Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-06-01T19:09:23.447Z Has data issue: false hasContentIssue false

Exploring human biology with N-of-1 clinical trials

Published online by Cambridge University Press:  10 January 2023

N. J. Schork*
Affiliation:
Department of Quantitative Medicine, The Translational Genomics Research Institute (TGen), Phoenix, AZ, USA Net.bio Inc., Los Angeles, CA, USA
B. Beaulieu-Jones
Affiliation:
Net.bio Inc., Los Angeles, CA, USA University of Chicago, Chicago, IL, USA
W. S. Liang
Affiliation:
Net.bio Inc., Los Angeles, CA, USA
S. Smalley
Affiliation:
Net.bio Inc., Los Angeles, CA, USA The University of California Los Angeles, Los Angeles, CA, USA
L. H. Goetz
Affiliation:
Department of Quantitative Medicine, The Translational Genomics Research Institute (TGen), Phoenix, AZ, USA Net.bio Inc., Los Angeles, CA, USA
*
Author for correspondence: N. J. Schork, Email: nschork@tgen.org
Rights & Permissions [Opens in a new window]

Abstract

Studies on humans that exploit contemporary data-intensive, high-throughput ‘omic’ assay technologies, such as genomics, transcriptomics, proteomics and metabolomics, have unequivocally revealed that humans differ greatly at the molecular level. These differences, which are compounded by each individual’s distinct behavioral and environmental exposures, impact individual responses to health interventions such as diet and drugs. Questions about the best way to tailor health interventions to individuals based on their nuanced genomic, physiologic, behavioral, etc. profiles have motivated the current emphasis on ‘precision’ medicine. This review’s purpose is to describe how the design and execution of N-of-1 (or personalized) multivariate clinical trials can advance the field. Such trials focus on individual responses to health interventions from a whole-person perspective, leverage emerging health monitoring technologies, and can be used to address the most relevant questions in the precision medicine era. This includes how to validate biomarkers that may indicate appropriate activity of an intervention as well as how to identify likely beneficial interventions for an individual. We also argue that multivariate N-of-1 and aggregated N-of-1 trials are ideal vehicles for advancing biomedical and translational science in the precision medicine era since the insights gained from them can not only shed light on how to treat or prevent diseases generally, but also provide insight into how to provide real-time care to the very individuals who are seeking attention for their health concerns in the first place.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2023. Published by Cambridge University Press

Impact statement

Individuals do not respond to health interventions in the same way. This creates a need for identifying what it is (e.g., a behavior, a gene, a biomarker, or their combinations) that may indicate which interventions should be provided to different individuals. In fact, a great deal of modern biomedical science has focused on the identification of the mechanisms that contribute to disease, and relevant research has revealed that most disease processes are indeed multifactorial and can differ substantially between individuals. However, only now are studies being pursued in earnest that seek to identify links between measurable factors and likely response to health interventions. In this light, studies designed to identify unequivocal individual responders and non-responders to health interventions are needed. Current approaches, specifically those involving large cohort-based clinical trials with single endpoints and a focus on average effects of an intervention, are not necessarily designed for this. Rather, emerging N-of-1 trial designs that focus on individual responses to an intervention by collecting enough data on a participant to statistically determine and quantify their responses are better suited for this. We provide the basic motivation and techniques used in N-of-1 studies, contrasting them with standard population-based clinical trials, and focus on directions in which the research community is going that could accelerate the use of strategies for providing health interventions to the individuals most likely to benefit from them. One key area where clinical studies of health interventions have fallen short is in limiting their focus on one health outcome or measure. It is underappreciated that what individuals put in their bodies may impact them in a wide variety of ways – both good and bad – and N-of-1 studies have the potential to help overcome this and thereby push the understanding of human biology in unprecedented ways.

Introduction

The rapid development of high-throughput, cost-efficient and data-intensive assays for use in molecular biology and the biomedical sciences (e.g., DNA sequencing, proteomics, metabolomics, etc.) is revolutionizing the manner in which studies are pursued by seeking a deeper understanding of the pathological processes underlying diseases of all sorts. The application of such technologies to, for example, explorations of the differences between diseased and non-diseased human tissue specimens or genome-wide association studies (GWAS) interrogating DNA collected on tens of thousands of individuals with and without a particular condition, has led to many very useful insights into how to combat diseases (Karczewski and Snyder, Reference Karczewski and Snyder2018). However, such investigations have also exposed one very complicated set of issues: most pathological processes underlying diseases are heterogeneous and nuanced, to the point where mechanisms contributing to disease in one individual may be different from those in another individual. Given this, it has also been shown that available treatments or preventive interventions for different diseases tend not to work in everyone with the same general diagnosis. These two facts have led to concerted efforts to promote ‘precision’ or ‘personalized’ medicine and nutrition whereby health interventions are tailored to the unique genomic, physiologic, clinical, behavioral and exposure profiles of individuals who could benefit from them (Ginsburg and Willard, Reference Ginsburg and Willard2016; Karczewski and Snyder, Reference Karczewski and Snyder2018; Zeggini et al., Reference Zeggini, Gloyn, Barton and Wain2019).

The two largest impediments to enabling and deploying precision medicine at scale are (1) simply not having a more complete understanding of human in vivo biology and (2) not having insight into whether the differences exhibited by individuals at the molecular level – that have largely been identified from in vitro or ex vivo studies of human tissues – are truly clinically meaningful. Comprehensive longitudinal evaluations of humans using state-of-the-field assays have been pursued, but they have focused on identifying patterns among individuals in their natural environments without any controlled perturbation or design to relevant data collections (Chen et al., Reference Chen, Mias, Li-Pook-Than, Jiang, Lam, Chen, Miriami, Karczewski, Hariharan, Dewey, Cheng, Clark, Im, Habegger, Balasubramanian, O’Huallachain, Dudley, Hillenmeyer, Haraksingh, Sharon, Euskirchen, Lacroute, Bettinger, Boyle, Kasowski, Grubert, Seki, Garcia, Whirl-Carrillo, Gallardo, Blasco, Greenberg, Snyder, Klein, Altman, Butte, Ashley, Gerstein, Nadeau, Tang and Snyder2012; Li-Pook-Than and Snyder, Reference Li-Pook-Than and Snyder2013; Price et al., Reference Price, Magis, Earls, Glusman, Levy, Lausted, McDonald, Kusebauch, Moss, Zhou, Qin, Moritz, Brogaard, Omenn, Lovejoy and Hood2017; Earls et al., Reference Earls, Rappaport, Heath, Wilmanski, Magis, Schork, Omenn, Lovejoy, Hood and Price2019; Schussler-Fiorenza Rose et al., Reference Schussler-Fiorenza Rose, Contrepois, Moneghetti, Zhou, Mishra, Mataraso, Dagan-Rosenfeld, Ganz, Dunn, Hornburg, Rego, Perelman, Ahadi, Sailani, Zhou, Leopold, Chen, Ashland, Christle, Avina, Limcaoco, Ruiz, Tan, Butte, Weinstock, Slavich, Sodergren, McLaughlin, Haddad and Snyder2019; Levy et al., Reference Levy, Magis, Earls, Manor, Wilmanski, Lovejoy, Gibbons, Omenn, Hood and Price2020; Sailani et al., Reference Sailani, Metwally, Zhou, Rose, Ahadi, Contrepois, Mishra, Zhang, Kidzinski, Chu and Snyder2020; Zimmer et al., Reference Zimmer, Korem, Rappaport, Wilmanski, Baloni, Jade, Robinson, Magis, Lovejoy, Gibbons, Hood and Price2021; Metwally et al., Reference Metwally, Zhang, Wu, Kellogg, Zhou, Contrepois, Tang and Snyder2022). Such studies are essential to explore human intra- and inter-individual variation but leave open the question of how different factors might contribute to different responses to health interventions (Atkinson and Batterham, Reference Atkinson and Batterham2015; Atkinson et al., Reference Atkinson, Williamson and Batterham2019; McInnes et al., Reference McInnes, Yee, Pershad and Altman2021). We note that there are examples of specific therapeutic modalities whose development is consistent with and motivated by a precision medicine orientation in the discussion section.

The purpose of this review is to provide an argument that clinical trials can be pursued that will allow researchers to probe human physiology in ethically-sound ways with unprecedented sophistication. Relevant trials should be rooted in N-of-1 and aggregated N-of-1 designs (Schork, Reference Schork2015; Nikles et al., Reference Nikles, Onghena, Vlaeyen, Wicksell, Simons, McGree and McDonald2021) and focus on exploring multiple phenotypes simultaneously and identifying causal relationships between phenotypes by leveraging emerging, largely non-invasive, health monitoring devices and assays (Izmailova et al., Reference Izmailova, Wagner and Perakslis2018; Bentley et al., Reference Bentley, Kleiman, Elliott, Huffman and Nock2019; Tehrani et al., Reference Tehrani, Teymourian, Wuerstle, Kavner, Patel, Furmidge, Aghavali, Hosseini-Toudeshki, Brown, Zhang, Mahato, Li, Barfidokht, Yin, Warren, Huang, Patel, Mercier and Wang2022). We do not provide an exhaustive review of N-of-1 trials, as there are many excellent resources and introductions to the basic motivation and methodologies (Lillie et al., Reference Lillie, Patay, Diamant, Issell, Topol and Schork2011; Nikles et al., Reference Nikles, Onghena, Vlaeyen, Wicksell, Simons, McGree and McDonald2021; Davidson et al., Reference Davidson, Cheung, Friel and Suls2022), including comprehensive reviews of the applications of N-of-1 trials (Gabler et al., Reference Gabler, Duan, Vohra and Kravitz2011; Li et al., Reference Li, Gao, Punja, Ma, Vohra, Duan, Gabler, Yang and Kravitz2016; Mirza et al., Reference Mirza, Punja, Vohra and Guyatt2017) as well as practical guides as to how to conduct N-of-1 trials (Guyatt et al., Reference Guyatt, Sackett, Adachi, Roberts, Chong, Rosenbloom and Keller1988; Kravitz et al., Reference Kravitz and Duan2014; Nikles et al., Reference Nikles, Onghena, Vlaeyen, Wicksell, Simons, McGree and McDonald2021; Duan et al., Reference Duan, Norman, Schmid, Sim and Kravitz2022). In fact, N-of-1 trials are now receiving attention as strategies for improving health care generally (Keller et al., Reference Keller, Guyatt, Roberts, Adachi and Rosenbloom1988; Senn, Reference Senn1998; Derby et al., Reference Derby, Kronish, Wood, Cheung, Cohn, Duan, St Onge, Duer-Hefele, Davidson and Moise2021; McDonald and Nikles, Reference McDonald and Nikles2021; Selker et al., Reference Selker, Cohen, D’Agostino, Dere, Ghaemi, Honig, Kaitin, Kaplan, Kravitz, Larholt, McElwee, Oye, Palm, Perfetto, Ramanathan, Schmid, Seyfert-Margolis, Trusheim and Eichler2022). Rather, we focus on N-of-1 trials that can address issues plaguing precision medicine and can provide a better understanding of human biology for at least four reasons: (1) They can provide unprecedented insights into human biology, including intra-individual causal claims about interventions and health measures. (2) They provide very comprehensive ways of vetting interventions to see if they work and for whom they work. (3) Their results provide insight into an individual’s health that may benefit them almost immediately, as opposed to much later after all relevant data have been collected and analyzed as part of a larger study. (4) Their results can be aggregated to explore patterns among individuals who exhibit robust responses to interventions. The organization of the review is as follows. We first provide greater insight into why legacy population-wide effect-focused randomized clinical trials (RCTs) are inadequate to address fundamental questions about human biology. We then consider different aspects of, and settings for, the proposed multivariate N-of-1 clinical trials, including the need for better markers of drug activity and availability. We end with a brief discussion of a few emerging therapeutic areas that could benefit from the proposed trials as well as suggestions for future research.

Human biology and legacy clinical trials

Strategies to understand how systems function as a whole, and which components may be dependent on other components, typically involve inducing perturbations to those systems and then determining how the systems respond (e.g., in cellular or mouse physiology studies). Studies seeking to perturb living humans systematically in this way are at worst unethical and at best logistically complicated. However, humans voluntarily subject themselves to perturbations of all sorts via pharmacologic interventions, dietary manipulations, environmental exposures, etc. In fact, clinical trials are routinely pursued to explore responses to such perturbations. Unfortunately, most clinical trials tend to focus on a singular indication (i.e., health or response measure) and the average response to the intervention in the population at large and therefore do not address broader questions about human physiology. We do not provide an in-depth review of clinical trials here (see, e.g., Friedman et al., Reference Friedman, Furburg, DeMets, Reboussin and Granger2015), but rather highlight a few of their key aspects so they can be contrasted with the N-of-1 studies. Typically, health interventions are evaluated in stages to ensure their safety and efficacy, from small (n = 5–20) phase I safety trials, to moderately sized (n = 25–200) phase II efficacy trials, to large (n = 250–10,000) phase III comparative and phase IV post-marketing surveillance studies. Some phase II and virtually all phase III and IV trials are pursued as RCTs where individuals are randomized to receive or not receive the intervention in question to avoid confounding. The health measures collected on these individuals are then compared to determine what effect the intervention may have on the typical person in the population at large.

There are at least six issues in the conduct of phase I–phase IV clinical trials (Deaton and Cartwright, Reference Deaton and Cartwright2018; Schork, Reference Schork2018) that motivate complementary N-of-1 trials: (1) Most standard clinical trials have inclusion and exclusion criteria to make sure the trial has been carried out in individuals likely to benefit, as well as for ensuring safety and avoiding confounding effects, which can complicate their generalizability. (2) Most, if not all, trials focus on the effect of an intervention on a single well-defined endpoint (e.g., such as blood pressure, pain, or rheumatoid arthritis symptoms). (3) Most failures of interventions in clinical trials testing occur in the phase II stage of testing; that is, despite being shown to have potential in ‘pre-clinical’ cellular and non-human experiments and to be safe in phase I trials, many interventions are shown not to modulate or affect the phenotype they were designed to impact, calling into question the pre-clinical, basic-science driven evidence suggesting that they may have benefit in humans in vivo (of course there are other reasons why an intervention may fail in a Phase II trial, for example, due to biased sampling, focusing on the wrong endpoint, measurement error, etc.). (4) Most late phase clinical trials, despite having inclusion and exclusion criteria, are expensive as they are conducted on very large numbers of people to ensure the trial results are generalizable and to overcome often hypothesized weak average effect sizes. (5) The results of clinical trials may identify interventions with the potential to benefit individuals, but unless it is known a priori how to identify individuals most likely to benefit from each intervention, it will be unclear how to optimally provide the interventions (see Figure 1). (6) Standard population-based RCTs can take a very long time to pursue and analyze, whereas more focused participant or patient-oriented alternative trial designs can be aggregated sequentially to enable population-based inferences (Schork, Reference Schork2022).

Figure 1. A tree or dendrogram reflecting how similar a number of individuals are with respect to phenotypes of relevance to drug response: the closer the bottommost branches of Figure 1 are – which represent individuals – the more similar the phenotypic profiles of those individuals are. The darkness of the shaded human figures at the bottom of the figure at different positions in the tree reflects the degree to which individuals at those positions in the tree possess a certain characteristic or profile. The circles represent interventions that can benefit different groups of individuals, such that the different locations where the shaded circles are situated represent convergence points for all individuals connected beneath that point who can benefit from the specific intervention. Thus, the topmost circle indicates that all individuals may benefit from that intervention (since all the individual tree branches converge back to that point), whereas the leftmost circle is likely to benefit the first ~25–30% of individuals. The two circles second and third from the left indicate interventions that may benefit a small number of individuals (e.g., only ~10% of individuals). The circle to which the arrow is pointing indicates an intervention that may benefit a large number of individuals but for whom other interventions (reflected by the 5th and 6th circles from the right) may benefit smaller subsets of individuals. Identifying points on trees like this that are consistent with who benefits from an intervention based on understanding of the factors responsible for mediating response is the motivation behind precision medicine and nutrition.

Basic N-of-1 trial designs

Basic designs

As emphasized, the ultimate goal of N-of-1 trials is to determine, in an appropriately powered way, if an intervention is actually benefitting a target individual by leveraging data collections and analytical methods focused on that target individual’s response. An element common to all N-of-1 clinical trial designs is an intervention ‘crossover’ component in which measurements on a health-related phenotype (e.g., blood pressure, mood, weight, symptoms, etc.) are made while the target individual is receiving, and not receiving, an intervention. This contrast between measures while on and off the intervention can then be exploited to quantify and characterize the individual’s response to the intervention but only if enough reliable measurements are made during each of the intervention periods and data analysis methods are used to control for confounding due to, for example, placebo or unmeasured covariate effects (Lillie et al., Reference Lillie, Patay, Diamant, Issell, Topol and Schork2011; Kravitz et al., Reference Kravitz and Duan2014; Wang and Schork, Reference Wang and Schork2019; Kravitz and Duan, Reference Kravitz and Duan2022). Note that many of the most widely used strategies for avoiding confounding in standard RCTs can be exploited in the design and execution of N-of-1 trials, such as randomizing the order in which the interventions are provided, blinding of the received interventions to the participants and/or researchers analyzing the data, washout periods to avoid carryover effects, etc. (Lillie et al., Reference Lillie, Patay, Diamant, Issell, Topol and Schork2011; Duan et al., Reference Duan, Kravitz and Schmid2013; Kravitz et al., Reference Kravitz and Duan2014; Duan et al., Reference Duan, Norman, Schmid, Sim and Kravitz2022; Kravitz and Duan, Reference Kravitz and Duan2022).

Figure 2 depicts some basic N-of-1 designs. We note that there is growing, but not complete, consensus on the definition of an N-of-1 clinical trial – which many believe requires a randomized order of interventions with, for example, blinding – as opposed to a simple ‘single case study’ which may not include randomization or blinding. We argue that both N-of-1 clinical trials and some single case studies are appropriate for advancing precision medicine (Davidson et al., Reference Davidson, Cheung, Friel and Suls2022) and consider them both as N-of-1 clinical trials. Panel A depicts the simple and often used ‘interrupted time series single case design’ – or basic ‘AB’ design, where ‘A’ and ‘B’ correspond to interventions, one of which could be a placebo or simply no intervention (see, e.g., part V of the book by Huitema, Reference Huitema2011 for an excellent introduction). Panel B depicts the ‘reversal’ or ‘ABAB’ design in which the intervention periods in the interrupted time series design are repeated to ensure the initial set of observations do not reflect false positive or negative results. Panel C depicts the reversal design with washout periods (i.e., periods where no administration of an intervention, including a placebo, are provided) between each administration of an intervention to avoid confounding carryover effects (an ‘AwBwAwB’ design). Note that the number of intervention administration periods and the order of the interventions can vary depending on the sophistication of the design (e.g., ‘ABwBA’ or ‘AwAwBwAwBwBwA’).

Figure 2. Different, very basic, types of N-of-1 clinical trial designs in which an intervention had a lowering effect on a health measure (like blood pressure). The black and red lines reflect hypothetical health measure trajectories (i.e., longitudinal data) while an individual is not receiving (black) or receiving (red) an intervention. The vertical dashed lines indicate when interventions were provided or changed. Panel A depicts the basic ‘interrupted time’ series design, Panel B the ‘reversal’ design and panel C a reversal design with washout periods (green lines).

The power of N-of-1 trials

N-of-1 trials derive their power to make inferences about the effect of an intervention on an individual from the number of measurements made on the participant while on and off an intervention (Huitema, Reference Huitema2011). However, serial correlations between the measurements can complicate the analysis if not appropriately accounted for, as can aforementioned covariate effects, carryover effects, missing data, non-uniform time points between measurement collections and placebo effects (Rochon, Reference Rochon1990; Huitema, Reference Huitema2011; Lillie et al., Reference Lillie, Patay, Diamant, Issell, Topol and Schork2011; Wang and Schork, Reference Wang and Schork2019; Somer et al., Reference Somer, Gische and Miocevic2022). Many offshoots of N-of-1 trials exist to improve their efficiency and comprehensiveness; for example, sequential designs can be used to minimize the number of measurements made while preserving appropriate false positive and false negative rates (Schork and Goetz, Reference Schork and Goetz2017; Schork, Reference Schork2022). In addition, there is no reason that N-of-1 trial methodology cannot be used in other settings, for example, assessing intervention effects in cell lines, tissue samples, mice, etc. In fact, such studies often make use of samples from a single individual or strain of mice and so, from a biological standpoint, they are, by their nature, assuming that insights from a single individual can shed light on very general biological questions. There are many recent examples of N-of-1 studies, which we will not review exhaustively here (Gabler et al., Reference Gabler, Duan, Vohra and Kravitz2011; Kronish et al., Reference Kronish, Hampsey, Falzon, Konrad and Davidson2018; Nikles et al., Reference Nikles, Evans, Hams and Sterling2022; Samuel et al., Reference Samuel, Wootton, Holder and Molony2022), but rather simply emphasize that they are growing in number and sophistication (Kim et al., Reference Kim, Hu, El Achkar, Black, Douville, Larson, Pendergast, Goldkind, Lee, Kuniholm, Soucy, Vaze, Belur, Fredriksen, Stojkovska, Tsytsykova, Armant, DiDonato, Choi, Cornelissen, Pereira, Augustine, Genetti, Dies, Barton, Williams, Goodlett, Riley, Pasternak, Berry, Pflock, Chu, Reed, Tyndall, Agrawal, Beggs, Grant, Urion, Snyder, Waisbren, Poduri, Park, Patterson, Biffi, Mazzulli, Bodamer, Berde and Yu2019; Lamb et al., Reference Lamb, Stone, D’Adamo, Volkov, Metti, Aronica, Minich, Leary, Class, Carullo, Ryan, Larson, Lundquist, Contractor, Eck, Ordovas and Bland2022; Phyland et al., Reference Phyland, McKay, Olver, Walterfang, Hopwood, Ponsford and Ponsford2022).

Beyond the basics

There are three important aspects of N-of-1 trials that are receiving the attention which are motivating newer approaches. First, the data and results associated with individual N-of-1 trials can be aggregated and analyzed to explore trends among the participants and their responses (Zucker et al., Reference Zucker, Ruthazer and Schmid2010; Araujo et al., Reference Araujo, Julious and Senn2016; Punja et al., Reference Punja, Xu, Schmid, Hartling, Urichuk, Nikles and Vohra2016; Schork and Goetz, Reference Schork and Goetz2017; Barbosa Mendes et al., Reference Barbosa Mendes, Jamshidi, Van den Noortgate and Fernandez-Castilla2022). Second, with sufficient data collected over time, one could characterize causal relationships among the intervention and other measures (Molenaar, Reference Molenaar2019; Izem and McCarter, Reference Izem and McCarter2021; Yeboah et al., Reference Yeboah, Mauer, Hufstedler, Carr, Matthay, Maxwell, Rahman, Debray, de Jong, Campbell, Gustafson, Janisch and Barnighausen2021) (note: an entire recent issue of the journal ‘Evaluation and the Health Professions’ was devoted to causal analysis in N-of-1 trials (Miocevic et al., Reference Miocevic, Moeyaert, Mayer and Montoya2022). Such analyses could provide unprecedented insight into human physiology. The third is that the execution of N-of-1 trials focusing on important physiologic endpoints can be greatly enhanced with emerging digital health-based monitoring devices (such as the Apple Watch and continuous glucose monitors), survey instruments made available through smartphone apps, and largely pain-free and convenient methods for obtaining blood, urine, stool and saliva samples (Enderle et al., Reference Enderle, Foerster and Burhenne2016; Izmailova et al., Reference Izmailova, Wagner and Perakslis2018).

Multivariate n-of-1 trials

N-of-1 clinical trials can be pursued to characterize the effect of an intervention on a specific phenotype (blood pressure) for a target individual and as such complement population-based RCTs, especially when it is unclear if an individual is likely to benefit from the intervention. However, many diseases are not associated with singular phenotypes and, in fact, most individuals who suffer from them do not only have one major symptom or problem (Ong et al., Reference Ong, Lee and Lee2020). This is especially the case for older individuals with many comorbidities (Pearson-Stuttard et al., Reference Pearson-Stuttard, Ezzati and Gregg2019; Onder et al., Reference Onder, Bernabei, Vetrano, Palmer and Marengoni2020; Skou et al., Reference Skou, Mair, Fortin, Guthrie, Nunes, Miranda, Boyd, Pati, Mtenga and Smith2022). As a result, it makes sense to pursue appropriately powered N-of-1 trials that explore the impact of an intervention on more than one outcome (i.e., multivariate N-of-1 trials). Although multivariate trials have been proposed in the context of standard RCTs, there are few, if any, precedents in N-of-1 study contexts (Zhao et al., Reference Zhao, Hu and Lagakos2009). Few published precision medicine studies have measured more than one clinically relevant health measure despite the availability of newer health monitoring technologies (Viana et al., Reference Viana, Edney, Gondalia, Mauch, Sellak, O’Callaghan and Ryan2021). Although we will not go into the mathematical or statistical details here for how such trials can achieve sufficient power, it is arguable that if health is defined broadly (e.g., normal blood pressure, quality sleep, good blood biochemistry profile, etc.) then a good health intervention should at a minimum not negatively affect any of them and at best positively affect them all. In this light, testing multiple measures for intervention effects simultaneously using an omnibus statistical test of the hypothesis that an intervention positively effects them all could lead to an increase in power (Huitema, Reference Huitema2011; Tabachnick and Fidell, Reference Tabachnick and Fidell2012), but only if the number of measures is large (Leroy et al., Reference Leroy, Frongillo, Kase, Alonso, Chen, Dohoo, Huybregts, Kadiyala and Saville2022). Reaching appropriate numbers of observations could be achieved, for example, through the use of the aforementioned continuous wireless devices or microsampling techniques which involve collecting minute amounts of blood or urine for analyses to avoid a standard blood draw or logistically challenging biospecimen collections (Enderle et al., Reference Enderle, Foerster and Burhenne2016; Bentley et al., Reference Bentley, Kleiman, Elliott, Huffman and Nock2019; Anderson et al., Reference Anderson, Razavi, Pope, Yip, Cameron, Bassini-Cameron and Pearson2020).

There are many settings beyond multimorbidity issues that justify an evaluation of multiple health measures in N-of-1 clinical trials. For example, depression is known to impact virtually all aspects of a person’s health due to the various behaviors adopted by depressed individuals (Triolo et al., Reference Triolo, Harber-Aschan, Murri, Calderon-Larranaga, Vetrano, Sjoberg, Marengoni and Dekhtyar2020; Aprahamian et al., Reference Aprahamian, Borges, Hanssen, Jeuring and Oude Voshaar2022). Testing the effect of an antidepressant on mood and depressive symptoms in addition to, perhaps, weight, blood pressure, sleep quality, etc. makes sense. Another example involves geroprotectors, or interventions meant to slow the aging rate and thereby influence susceptibility to, or processes associated with, many different age-related diseases (Mahmoudi et al., Reference Mahmoudi, Xu and Brunet2019; Kritchevsky and Justice, Reference Kritchevsky and Justice2020; Triolo et al., Reference Triolo, Harber-Aschan, Murri, Calderon-Larranaga, Vetrano, Sjoberg, Marengoni and Dekhtyar2020; Aprahamian et al., Reference Aprahamian, Borges, Hanssen, Jeuring and Oude Voshaar2022; Moskalev et al., Reference Moskalev, Guvatova, Lopes, Beckett, Kennedy, De Magalhaes and Makarov2022). Thus, by definition, a geroprotector should affect multiple systems and hence could be tested for this. In fact, if only one or some subset of health measures among many different measures is in fact affected by a purported geroprotector, then the intervention is probably not a geroprotector (Schork et al., Reference Schork, Beaulieu-Jones, Liang, Smalley and Goetz2022).

In addition to testing for the effect of an intervention on multiple health measures, N-of-1 and aggregated N-of-1 studies can be pursued to exploit interventions as ways of perturbing or probing human physiology – the goal being to identify relationships among different health measures or processes. Thus, if enough measures are collected over the time an individual is both receiving and not receiving an intervention, then temporal relationships between the measures can reveal likely causal relationships among them based on, for example, time series analysis, Granger regression and other techniques (McCracken, Reference McCracken2016; Molenaar, Reference Molenaar2019). Such analyses would again be significantly enhanced if the relevant health measures were collected continuously (Enderle et al., Reference Enderle, Foerster and Burhenne2016; Bentley et al., Reference Bentley, Kleiman, Elliott, Huffman and Nock2019; Anderson et al., Reference Anderson, Razavi, Pope, Yip, Cameron, Bassini-Cameron and Pearson2020). In addition, by assessing the effect of the intervention on health measures beyond a primary measure in relevant trials, potential intervention ‘repurposing’ opportunities could arise (Pushpakom et al., Reference Pushpakom, Iorio, Eyers, Escott, Hopper, Wells, Doig, Guilliams, Latimer, McNamee, Norris, Sanseau, Cavalla and Pirmohamed2019; Krishnamurthy et al., Reference Krishnamurthy, Grimshaw, Axson, Choe and Miller2022; Mucke, Reference Mucke2022). In this way, N-of-1 trials can be pursued as proof-of-concept studies for identifying multiple indications, or at least one on solid footing, for an intervention (Pushpakom et al., Reference Pushpakom, Iorio, Eyers, Escott, Hopper, Wells, Doig, Guilliams, Latimer, McNamee, Norris, Sanseau, Cavalla and Pirmohamed2019; Mucke, Reference Mucke2022). In addition, by collecting multiple health measures on an individual N-of-1 trial participant, possibly continuously and in real time, insights into that participant’s health and health trajectory can be obtained even if an intervention being tested is shown not to benefit the participant.

Whole body, biomarker validation and therapeutic drug monitoring studies

There are some very specific areas where multivariate N-of-1 trials can be pursued that will enhance the assessment of individual intervention response and enable deeper insight into human physiology, as emphasized throughout this review. We briefly describe four such areas below.

General assessment of inter-individual variation in intervention response

As noted, given that N-of-1 trials focus on individuals’ responses, they can be used to more precisely identify responders to particular interventions. In addition, if relevant studies collected sufficient data on more than one health measure then they can be used to identify potential side effects, alternative uses for the intervention and different mechanisms of action or physiological processes modulated by the intervention. In fact, it might make sense for all interventions to be evaluated for their whole-body effects in a small number of individuals as they are being developed. If done along the lines outlined in the review, such trials could shed enormous light on how substances put into the human body affect it systemically (see Figure 3).

Figure 3. Contrasting clinical trial designs. The design depicted on the left is consistent with standard RCTs focusing on a singular health measure or indication (the gray colored dot on the left side of the head of the human figures indicating a single phenotype of interest; for example, depression symptoms). If individuals are found not to respond (NR = Non-Responders) then a future study seeking to identify biomarkers of response could be pursued, whereby a new biomarker phenotype is associated with the response/non-response phenotype (e.g., genomic profile). The design depicted on the right provides the motivation for complementary trials to traditional RCTs whereby the effect of an intervention is evaluated on an individual from a whole-body perspective. The results of this trial are aggregated with trials on other individuals and patterns that could identify responders and non-responders are explored that may also reveal intervention effects on different phenotypes and how those phenotypes interact.

Biomarker and surrogate endpoint validation

There is great interest in identifying better biomarkers of an intervention’s activity so that these biomarkers can be correlated with other health measures of interest (see, e.g., ‘Therapeutic Drug Monitoring Studies’ section below) (Hendrickson et al., Reference Hendrickson, Thomas, Schork and Raskind2020). In addition, there is also interest in identifying ‘surrogate endpoints’ for clinical trials that initially focus on expensive, lengthy and logistically challenging health outcome measures, and N-of-1 trials are excellent vehicles for validating biomarkers and surrogate endpoints (Burzykowski et al., Reference Burzykowski, Molenberghs and Buyse2005). As an example, consider the development and use of epigenetic clocks as surrogate endpoints in trials of geroprotectors (Schork et al., Reference Schork, Beaulieu-Jones, Liang, Smalley and Goetz2022). The belief is that if an intervention modulates or changes an epigenetic clock among participants in a trial in positive ways – thereby indicating that the intervention in question is slowing the aging rate of the individuals – then those individuals do not necessarily have to be tracked longitudinally until they develop (or do not develop) age-related diseases that the candidate geroprotector is hypothesized to prevent or treat (Mahmoudi et al., Reference Mahmoudi, Xu and Brunet2019; Kritchevsky and Justice, Reference Kritchevsky and Justice2020; Schork et al., Reference Schork, Beaulieu-Jones, Liang, Smalley and Goetz2022). Thus, the epigenetic clocks would act as a surrogate endpoint for the processes that are associated with the disease endpoints of real interest, which are modulated by the intervention. Although epigenetic clocks have been shown to be correlated with disease endpoints, they have been done so via large epidemiological studies and not in focused clinical trials measuring appropriate health measures. Therefore, it is arguable that by measuring epigenetic clocks along with health measures that underlie many common chronic age-related diseases and conditions, such as blood pressure, cholesterol level, sleep quality, etc. in appropriately powered N-of-1 trials, one might not only show that the geroprotector influences these health measures in positive ways, but also that an epigenetic clock is correlated with them as well. This would in effect validate surrogacy of the epigenetic clock at the ‘level of the individuals and the trial’ (Burzykowski et al., Reference Burzykowski, Molenberghs and Buyse2005; Buyse et al., Reference Buyse, Saad, Burzykowski, Regan and Sweeney2022).

Therapeutic drug monitoring studies

Therapeutic drug monitoring (TDM) studies consider the measurement of a drug’s concentration in an individual’s bloodstream in order to correlate the levels of the drug with the phenotype that the drug is hypothesized to modulate (Dasgupta, Reference Dasgupta2012; Clarke and Dasgupta, Reference Clarke and Dasgupta2019). Most drugs do not undergo such evaluation and testing, which is unfortunate since such studies could in theory better characterize mechanisms of action of the drug and its effects on different phenotypic endpoints. Of course, TDM studies are predicated on the assumption that there is a definable relationship between drug dose and plasma or blood drug concentration, and between concentrations and therapeutic effects. In addition, TDM studies require ways of measuring blood levels of a drug which may not be trivial. However, by more precisely measuring drug bioavailability and activity in N-of-1 trials, especially in trials for which participants are monitored for multiple health measures, one could explore temporal relationships between drug bioavailability and activity and not just, for example, pill count-based dosing and outcomes (Dasgupta, Reference Dasgupta2012; Clarke and Dasgupta, Reference Clarke and Dasgupta2019; Irving and Gecse, Reference Irving and Gecse2022; Ordutowski et al., Reference Ordutowski, Dal Dosso, De Wispelaere, Van Tricht, Vermeire, Geukens, Gils, Spasic and Lammertyn2022).

Matching based on data aggregation

As noted previously, if enough N-of-1 trials are pursued using the same interventions, and baseline health assessments with common measures have been collected on each participant, then the data and results can be aggregated and analyzed. The common baseline health examination profiles of the individuals could then be explored for patterns and correlations with intervention responses. This can enable matching a future target individual’s baseline health profile with others’ profiles who previously went through N-of-1 trials. If good matches (however defined) are found, then the interventions to which those individuals matching the target individual responded, would be reasonable first-choice interventions for the target individual (Wicks et al., Reference Wicks, Vaughan, Massagli and Heywood2011; Schork and Goetz, Reference Schork and Goetz2017; Schork et al., Reference Schork, Goetz, Lowey and Trent2020; Davidson et al., Reference Davidson, Cheung, Friel and Suls2022). Different strategies for identifying the matches could be pursued based on, for example, propensity scores and related techniques (Guo and Fraser, Reference Guo and Fraser2014; Liu and Meng, Reference Liu and Meng2016).

Conclusions and future directions

There are few health interventions whose effectiveness is ubiquitous. This can be attributed to the great genetic, physiologic, clinical, behavioral and exposure profile variation exhibited by individuals susceptible to or suffering from diseases (Schork, Reference Schork2015). Identifying interventions that benefit individuals on the basis of their nuanced and possibly unique profiles is the goal of precision or personalized medicine. However, tailoring or matching interventions to individuals will require greater understanding of intra- and inter-individual variation and intervention response and, as argued throughout, can be enabled or enhanced through the use of whole-body N-of-1 clinical trials (Figures 1 and 3).

In this light, many emerging interventions, such as cytotoxic T-cell therapies (Kiyotani et al., Reference Kiyotani, Toyoshima and Nakamura2021; Roesler and Anderson, Reference Roesler and Anderson2022), brain anatomy-guided Transcranial Magnetic Stimulation (TMS) therapies (Siddiqi et al., Reference Siddiqi, Weigand, Pascual-Leone and Fox2021; Williams et al., Reference Williams, Coman, Stetz, Walker, Kozel, George, Yoon, Hack, Madore, Lim, Philip and Holtzheimer2021) and sequence-based antisense oligonucleotide therapies (Kim et al., Reference Kim, Hu, El Achkar, Black, Douville, Larson, Pendergast, Goldkind, Lee, Kuniholm, Soucy, Vaze, Belur, Fredriksen, Stojkovska, Tsytsykova, Armant, DiDonato, Choi, Cornelissen, Pereira, Augustine, Genetti, Dies, Barton, Williams, Goodlett, Riley, Pasternak, Berry, Pflock, Chu, Reed, Tyndall, Agrawal, Beggs, Grant, Urion, Snyder, Waisbren, Poduri, Park, Patterson, Biffi, Mazzulli, Bodamer, Berde and Yu2019; Helm et al., Reference Helm, Schols and Hauser2022), are designed to only work on specific individuals given that the targets they exploit and constructs they use are based on the unique features underlying the pathologies of the individuals for whom they are designed. Testing the effectiveness of these interventions, given that no two individuals with the same condition will likely get exactly the same intervention, could make use of the proposed N-of-1 strategies. Of course, one could address very broad questions about the utility of such interventions using standard RCTs, such as whether individuals who receive the personalized interventions fare better than individuals who receive a more ‘one-size-fits-all’ intervention (Schork et al., Reference Schork, Goetz, Lowey and Trent2020).

Ultimately, the current emphasis on precision medicine, the emergence of sophisticated health monitoring technologies, and the desire of individuals to optimize their health and not simply contribute to studies that may only benefit future generations, demand better approaches to biomedical and translational science. We recognize that there might be impediments to the implementation of multivariate N-of-1 trials of the type described. For example, a greater patient burden for data collection, logistical complications in collecting different data types, and the costs of conducting and monitoring the individual participants may create barriers to the adoption and use of multivariate N-of-1 trials. However, efficient, cost-effective and participant-friendly N-of-1 clinical trials – to the degree that they can be pursued – are very likely to be an appropriate addition to biomedical and translational studies in the future given that they have at least 4 very overt advantages, including: (1) the ability to shed light on fundamental questions about human biology; (2) determine which interventions work and on whom; (3) benefit the participants in the trials directly and almost immediately by collecting vast amounts of health data on them possibly continuously and with real-time interpretive ability; and (4) pave the way for their aggregation and analysis to identify patterns that may inform their use and execution in the future.

Open peer review

To view the open peer review materials for this article, please visit http://doi.org/10.1017/pcm.2022.15.

Acknowledgements

The authors would like to thank Drs. Mark Adler and Stephanie Venn Watson for commenting on earlier versions of this manuscript.

Author contributions

N.J.S. conceived of the orientation and format for the review, and N.J.S., B.B.-J., W.S.L., S.S. and L.H.G. pursued relevant literature reviews. N.J.S. wrote the initial draft, L.H.G. edited the initial draft and B.B.-J., W.S.L. and S.S. edited the subsequent drafts.

Financial support

N.J.S. is supported in part by the following grant support from the National Institutes of Health: 1 U19 AG056169-01A1; U2C CA252973; UH3 AG064706; UH2 AG06470602S1 and U19 AG023122.

Competing interest

N.J.S., S.S. and L.H.G. are founders of net. Bio, a company focusing on pursuing novel clinical protocols to ensure that individuals benefit from health interventions of all sorts. B.B.-J. and W.S.L. are paid consultants for net. Bio.

References

Anderson, L, Razavi, M, Pope, ME, Yip, R, Cameron, LC, Bassini-Cameron, A and Pearson, TW (2020 ) Precision multiparameter tracking of inflammation on timescales of hours to years using serial dried blood spots. Bioanalysis 12, 937955.CrossRefGoogle Scholar
Aprahamian, I, Borges, MK, Hanssen, DJC, Jeuring, HW and Oude Voshaar, RC (2022 ) The frail depressed patient: A narrative review on treatment challenges Clinical Interventions in Aging 17, 979990.CrossRefGoogle Scholar
Araujo, A, Julious, S and Senn, S (2016) Understanding variation in sets of N-of-1 trials. PLoS One 11, e0167167.CrossRefGoogle ScholarPubMed
Atkinson, G and Batterham, AM (2015 ) True and false interindividual differences in the physiological response to an intervention. Experimental Physiology 100, 577588.CrossRefGoogle ScholarPubMed
Atkinson, G, Williamson, P and Batterham, AM (2019) Issues in the determination of ‘responders’ and ‘non-responders’ in physiological research. Experimental Physiology 104, 12151225.CrossRefGoogle ScholarPubMed
Barbosa Mendes, A, Jamshidi, L, Van den Noortgate, W and Fernandez-Castilla, B (2022) Network meta-analysis for single-case design studies: An illustration. Evaluation and the Health Professions 45, 6675.CrossRefGoogle ScholarPubMed
Bentley, KH, Kleiman, EM, Elliott, G, Huffman, JC and Nock, MK (2019) Real-time monitoring technology in single-case experimental design research: Opportunities and challenges. Behavior Research and Therapy 117, 8796.CrossRefGoogle ScholarPubMed
Burzykowski, T, Molenberghs, G and Buyse, M (eds.) (2005) The Evaluation of Surrogate Endpoints. New York: Springer.CrossRefGoogle Scholar
Buyse, M, Saad, ED, Burzykowski, T, Regan, MM and Sweeney, CS (2022 ) Surrogacy beyond prognosis: The importance of “trial-level” surrogacy. The Oncologist 27, 266271.CrossRefGoogle ScholarPubMed
Chen, R, Mias, GI, Li-Pook-Than, J, Jiang, L, Lam, HY, Chen, R, Miriami, E, Karczewski, KJ, Hariharan, M, Dewey, FE, Cheng, Y, Clark, MJ, Im, H, Habegger, L, Balasubramanian, S, O’Huallachain, M, Dudley, JT, Hillenmeyer, S, Haraksingh, R, Sharon, D, Euskirchen, G, Lacroute, P, Bettinger, K, Boyle, AP, Kasowski, M, Grubert, F, Seki, S, Garcia, M, Whirl-Carrillo, M, Gallardo, M, Blasco, MA, Greenberg, PL, Snyder, P, Klein, TE, Altman, RB, Butte, AJ, Ashley, EA, Gerstein, M, Nadeau, KC, Tang, H and Snyder, M (2012) Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell 148, 12931307.CrossRefGoogle ScholarPubMed
Clarke, W and Dasgupta, A (2019) Clinical Challenges in Therapeutic Drug Monitoring: Special Populations, Physiological Conditions and Pharmacogenomics. Amsterdam: Elsevier.Google Scholar
Dasgupta, A (2012) Therapeutic Drug Monitoring: Newer Drugs and Biomarkers. Cambridge, MA: Academic Press.CrossRefGoogle Scholar
Davidson, K, Cheung, K, Friel, C and Suls, J (2022) Introducing data sciences to N-of-1 designs, statistics, use-cases, the future, and the moniker ‘N-of-1’ Trial. Harvard Data Science Review, (Special Issue 3). https://doi.org/10.1162/99608f92.116c43feCrossRefGoogle Scholar
Deaton, A and Cartwright, N (2018) Understanding and misunderstanding randomized controlled trials. Social Science and Medicine 210, 221.CrossRefGoogle ScholarPubMed
Derby, L, Kronish, IM, Wood, D, Cheung, YKK, Cohn, E, Duan, N, St Onge, T, Duer-Hefele, J, Davidson, KW and Moise, N (2021). Using a multistakeholder collaboratory and patient surveys to inform the conduct of personalized (N-of-1) trials. Health Psychology 40, 230241.CrossRefGoogle ScholarPubMed
Duan, N, Kravitz, RL and Schmid, CH (2013) Single-patient (n-of-1) trials: A pragmatic clinical decision methodology for patient-centered comparative effectiveness research. Journal of Clinical Epidemiology 66, S21S28.CrossRefGoogle ScholarPubMed
Duan, N, Norman, D, Schmid, C, Sim, I and Kravitz, RL (2022) Personalized data science and personalized (N-of-1) trials: Promising paradigms for individualized health care. Harvard Data Science Review, (Special Issue 3). https://doi.org/10.1162/99608f92.8439a336CrossRefGoogle Scholar
Earls, JC, Rappaport, N, Heath, L, Wilmanski, T, Magis, AT, Schork, NJ, Omenn, GS, Lovejoy, J, Hood, L and Price, ND (2019) Multi-omic biological age estimation and its correlation with wellness and disease phenotypes: A longitudinal study of 3,558 individuals. Journals of Gerontology Series A: Biological Sciences and Medical Sciences 74, S52S60.CrossRefGoogle ScholarPubMed
Enderle, Y, Foerster, K and Burhenne, J (2016) Clinical feasibility of dried blood spots: Analytics, validation, and applications. Journal of Pharmaceutical and Biomedical Analysis 130, 231243.CrossRefGoogle ScholarPubMed
Friedman, LM, Furburg, CD, DeMets, DL, Reboussin, DR and Granger, CB (2015) Fundamentals of Clinical Trials. New York: Springer.CrossRefGoogle Scholar
Gabler, NB, Duan, N, Vohra, S and Kravitz, RL (2011) N-of-1 trials in the medical literature: A systematic review. Medical Care 49(8), 761.CrossRefGoogle ScholarPubMed
Ginsburg, GS and Willard, HF (ed.) (2016) Genomic and Precision Medicine: Foundations, Translation, and Implementation. London: Academic Press.Google Scholar
Guo, S and Fraser, MW (2014) Propensity Score Analysis: Statistical Methods and Applications. Thousand Oaks, CA: SAGE Publications.Google Scholar
Guyatt, G, Sackett, D, Adachi, J, Roberts, R, Chong, J, Rosenbloom, D and Keller, J (1988) A clinician’s guide for conducting randomized trials in individual patients. Canadian Medical Association Journal 139, 497503.Google ScholarPubMed
Helm, J, Schols, L and Hauser, S (2022) Towards personalized allele-specific antisense oligonucleotide therapies for toxic gain-of-function neurodegenerative diseases. Pharmaceutics 14, 1708.CrossRefGoogle ScholarPubMed
Hendrickson, RC, Thomas, RG, Schork, NJ and Raskind, MA (2020) Optimizing aggregated N-Of-1 trial designs for predictive biomarker validation: Statistical methods and theoretical findings. Frontiers in Digital Health 2, 13.CrossRefGoogle ScholarPubMed
Huitema, BE (2011) The Analysis of Covariance and Alternatives: Statistical Methods for Experiments, Quasi-Experiments, and Single-Case Studies. New Jersey: Wiley.CrossRefGoogle Scholar
Irving, PM and Gecse, KB (2022 ) Optimizing therapies using therapeutic drug monitoring: Current strategies and future perspectives. Gastroenterology 162, 15121524.CrossRefGoogle ScholarPubMed
Izem, R and McCarter, R (2021) Randomized and non-randomized designs for causal inference with longitudinal data in rare disorders. Orphanet Journal of Rare Diseases 16, 491.CrossRefGoogle ScholarPubMed
Izmailova, ES, Wagner, JA and Perakslis, ED (2018) Wearable devices in clinical trials: Hype and hypothesis. Clinical Pharmacology and Therapeutics 104, 4252.CrossRefGoogle ScholarPubMed
Karczewski, KJ and Snyder, MP (2018) Integrative omics for health and disease. Nature Reviews Genetics 19, 299310.CrossRefGoogle ScholarPubMed
Keller, JL, Guyatt, GH, Roberts, RS, Adachi, JD and Rosenbloom, D (1988 ) An N of 1 service: Applying the scientific method in clinical practice. Scandinavian Journal of Gastroenterology Supplement 147, 2229.CrossRefGoogle Scholar
Kim, J, Hu, C, El Achkar, CM, Black, LE, Douville, J, Larson, A, Pendergast, MK, Goldkind, SF, Lee, EA, Kuniholm, A, Soucy, A, Vaze, J, Belur, NR, Fredriksen, K, Stojkovska, I, Tsytsykova, A, Armant, M, DiDonato, RL, Choi, J, Cornelissen, L, Pereira, LM, Augustine, EF, Genetti, CA, Dies, K, Barton, B, Williams, L, Goodlett, BD, Riley, BL, Pasternak, A, Berry, ER, Pflock, KA, Chu, S, Reed, C, Tyndall, K, Agrawal, PB, Beggs, AH, Grant, PE, Urion, DK, Snyder, RO, Waisbren, SE, Poduri, A, Park, PJ, Patterson, A, Biffi, A, Mazzulli, JR, Bodamer, O, Berde, CB and Yu, TW (2019 ) Patient-customized oligonucleotide therapy for a rare genetic disease. New England Journal of Medicine 381, 16441652.CrossRefGoogle ScholarPubMed
Kiyotani, K, Toyoshima, Y and Nakamura, Y (2021) Personalized immunotherapy in cancer precision medicine. Cancer Biology and Medicine 18, 955965.CrossRefGoogle ScholarPubMed
Kravitz, RL and Duan, N (2022) Conduct and implementation of personalized trials in research and practice. Harvard Data Science Review, (SpecialIssue 3). https://doi.org/10.1162/99608f92.901255e7CrossRefGoogle Scholar
Kravitz, RL, Duan, N and DEcIDE Methods Center N-of-1 Guidance Panel (2014) Design and implementation of N-of-1 trials: A user’s guide. In AHRQ Publication No. 13(14)-EHC122-EF. Rockville, MD: Agency for Healthcare Research and Quality.Google Scholar
Krishnamurthy, N, Grimshaw, AA, Axson, SA, Choe, SH and Miller, JE (2022) Drug repurposing: A systematic review on root causes, barriers and facilitators. BMC Health Services Research 22, 970.CrossRefGoogle ScholarPubMed
Kritchevsky, SB and Justice, JN (2020) Testing the geroscience hypothesis: Early days. Journals of Gerontology. Series A: Biological Sciemce and Medical Science 75, 99101.CrossRefGoogle ScholarPubMed
Kronish, IM, Hampsey, M, Falzon, L, Konrad, B and Davidson, KW (2018) Personalized (N-of-1) trials for depression: A systematic review. Journal of Clinical Psychopharmacology 38, 218225.CrossRefGoogle ScholarPubMed
Lamb, JJ, Stone, M, D’Adamo, CR, Volkov, A, Metti, D, Aronica, L, Minich, D, Leary, M, Class, M, Carullo, M, Ryan, JJ, Larson, IA, Lundquist, E, Contractor, N, Eck, B, Ordovas, JM and Bland, JS (2022) Personalized lifestyle intervention and functional evaluation health outcomes SurvEy: Presentation of the LIFEHOUSE study using N-of-one tent-umbrella-bucket design. Journal of Personalized Medicine 12, 115.CrossRefGoogle ScholarPubMed
Leroy, JL, Frongillo, EA, Kase, BE, Alonso, S, Chen, M, Dohoo, I, Huybregts, L, Kadiyala, S and Saville, NM (2022) Strengthening causal inference from randomised controlled trials of complex interventions. British Medical Journal Global Health 7(6):e008597. doi: 10.1136/bmjgh-2022-008597. PMID: 35688484; PMCID: PMC9189821.Google ScholarPubMed
Levy, R, Magis, AT, Earls, JC, Manor, O, Wilmanski, T, Lovejoy, J, Gibbons, SM, Omenn, GS, Hood, L and Price, ND (2020) Longitudinal analysis reveals transition barriers between dominant ecological states in the gut microbiome. Proceedings of the National Academy of Sciences: U S A 117, 1383913845.CrossRefGoogle ScholarPubMed
Li, J, Gao, W, Punja, S, Ma, B, Vohra, S, Duan, N, Gabler, N, Yang, K and Kravitz, RL (2016) Reporting quality of N-of-1 trials published between 1985 and 2013: A systematic review. Journal of Clinical Epidemiology 76, 5764.CrossRefGoogle ScholarPubMed
Lillie, EO, Patay, B, Diamant, J, Issell, B, Topol, EJ and Schork, NJ (2011) The n-of-1 clinical trial: The ultimate strategy for individualizing medicine? Personalized Medicine 8, 161173.CrossRefGoogle ScholarPubMed
Li-Pook-Than, J and Snyder, M (2013 ) iPOP goes the world: Integrated personalized omics profiling and the road toward improved health care. Chemistry and Biology 20, 660666.CrossRefGoogle ScholarPubMed
Liu, K and Meng, XL (2016) There is individualized treatment. Why not individualized inference? Annual Review of Statistics and Its Application 3, 79111.CrossRefGoogle Scholar
Mahmoudi, S, Xu, L and Brunet, A (2019) Turning back time with emerging rejuvenation strategies. Nature Cell Biology 21, 3243.CrossRefGoogle ScholarPubMed
McCracken, JM (2016) Exploratory Causal Analysis with Time Series Data. San Rafael, CA: Morgan and Claypool.CrossRefGoogle Scholar
McDonald, S and Nikles, J (2021) N-of-1 trials in healthcare. Healthcare (Basel) 9, 330.CrossRefGoogle ScholarPubMed
McInnes, G, Yee, SW, Pershad, Y and Altman, RB (2021 ) Genomewide association studies in pharmacogenomics. Clinical Pharmacology and Therapeutics 110, 637648.CrossRefGoogle ScholarPubMed
Metwally, AA, Zhang, T, Wu, S, Kellogg, R, Zhou, W, Contrepois, K, Tang, H and Snyder, M (2022) Robust identification of temporal biomarkers in longitudinal omics studies. Bioinformatics 38, 38023811.CrossRefGoogle ScholarPubMed
Miocevic, M, Moeyaert, M, Mayer, A and Montoya, AK (2022) Causal mediation analysis in single case experimental designs: Introduction to the special issue. Evaluation and the Health Professions 45, 37.CrossRefGoogle ScholarPubMed
Mirza, RD, Punja, S, Vohra, S and Guyatt, G (2017) The history and development of N-of-1 trials. Journal of the Royal Society of Medicine, 110, 330340.CrossRefGoogle ScholarPubMed
Molenaar, PCM (2019 ) Granger causality testing with intensive longitudinal data. Prevention Science 20, 442451.CrossRefGoogle ScholarPubMed
Moskalev, A, Guvatova, Z, Lopes, IA, Beckett, CW, Kennedy, BK, De Magalhaes, JP and Makarov, AA (2022 ) Targeting aging mechanisms: Pharmacological perspectives. Trends in Endocrinology and Metabolism 33, 266280.CrossRefGoogle ScholarPubMed
Mucke, HAM (2022 ) Drug repurposing patent applications March-June 2022. Assay and Drug Development Technologies 20, 286293.CrossRefGoogle ScholarPubMed
Nikles, J, Evans, K, Hams, A and Sterling, M (2022) A systematic review of N-of-1 trials and single case experimental designs in physiotherapy for musculoskeletal conditions. Musculoskeletal Science and Practice 62, 102639.CrossRefGoogle ScholarPubMed
Nikles, J, Onghena, P, Vlaeyen, JWS, Wicksell, RK, Simons, LE, McGree, JM and McDonald, S (2021) Establishment of an international collaborative network for N-of-1 trials and single-case designs. Contemporary Clinical Trials Communications 23, 100826.CrossRefGoogle Scholar
Onder, G, Bernabei, R, Vetrano, DL, Palmer, K and Marengoni, A (2020) Facing multimorbidity in the precision medicine era. Mechanisms of Ageing and Development 190, 111287.CrossRefGoogle ScholarPubMed
Ong, KY, Lee, PSS and Lee, ES (2020 ) Patient-centred and not disease-focused: A review of guidelines and multimorbidity. Singapore Medical Journal 61, 584590.CrossRefGoogle Scholar
Ordutowski, H, Dal Dosso, F, De Wispelaere, W, Van Tricht, C, Vermeire, S, Geukens, N, Gils, A, Spasic, D and Lammertyn, J (2022) Next generation point-of-care test for therapeutic drug monitoring of adalimumab in patients diagnosed with autoimmune diseases. Biosensors & Bioelectronics 208, 114189.CrossRefGoogle ScholarPubMed
Pearson-Stuttard, J, Ezzati, M and Gregg, EW (2019) Multimorbidity-a defining challenge for health systems. The Lancet Public Health 4, e599e600.CrossRefGoogle ScholarPubMed
Phyland, RK, McKay, A, Olver, J, Walterfang, M, Hopwood, M, Ponsford, M and Ponsford, JL (2022) Use of olanzapine to treat agitation in traumatic brain injury: A series of N-of-one trials. Journal of Neurotrauma 40, 3351.CrossRefGoogle ScholarPubMed
Price, ND, Magis, AT, Earls, JC, Glusman, G, Levy, R, Lausted, C, McDonald, DT, Kusebauch, U, Moss, CL, Zhou, Y, Qin, S, Moritz, RL, Brogaard, K, Omenn, GS, Lovejoy, JC and Hood, L (2017) A wellness study of 108 individuals using personal, dense, dynamic data clouds. Nature Biotechnology 35, 747756.CrossRefGoogle ScholarPubMed
Punja, S, Xu, D, Schmid, CH, Hartling, L, Urichuk, L, Nikles, CJ and Vohra, S (2016) N-of-1 trials can be aggregated to generate group mean treatment effects: A systematic review and meta-analysis. Journal of Clinical Epidemiology 76, 6575.CrossRefGoogle ScholarPubMed
Pushpakom, S, Iorio, F, Eyers, PA, Escott, KJ, Hopper, S, Wells, A, Doig, A, Guilliams, T, Latimer, J, McNamee, C, Norris, A, Sanseau, P, Cavalla, D and Pirmohamed, M (2019) Drug repurposing: Progress, challenges and recommendations. Nature Reviews Drug Discovery 18, 4158.CrossRefGoogle ScholarPubMed
Rochon, J (1990) A statistical model for the “N-of-1” study. Journal of Clinical Epidemiology 43, 499508.CrossRefGoogle ScholarPubMed
Roesler, AS and Anderson, KS (2022 ) Beyond sequencing: Prioritizing and delivering neoantigens for cancer vaccines. Methods in Molecular Biology 2410, 649670.CrossRefGoogle ScholarPubMed
Sailani, MR, Metwally, AA, Zhou, W, Rose, SMS, Ahadi, S, Contrepois, K, Mishra, T, Zhang, MJ, Kidzinski, L, Chu, TJ and Snyder, MP (2020) Deep longitudinal multiomics profiling reveals two biological seasonal patterns in California. Nature Communications 11, 4933.CrossRefGoogle ScholarPubMed
Samuel, JP, Wootton, SH, Holder, T and Molony, D (2022) A scoping review of randomized trials assessing the impact of n-of-1 trials on clinical outcomes. PLoS One 17, e0269387.CrossRefGoogle ScholarPubMed
Schork, NJ (2015) Personalized medicine: Time for one-person trials. Nature 520, 609611.CrossRefGoogle ScholarPubMed
Schork, NJ (2018) Randomized clinical trials and personalized medicine: A commentary on deaton and cartwright. Social Science and Medicine 210, 7173.CrossRefGoogle ScholarPubMed
Schork, NJ (2022) Accommodating serial correlation and sequential design elements in personalized studies and aggregated personalized studies. Harvard Data Science Review, (Special Issue 3). https://doi.org/10.1162/99608f92.f1eef6f4.CrossRefGoogle Scholar
Schork, NJ, Beaulieu-Jones, B, Liang, W, Smalley, S and Goetz, LH (2022) Does modulation of an epigenetic clock define a geroprotector? Advances in Geriatric Medicine and Research 4, e220002.Google ScholarPubMed
Schork, NJ and Goetz, LH (2017) Single-subject studies in translational nutrition research. Annual Review of Nutrition 37, 395422.CrossRefGoogle ScholarPubMed
Schork, NJ, Goetz, LH, Lowey, J and Trent, J (2020 ) Strategies for testing intervention matching schemes in cancer. Clinical Pharmacology and Therapeutics 108, 542552.CrossRefGoogle ScholarPubMed
Schussler-Fiorenza Rose, SM, Contrepois, K, Moneghetti, KJ, Zhou, W, Mishra, T, Mataraso, S, Dagan-Rosenfeld, O, Ganz, AB, Dunn, J, Hornburg, D, Rego, S, Perelman, D, Ahadi, S, Sailani, MR, Zhou, Y, Leopold, SR, Chen, J, Ashland, M, Christle, JW, Avina, M, Limcaoco, P, Ruiz, C, Tan, M, Butte, AJ, Weinstock, GM, Slavich, GM, Sodergren, E, McLaughlin, TL, Haddad, F and Snyder, MP (2019) A longitudinal big data approach for precision health. Nature Medicine 25, 792804.CrossRefGoogle ScholarPubMed
Selker, HP, Cohen, T, D’Agostino, RB, Dere, WH, Ghaemi, SN, Honig, PK, Kaitin, KI, Kaplan, HC, Kravitz, RL, Larholt, K, McElwee, NE, Oye, KA, Palm, ME, Perfetto, E, Ramanathan, C, Schmid, CH, Seyfert-Margolis, V, Trusheim, M and Eichler, HG (2022 ) A useful and sustainable role for N-of-1 trials in the healthcare ecosystem. Clinical Pharmacology and Therapeutics 112, 224232.CrossRefGoogle ScholarPubMed
Senn, S (1998 ) Applying results of randomised trials to patients. N of 1 trials are needed. British Medical Journal 317, 537538.CrossRefGoogle ScholarPubMed
Siddiqi, SH, Weigand, A, Pascual-Leone, A and Fox, MD (2021) Identification of personalized transcranial magnetic stimulation targets based on subgenual cingulate connectivity: An independent replication. Biological Psychiatry 90, e55e56.CrossRefGoogle ScholarPubMed
Skou, ST, Mair, FS, Fortin, M, Guthrie, B, Nunes, BP, Miranda, JJ, Boyd, CM, Pati, S, Mtenga, S and Smith, SM (2022) Multimorbidity. Nature Reviews Disease Primers 8, 48.CrossRefGoogle ScholarPubMed
Somer, E, Gische, C and Miocevic, M (2022) Methods for modeling autocorrelation and handling missing data in mediation analysis in single case experimental designs (SCEDs). Evaluation and the Health Professions 45, 3653.CrossRefGoogle ScholarPubMed
Tabachnick, GB and Fidell, LS (2012) Using Multivariate Statistics. New York: Pearson.Google Scholar
Tehrani, F, Teymourian, H, Wuerstle, B, Kavner, J, Patel, R, Furmidge, A, Aghavali, R, Hosseini-Toudeshki, H, Brown, C, Zhang, F, Mahato, K, Li, Z, Barfidokht, A, Yin, L, Warren, P, Huang, N, Patel, Z, Mercier, PP and Wang, J (2022) An integrated wearable microneedle array for the continuous monitoring of multiple biomarkers in interstitial fluid. Nature Biomedical Engineering 6, 12141224.CrossRefGoogle ScholarPubMed
Triolo, F, Harber-Aschan, L, Murri, MB, Calderon-Larranaga, A, Vetrano, DL, Sjoberg, L, Marengoni, A and Dekhtyar, S (2020) The complex interplay between depression and multimorbidity in late life: Risks and pathways. Mechanisms of Ageing and Development 192, 111383.CrossRefGoogle ScholarPubMed
Viana, JN, Edney, S, Gondalia, S, Mauch, C, Sellak, H, O’Callaghan, N and Ryan, JC (2021) Trends and gaps in precision health research: A scoping review. British Medical Journal Open 11, e056938.Google ScholarPubMed
Wang, Y and Schork, NJ (2019) Power and design issues in crossover-based N-of-1 clinical trials with fixed data collection periods. Healthcare (Basel) 7, E84.CrossRefGoogle ScholarPubMed
Wicks, P, Vaughan, TE, Massagli, MP and Heywood, J (2011) Accelerated clinical discovery using self-reported patient data collected online and a patient-matching algorithm. Nature Biotechnology 29, 411414.CrossRefGoogle Scholar
Williams, LM, Coman, JT, Stetz, PC, Walker, NC, Kozel, FA, George, MS, Yoon, J, Hack, LM, Madore, MR, Lim, KO, Philip, NS and Holtzheimer, PE (2021) Identifying response and predictive biomarkers for transcranial magnetic stimulation outcomes: Protocol and rationale for a mechanistic study of functional neuroimaging and behavioral biomarkers in veterans with pharmacoresistant depression. BMC Psychiatry 21, 35.CrossRefGoogle ScholarPubMed
Yeboah, E, Mauer, NS, Hufstedler, H, Carr, S, Matthay, EC, Maxwell, L, Rahman, S, Debray, T, de Jong, VMT, Campbell, H, Gustafson, P, Janisch, T and Barnighausen, T (2021) Current trends in the application of causal inference methods to pooled longitudinal non-randomised data: A protocol for a methodological systematic review. British Medical Journal Open 11, e052969.Google Scholar
Zeggini, E, Gloyn, AL, Barton, AC and Wain, LV (2019 ) Translational genomics and precision medicine: Moving from the lab to the clinic. Science 365, 14091413.CrossRefGoogle ScholarPubMed
Zhao, L, Hu, XJ and Lagakos, SW (2009 ) Statistical monitoring of clinical trials with multivariate response and/or multiple arms: A flexible approach. Biostatistics 10, 310323.CrossRefGoogle ScholarPubMed
Zimmer, A, Korem, Y, Rappaport, N, Wilmanski, T, Baloni, P, Jade, K, Robinson, M, Magis, AT, Lovejoy, J, Gibbons, SM, Hood, L and Price, ND (2021) The geometry of clinical labs and wellness states from deeply phenotyped humans. Nature Communications 12, 3578.CrossRefGoogle ScholarPubMed
Zucker, DR, Ruthazer, R and Schmid, CH (2010 ) Individual (N-of-1) trials can be combined to give population comparative treatment effect estimates: Methodologic considerations. Journal of Clinical Epidemiology 63, 13121323.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. A tree or dendrogram reflecting how similar a number of individuals are with respect to phenotypes of relevance to drug response: the closer the bottommost branches of Figure 1 are – which represent individuals – the more similar the phenotypic profiles of those individuals are. The darkness of the shaded human figures at the bottom of the figure at different positions in the tree reflects the degree to which individuals at those positions in the tree possess a certain characteristic or profile. The circles represent interventions that can benefit different groups of individuals, such that the different locations where the shaded circles are situated represent convergence points for all individuals connected beneath that point who can benefit from the specific intervention. Thus, the topmost circle indicates that all individuals may benefit from that intervention (since all the individual tree branches converge back to that point), whereas the leftmost circle is likely to benefit the first ~25–30% of individuals. The two circles second and third from the left indicate interventions that may benefit a small number of individuals (e.g., only ~10% of individuals). The circle to which the arrow is pointing indicates an intervention that may benefit a large number of individuals but for whom other interventions (reflected by the 5th and 6th circles from the right) may benefit smaller subsets of individuals. Identifying points on trees like this that are consistent with who benefits from an intervention based on understanding of the factors responsible for mediating response is the motivation behind precision medicine and nutrition.

Figure 1

Figure 2. Different, very basic, types of N-of-1 clinical trial designs in which an intervention had a lowering effect on a health measure (like blood pressure). The black and red lines reflect hypothetical health measure trajectories (i.e., longitudinal data) while an individual is not receiving (black) or receiving (red) an intervention. The vertical dashed lines indicate when interventions were provided or changed. Panel A depicts the basic ‘interrupted time’ series design, Panel B the ‘reversal’ design and panel C a reversal design with washout periods (green lines).

Figure 2

Figure 3. Contrasting clinical trial designs. The design depicted on the left is consistent with standard RCTs focusing on a singular health measure or indication (the gray colored dot on the left side of the head of the human figures indicating a single phenotype of interest; for example, depression symptoms). If individuals are found not to respond (NR = Non-Responders) then a future study seeking to identify biomarkers of response could be pursued, whereby a new biomarker phenotype is associated with the response/non-response phenotype (e.g., genomic profile). The design depicted on the right provides the motivation for complementary trials to traditional RCTs whereby the effect of an intervention is evaluated on an individual from a whole-body perspective. The results of this trial are aggregated with trials on other individuals and patterns that could identify responders and non-responders are explored that may also reveal intervention effects on different phenotypes and how those phenotypes interact.

Author comment: EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS — R0/PR1

Comments

To whom it may concern,

Please find a manuscript entitled 'EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS' which we were invited to submit to Cambridge PRISMS by Laetitia Beck. The manuscript has not been submitted to another journal and reviews aspects of N-of-1 trials that make them appealing in an era of precision medicine.

Thanks,

Nicholas J. Schork

Review: EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS — R0/PR2

Comments

Comments to Author: The authors of this manuscript deliver an excellent commentary on what N-of-1 trials have to offer precision medicine. They share a vision for the role of multivariate N-of-1 trials and provide a compelling argument for why such trials can provide a valuable contribution to the evidence base in many clinical areas. The authors make the case for multivariate N-of-1 trials addressing fundamental unanswered questions about human biology and being used as a tool to facilitate drug repurposing, which has received considerable attention in the scientific literature of late (e.g. Pushpakom et al., 2019; Krishnamurthy et al., 2022)

There are two issues I think the authors could consider addressing in a revision of the manuscript:

(1) Some single-case designs mentioned briefly in this paper (AB, ABAB), and visually represented in Figure 1, are not typically considered an “N-of-1 trial” design. I think it is generally accepted that N-of-1 trials are those that involve multiple crossovers that are randomly determined (+/- blinding) and therefore mention of the AB and ABAB design may not be needed. Alternatively, these designs could be described under the broad umbrella term “single-case designs”.

(2) I think it would be appropriate to outline some challenges for multivariate N-of-1 trials; they will require frequent measurement of all outcomes of interest to achieve the statistical power needed for the analysis but some outcomes may not be amendable to such frequent measurement (yet). A couple of sentences to briefly acknowledge these challenges may be useful. In addition, it may not be clear to the reader what the authors mean by doing N-of-1 trials “properly”. Perhaps some extra detail here to clarify would also be useful.

Some other minor points the authors could consider are listed below.

• Although in N-of-1 trials patients may not have to wait for as long as they do in RCTs to receive any results shared with them, it is not clear from the paper how using N-of-1 trials enable them to receive “real time” care (mentioned in the abstract). There may be statistical techniques/packages and technology that can fast-track or automate the analysis of N-of-1 trial data, but it might be good to cover this briefly in the manuscript to endorse the possibility of delivering real-time care.

• The authors state: “Most standard clinical trials have inclusion and exclusion criteria to make sure the trial has been carried out in individuals likely to benefit” (page 9) – other reasons could include safety reasons, to avoid confounds and to ensure the individuals are similar so the results can apply to this group of individuals.

• The authors state: “many interventions are shown not to modulate or affect the phenotype they were designed to impact, calling into question the ‘pre-clinical,’ basic-science driven evidence suggesting that they may have benefit in humans in vivo” (page 9) – this could happen for other reasons also.

• A fifth point to extend the section on page 9-10 could be the lag time to obtain results.

• It may not be clear to the reader what is meant by micro-sampling techniques (page 14). Is this questionnaire sampling methods like ecological momentary assessments or something else? Perhaps the authors could add one or two examples in parenthesis.

• The second paragraph in the conclusion (page 19) might fit better in the main manuscript as an additional future direction/opportunity. ASO is an exciting area that perhaps deserves more space?

• In the last paragraph in the conclusion (page 20) “N-of-1 trials have a 4 fold advantage” - I wondered what you were comparing them to in this statement, as there are possibly many more than just four advantages (depending on what the comparator is).

• If appropriate, the authors may wish to consider mentioning the International Collaborative Network for N-of-1 Trials and Single-Case Designs (www.nof1sced.org) as a resource for those interested in this design (Nikles, J., Onghena, P., Vlaeyen, J. W., Wicksell, R. K., Simons, L. E., McGree, J. M., & McDonald, S. (2021). Establishment of an International Collaborative Network for N-of-1 Trials and Single-Case Designs. Contemporary clinical trials communications, 23, 100826).

Review: EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS — R0/PR3

Conflict of interest statement

Reviewer declares none.

Comments

Comments to Author: In this review, the authors wrote about exploring human biology with N-of-1 clinical trials. This is an interesting topic. The comments from this reviewer are as follows:

1. In Abstract, the authors should clearly state the purpose of the review, so that the readers could understand the content and structure of this review better.

2. The main text of the review included six parts: Introduction, Human biology and legacy clinical trials, Basic N-of-1 trial designs, Multivariate N-of-1 trials, Whole body, biomarker validation, and therapeutic drug monitoring studies, Conclusions and future directions. The authors should clearly state the purpose and the structure of the review in the first part.

3. In the paragraph of ‘Therapeutic drug monitoring studies’, the authors wrote ‘However, by more precisely measuring drug bioavailability and activity in N-of-1 trials, especially in trials for which participants are monitored for multiple health measures, one could explore temporal relationships between drug bioavailability and activity and not just, e.g., pill count-based dosing and outcomes.’ Please give reference(s) for this sentence.

4. For Figure 1 and Figure 2, although Figure legends were given, it is recommended to add symbol notes in the figures as well.

Recommendation: EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS — R0/PR4

Comments

No accompanying comment.

Decision: EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS — R0/PR5

Comments

No accompanying comment.

Author comment: EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS — R1/PR6

Comments

No accompanying comment.

Review: EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS — R1/PR7

Comments

Comments to Author: Excellent article. I look forward to seeing it published.

Review: EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS — R1/PR8

Conflict of interest statement

Reviewer declares none.

Comments

Comments to Author: None

Recommendation: EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS — R1/PR9

Comments

No accompanying comment.

Decision: EXPLORING HUMAN BIOLOGY WITH N-OF-1 CLINICAL TRIALS — R1/PR10

Comments

No accompanying comment.