordjournals.org.

Funding

The National Institute Of Allergy and Infectious Diseases of the National Institutes of Health (UM1AI068635 and R37AI054165 to P.B.G.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

References

  1. Baden, L. R., ElSahly, H., Essink, B., Kotloff, K., Frey, S., Novak, R., Diemert, D., Spector, S. A., Rouphael, N., Creech, C. B.. and others. (2021). Efficacy and safety of the mRNA-1273 SARS-CoV-2 vaccine. New England Journal of Medicine 384, 403–416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Benkeser, D., Gilbert, P. B. and Carone, M. (2019). Estimating and testing vaccine sieve effects using machine learning. Journal of the American Statistical Association, 114, 1038–1049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Breslow, N. E. and Holubkov, R. (1997). Maximum likelihood estimation of logistic regression parameters under two-phase, outcome-dependent sampling. Journal of the Royal Statistical Society: Series B (Statistical Methodology) 59, 447–461. [Google Scholar]
  4. Capeding, M. R., Tran, N. H., Hadinegoro, S., Muhammad, I., Chotpitayasunondh, T., Chua, M. N., Luong, C. Q., Rusmil, K., Wirawan, D. N., Nallusamy, R.. and others. (2014). Clinical efficacy and safety of a novel tetravalent dengue vaccine in healthy children in Asia: a phase 3, randomised, observer-masked, placebo-controlled trial. The Lancet 384, 1358–1365. [DOI] [PubMed] [Google Scholar]
  5. Cowling, B. J., Lim, W. W., Perera, R. A., Fang, V. J., Leung, G. M., Peiris, J. M. and Tchetgen Tchetgen, E. J. (2019). Influenza hemagglutination-inhibition antibody titer as a mediator of vaccine-induced protection for influenza B. Clinical Infectious Diseases 68, 1713–1717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Ding, P. and VanderWeele, T. J. (2016). Sensitivity analysis without assumptions. Epidemiology 27, 368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. FDA, U.S. (2018). Bioanalytical method validation guidance for industry, US department of health and human services. Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Veterinary Medicine (CVM), Biopharmaceutics, 1–44. https://www.fda.gov/files/drugs/published/Bioanalytical-Method-Validation-Guidance-for-Industry.pdf [Google Scholar]
  8. Fleming, T. R. and Powers, J. H. (2012). Biomarkers and surrogate endpoints in clinical trials. Statistics in Medicine 31, 2973–2984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Follmann, D. (2006). Augmented designs to assess immune response in vaccine trials. Biometrics 62, 1161–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Gilbert, P. B. and Hudgens, M. G. (2008). Evaluating candidate principal surrogate endpoints. Biometrics 64, 1146–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Gilbert, P. B., Hudgens, M. G. and Wolfson, J. (2011). Commentary on “Inline graphic stratification – a goal or a tool?” by Inline graphic. The International Journal of Biostatistics 7, Article 36, 1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Gilbert, P. B., Montefiori, D. C., McDermott, A. B., Fong, Y., Benkeser, D., Deng, W., Zhou, H., Houchens, C. R., Martins, K., Jayashankar, L.. and others. (2022). Immune correlates analysis of the mRNA-1273 COVID-19 vaccine efficacy clinical trial. Science 375, 43–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Hejazi, N., van der Laan, M. J., Janes, H. E., Gilbert, P. B. and Benkeser, D. C. (2021). Efficient nonparametric inference on the effects of stochastic interventions under two-phase sampling, with applications to vaccine efficacy trials. Biometrics 77, 1241–1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Joffe, M.M. and Greene, T. (2009). Related causal frameworks for surrogate outcomes. Biometrics 65, 530–538. [DOI] [PubMed] [Google Scholar]
  15. Loux, T.M., Drake, C. and Smith-Gagen, J. (2017). A comparison of marginal odds ratio estimators. Statistical Methods in Medical Research 26, 155–175. [DOI] [PubMed] [Google Scholar]
  16. Molenberghs, G., Burzykowski, T., Alonso, A., Assam, P., Tilahum, A. and Buyse, M. (2008). The meta-analytic framework for the evaluation of surrogate endpoints in clinical trials. Journal of Statistical Planning and Inference 138, 432–449. [Google Scholar]
  17. Moodie, Z., Juraska, M., Huang, Y., Zhuang, Y., Fong, Y., Carpp, L.N., Self, S.G., Chambonneau, L., Small, R., Jackson, N.. and others. (2018). Neutralizing antibody correlates analysis of tetravalent dengue vaccine efficacy trials in Asia and Latin America. Journal of Infectious Diseases 217, 742–753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Pearl, J. (2001). Direct and Indirect Effects. San Francisco: Morgan Kaufmann. [Google Scholar]
  19. Plotkin, S. A. (2010). Correlates of protection induced by vaccination. Clinical Vaccine Immunology 17, 1055–1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Plotkin, S. A. and Gilbert, P. B. (2018). Correlates of protection. In: Plotkin, S., Orenstein, W., Offit, P. and Edwards, K. (editors), Vaccines, 7th edition. New York: Elsevier Inc., pp. 35–40. [Google Scholar]
  21. Prentice, R. L. (1986). A case-cohort design for epidemiologic cohort studies and disease prevention trials. Biometrika 73, 1–11. [Google Scholar]
  22. Prentice, R. L. (1989). Surrogate endpoints in clinical trials: definition and operational criteria. Statistics in Medicine 8, 431–440. [DOI] [PubMed] [Google Scholar]
  23. Price, B. L., Gilbert, P. B. and van der Laan, M. J. (2018). Estimation of the optimal surrogate based on a randomized trial. Biometrics 74, 1271–1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Qin, L., Gilbert, P. B., Corey, L., McElrath, J. and Self, S. G. (2007). A framework for assessing immunological correlates of protection in vaccine trials. The Journal of Infectious Diseases 196, 1304–1312. [DOI] [PubMed] [Google Scholar]
  25. Robins, J. M. and Greenland, S. (1992). Identifiability and exchangeability of direct and indirect effects. Epidemiology 3, 143–155. [DOI] [PubMed] [Google Scholar]
  26. Self, S. G. and Prentice, R. L. (1988). Asymptotic distribution theory and efficiency results for case-cohort studies. Annals of Statistics 16, 64–81. [Google Scholar]
  27. Son, H. and Fong, Y. (2021). Fast grid search and bootstrap-based inference for continuous two-phase polynomial regression models. Environmetrics 32, e2664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. US Food and Drug Administration. (1999). US Code of Federal Regulations FDA Subpart H – accelerated approval of new drugs for serious or life-threatening illnesses. secs. 314.500–314.560. 21 CFR. [Google Scholar]
  29. van der Laan, M. J. and Gruber, S. (2010). Collaborative double robust targeted maximum likelihood estimation. The International Journal of Biostatistics 6, Article 17, 1–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. VanderWeele, T. J. (2008). Simple relations between principal stratification and direct and indirect effects. Statistics and Probability Letters 78, 2957–2962. [Google Scholar]
  31. VanderWeele, T. J. (2013). Surrogate measures and consistent surrogates. Biometrics 69, 561–568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. VanderWeele, T. J. and Ding, P. (2017). Sensitivity analysis in observational research: introducing the E-value. Annals of Internal Medicine 167, 268–274. [DOI] [PubMed] [Google Scholar]
  33. VanderWeele, T. J. and Mathur, M. B. (2020). Commentary: developing best-practice guidelines for the reporting of E-values. International Journal of Epidemiology 49, 1495–1497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Vigne, C., Dupuy, M., Richetin, A., Guy, B., Jackson, N., Bonaparte, M., Hu, B., Saville, M., Chansinghakul, D., Noriega, F.. and others. (2017). Integrated immunogenicity analysis of a tetravalent dengue vaccine up to 4 years after vaccination. Human Vaccines & Immunotherapeutics 13, 2004–2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Villar, L., Dayan, G. H., Arredondo-García, J. L., Rivera, D. M., Cunha, R., Deseda, C., Reynales, H., Costa, M. S., Morales-Ramírez, J. O., Carrasquilla, G.. and others. (2015). Efficacy of a tetravalent dengue vaccine in children in Latin America. New England Journal of Medicine 372, 113–123. [DOI] [PubMed] [Google Scholar]
  36. Westling, T. and Carone, M. (2020). A unified study of nonparametric inference for monotone functions. Annals of Statistics 48, 1001–1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Westling, T., Luedtke, A., Gilbert, P. and Carone, M. (2021). Inference for treatment-specific survival curves using machine learning. arXiv preprint arXiv:2106.06602. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

kxac024_Supplementary_Data

Articles from Biostatistics (Oxford, England) are provided here courtesy of Oxford University Press

RESOURCES