Hostname: page-component-848d4c4894-p2v8j Total loading time: 0.001 Render date: 2024-05-14T04:56:29.734Z Has data issue: false hasContentIssue false

Mendelian randomization as a tool to inform drug development using human genetics

Published online by Cambridge University Press:  08 February 2023

Iyas Daghlas*
Affiliation:
Department of Neurology, University of California San Francisco, San Francisco, CA, USA
Dipender Gill
Affiliation:
Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK Chief Scientific Advisor Office, Research and Early Development, Novo Nordisk, Copenhagen, Denmark
*
Author for correspondence: Iyas Daghlas, Email: iyas.daghlas@ucsf.edu
Rights & Permissions [Opens in a new window]

Abstract

Drug development is essential to the advancement of human health, however, the process is slow, costly, and at high risk of failure at all stages. A promising strategy for expediting and improving the probability of success in the drug development process is the use of naturally randomized human genetic variation for drug target identification and validation. These data can be harnessed using the Mendelian randomization (MR) analytic paradigm to proxy the lifelong consequences of genetic perturbations of drug targets. In this review, we discuss the myriad applications of the MR paradigm for human drug target identification and validation. We review the methodology and applications of MR, key limitations of MR, and potential future opportunities for research. Throughout the review, we refer to illustrative examples of MR analyses investigating the consequences of genetic inhibition of interleukin 6 signaling which, in some cases, have anticipated results from randomized controlled trials. As human genetic data become more widely available, we predict that MR will serve as a key pillar of support for drug development efforts.

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

Mendelian randomization (MR) is a method that uses naturally randomized human genetic variation to study the lifelong effects of genetic perturbations of drug targets. This approach has great promise to help speed up and improvef the drug development process. In this review, we discuss how MR is used for identifying and testing drug targets, its limitations, and future opportunities for research. As more human genetic data become available, we expect MR to play a major role in drug development.

Introduction

The last century has seen major advances in pharmacotherapy within all medical specialties and with consequent reductions in morbidity and mortality (Fuchs, Reference Fuchs2010; Lichtenberg, Reference Lichtenberg2019). Despite this, there remain many unmet medical needs that necessitate ongoing drug development efforts. The challenges inherent to the process of drug development are highlighted by the poor success rate of drug development programs, which has been estimated to be as low as four percent (Hay et al., Reference Hay, Thomas, Craighead, Economides and Rosenthal2014; Finan et al., Reference Finan, Gaulton, Kruger, Lumbers, Shah, Engmann, Galver, Kelley, Karlsson, Santos, Overington, Hingorani and Casas2017). Challenges contributing to this high failure rate include substantial costs (DiMasi et al., Reference DiMasi, Hansen and Grabowski2003; Schlander et al., Reference Schlander, Hernandez-Villafuerte, Cheng, Mestre-Ferrandiz and Baumann2021), a low probability of passing preclinical testing (van Norman, Reference van Norman2019), poor concordance between efficacy in preclinical studies and clinical trials (Perel et al., Reference Perel, Roberts, Sena, Wheble, Briscoe, Sandercock, Macleod, Mignini, Jayaram and Khan2007), and limitations in accurately predicting drug toxicity in human (Bailey et al., Reference Bailey, Thew and Balls2014; Paglialunga et al., Reference Paglialunga, Morimoto, Clark and Friedrichs2019; van Norman, Reference van Norman2019). A common theme underlying these challenges is the poor translatability of findings from animal models to humans (Akhtar, Reference Akhtar2015).

These limitations of preclinical data in predicting drug efficacy and toxicity have motivated the use of alternative strategies for drug target selection and validation. An increasingly popular strategy is to leverage human genetic variation influencing protein-coding genes, as most small molecules and biologics target proteins (Santos et al., Reference Santos, Ursu, Gaulton, Bento, Donadi, Bologa, Karlsson, Al-Lazikani, Hersey, Oprea and Overington2016). There are several features of human genetic variation that make it an attractive source of data for this endeavor (Figure 1). First, germline genetic variants are randomly allocated at gametogenesis (Davies et al., Reference Davies, Holmes and Davey Smith2018). Thus, if confounding by genetic ancestry is appropriately controlled for (Price et al., Reference Price, Patterson, Plenge, Weinblatt, Shadick and Reich2006), the inheritance of genetic variants is not confounded by environmental variables. This is in contrast to conventional observational analyses that investigate efficacy or repurposing potential for drugs used in clinical practice (also referred to as pharmacoepidemiologic analyses). Second, the assignment of a germline genetic variant is fixed at gametogenesis and is therefore not susceptible to change following development of disease. This phenomenon of reverse causality commonly biases pharmacoepidemiologic analyses. These two inherent properties allow for genetic variants to be leveraged in naturally randomized experiments that can increase confidence in the causal relationship between an exposure and an outcome of interest (Davies et al., Reference Davies, Holmes and Davey Smith2018). Third, human genetic variants can be linked to clinically relevant efficacy and safety outcomes in humans rather than in model organisms. Fourth, large genetic datasets from millions of study participants are readily available for a wide variety of clinically relevant phenotypes including clinical risk factors, disease outcomes, circulating proteins, and diverse imaging phenotypes (Sudlow et al., Reference Sudlow, Gallacher, Allen, Beral, Burton, Danesh, Downey, Elliott, Green, Landray, Liu, Matthews, Ong, Pell, Silman, Young, Sprosen, Peakman and Collins2015; Elliott et al., Reference Elliott, Sharp, Alfaro-Almagro, Shi, Miller, Douaud, Marchini and Smith2018; Hemani et al., Reference Hemani, Zheng, Elsworth, Wade, Haberland, Baird, Laurin, Burgess, Bowden, Langdon, Tan, Yarmolinsky, Shihab, Timpson, Evans, Relton, Martin, Davey Smith, Gaunt and Haycock2018; Kurki et al., Reference Kurki, Karjalainen, Palta, Sipilä, Kristiansson, Donner, Reeve, Laivuori, Aavikko, Kaunisto, Loukola, Lahtela, Mattsson, Laiho, Della Briotta Parolo, Lehisto, Kanai, Mars, Rämö, Kiiskinen, Heyne, Veerapen, Rüeger, Lemmelä, Zhou, Ruotsalainen, Pärn, Hiekkalinna, Koskelainen, Paajanen, Llorens, Gracia-Tabuenca, Siirtola, Reis, Elnahas, Aalto-Setälä, Alasoo, Arvas, Auro, Biswas, Bizaki-Vallaskangas, Carpen, Chen, Dada, Ding, Ehm, Eklund, Färkkilä, Finucane, Ganna, Ghazal, Graham, Green, Hakanen, Hautalahti, Hedman, Hiltunen, Hinttala, Hovatta, Hu, Huertas-Vazquez, Huilaja, Hunkapiller, Jacob, Jensen, Joensuu, John, Julkunen, Jung, Junttila, Kaarniranta, Kähönen, Kajanne, Kallio, Kälviäinen, Kaprio, Kerimov, Kettunen, Kilpeläinen, Kilpi, Klinger, Kosma, Kuopio, Kurra, Laisk, Laukkanen, Lawless, Liu, Longerich, Mägi, Mäkelä, Mäkitie, Malarstig, Mannermaa, Maranville, Matakidou, Meretoja, Mozaffari, Niemi, Niemi, Niiranen, O’Donnell, Obeidat, Okafo, Ollila, Palomäki, Palotie, Partanen, Paul, Pelkonen, Pendergrass, Petrovski, Pitkäranta, Platt, Pulford, Punkka, Pussinen, Raghavan, Rahimov, Rajpal, Renaud, Riley-Gillis, Rodosthenous, Saarentaus, Salminen, Salminen, Salomaa, Schleutker, Serpi, Shen, Siegel, Silander, Siltanen, Soini, Soininen, Sul, Tachmazidou, Tasanen, Tienari, Toppila-Salmi, Tukiainen, Tuomi, Turunen, Ulirsch, Vaura, Virolainen, Waring, Waterworth, Yang, Nelis, Reigo, Metspalu, Milani, Esko, Fox, Havulinna, Perola, Ripatti, Jalanko, Laitinen, Mäkelä, Plenge, McCarthy, Runz, Daly and Palotie2022). In many cases, these data are publicly available, and statistical methods may be used to combine information from different datasets, thus rendering their use efficient and highly cost-effective.

Figure 1. Structure and assumptions of Mendelian randomization analyses (A) and application to the example of genetically proxied inhibition of interleukin 6 (IL-6) signaling (B).

The potential for human genetics to inform drug development is exemplified by the development of PCSK9 inhibitors, a cholesterol-lowering drug approved in record time for the prevention of coronary artery disease (CAD) (Hall, Reference Hall2013). This drug target was first discovered on the basis of damaging variants in the PCSK9 gene that reduced circulating low-density lipoprotein (LDL) cholesterol levels and reduced the risk of CAD (Cohen et al., Reference Cohen, Boerwinkle, Mosley and Hobbs2006). In effect, these variants served as proxies for the potential clinical benefits of LDL-cholesterol lowering via PCSK9 inhibition. The safety profile of PCSK9 inhibition was corroborated by identification of a patient with complete genetic inactivation of PCSK9 due to loss-of-function variants (Hall, Reference Hall2013). Careful phenotyping of this patient identified no major health consequences and served as proof-of-concept for the safety of complete PCSK9 inhibition. PCSK9 inhibitors, such as evolocumab and alirocumab, were subsequently approved (Shapiro et al., Reference Shapiro, Tavori and Fazio2018), and have demonstrated consistent efficacy in clinical trials for the lowering of LDL-cholesterol and prevention of CAD (Karatasakis et al., Reference Karatasakis, Danek, Karacsonyi, Rangan, Roesle, Knickelbine, Miedema, Khalili, Ahmad, Abdullah, Banerjee and Brilakis2017; Shapiro et al., Reference Shapiro, Tavori and Fazio2018).

Although the success of human genetics in identifying PCSK9 as a promising drug target is an outlier, there is broader evidence that drug targets with genetic support are more likely to be approved (Nelson et al., Reference Nelson, Tipney, Painter, Shen, Nicoletti, Shen, Floratos, Sham, Li, Wang, Cardon, Whittaker and Sanseau2015; King et al., Reference King, Wade Davis and Degner2019). This was initially evidenced by a seminal analysis (Nelson et al., Reference Nelson, Tipney, Painter, Shen, Nicoletti, Shen, Floratos, Sham, Li, Wang, Cardon, Whittaker and Sanseau2015) that matched drug-disease pairs with genetic association-phenotype pairs and determined that approved drugs were more likely to have supporting genetic mechanisms. Moreover, drug targets with this form of genetic support had higher odds of progression within each phase of a clinical trial (e.g., phase 1 to 2, phase 2 to 3). It was estimated that selecting a target with genetic support could double the success rate for drugs in clinical development. This finding was replicated in a subsequent study which also found that the strongest form of genetic evidence was in the form of genetic variation that impacts the protein-coding sequence of a gene (King et al., Reference King, Wade Davis and Degner2019). Of note, these retrospective analyses did not select for drug target programs that were initially motivated or supported by genetic data prior to approval of the drug. Thus, although these data are encouraging, it remains unclear the extent to which human genetic support may prospectively increase the odds of a successful drug program. Moreover, these estimates were obtained prior to the widespread use of the methods detailed in this review and may therefore be an underestimate of the true utility of genetics for drug discovery.

Mendelian randomization

Mendelian randomization (MR) is the paradigm by which randomly assorted germline genetic variants can be used as proxies for a disease risk factor or for drug target perturbation (Smith and Ebrahim, Reference Smith and Ebrahim2003, Reference Smith and Ebrahim2004; Davies et al., Reference Davies, Holmes and Davey Smith2018; Burgess et al., Reference Burgess, Davey Smith, Davies, Dudbridge, Gill, Glymour, Hartwig, Holmes, Minelli, Relton and Theodoratou2019). Specifically, MR leverages naturally randomized genetic variants as proxies (also referred to as instrumental variables) for modifying a given exposure to test causal effects on an outcome of interest. The features of human genetic variation outlined above – namely the random assortment at gametogenesis that renders genetic variants less susceptible to confounding and to reverse causality – increase confidence in causal inference from MR analyses (Davies et al., Reference Davies, Holmes and Davey Smith2018). Several key assumptions are necessary for causal inference in this paradigm, including that the genetic variants used as proxies are strongly associated with the exposure of interest (the relevance assumption), that the association of the genetic variants with the exposure and with the outcome are not confounded by environmental variables or by nearby genetic signals (the independence assumption), and that the association of the genetic variants with the outcome is not explained by pathways independent of the exposure of interest (also known as pleiotropy) (Davies et al., Reference Davies, Holmes and Davey Smith2018). These assumptions are summarized in Figure 1. A discussion of the impact of confounding due to nearby genetic signals and the statistical method of colocalization for overcoming this bias is discussed at length by Zuber et al. (Reference Zuber, Grinberg, Gill, Manipur, Slob, Patel, Wallace and Burgess2022). The last assumption, also referred to as the exclusion restriction condition, will be discussed later in this review.

An MR analysis may be conducted when a genetic variant(s) has met the above criteria to be used to proxy a drug target. In the simplest form of an MR analysis, these genetic proxies may be directly tested for their association with a clinical outcome of interest (Gill et al., Reference Gill, Georgakis, Walker, Schmidt, Gkatzionis, Freitag, Finan, Hingorani, Howson, Burgess, Swerdlow, Smith, Holmes, Dichgans, Zheng, Psaty and Davies2021). For PCSK9, this involved testing for an association of the genetic variants with risk of CAD. Statistical methods may be implemented to weigh the effect on the outcome by a unit increase in the biomarker of interest, such as mmol/L of LDL-cholesterol (Gill et al., Reference Gill, Georgakis, Walker, Schmidt, Gkatzionis, Freitag, Finan, Hingorani, Howson, Burgess, Swerdlow, Smith, Holmes, Dichgans, Zheng, Psaty and Davies2021).

The PCSK9 example exemplifies these key principles for the application of MR for drug development. First, the genetic variants proxying PCSK9 inhibition were strongly associated with lower LDL-cholesterol (Cohen et al., Reference Cohen, Boerwinkle, Mosley and Hobbs2006). These variants, therefore, satisfy the MR assumption of relevance. Second, the variants were positioned within the protein-coding sequence of PCSK9 and could thus more confidently, but not definitively, be predicted to exert their effects via influencing PCSK9 function. This contrasts with intronic variants in or around the gene that are more likely to tag causal variants in nearby genes (Acosta et al., Reference Acosta, Szejko and Falcone2021). Third, the genetic variants in PCSK9 were not associated with confounding variables such as age, sex, smoking status, and type 2 diabetes. This supports the MR independence assumption of no confounding and the exclusion restriction assumption that the variants do not exert pleiotropic effects via these pathways.

Scope of review

The recent years have seen an explosion in the methodology and application of drug target-focused MR (Acosta et al., Reference Acosta, Szejko and Falcone2021; Gill et al., Reference Gill, Georgakis, Walker, Schmidt, Gkatzionis, Freitag, Finan, Hingorani, Howson, Burgess, Swerdlow, Smith, Holmes, Dichgans, Zheng, Psaty and Davies2021). In this review, we discuss applications and case studies of these methodologies and approaches. The sections are structured to highlight how MR may be used to provide guidance and supporting data at all stages of drug development (Figure 2). It is our hope that this review will serve as an accessible resource for understanding applications and key limitations of MR for informing drug development.

Figure 2. Applications of Mendelian randomization (MR) to each phase of drug development. PheWAS, phenome-wide association study.

Throughout, we incorporate examples from genetic analyses investigating inhibition of interleukin 6 (IL-6) signaling, a pleiotropic inflammatory cytokine that is the target of Food and Drug Administration-approved drugs including tocilizumab, sarilumab, and satralizumab (Figure 1; Kang et al., Reference Kang, Tanaka, Narazaki and Kishimoto2019). These genetic analyses (summarized in Table 1) serve as examples for how MR may inform all stages of drug development, including anticipation of results from ongoing clinical trials investigating inhibition of IL-6 signaling for the prevention of cardiovascular disease and preservation of kidney function (Ridker and Rane, Reference Ridker and Rane2021). We focus on IL-6 as we anticipate that, in contrast to the established example of PCSK9 inhibitors, these ongoing trials will serve as prospective tests for how MR may inform drug development. Relevant citations were identified in the MEDLINE database using the keyword combinations ‘“IL-6” + “Mendelian randomization”’ and ‘“interleukin 6” + “Mendelian randomization”’. The present study was not designed as a systemic review.

Table 1. Selected examples of MR analyses cited in this Review that investigated effects of genetically proxied inhibition of IL-6 signaling on a diverse range of outcomes

Abbreviation: AA: aortic aneurysm; AAA: abdominal aortic aneurysm; AF: atrial fibrillation: CAD: coronary artery disease; CRP: c-reactive protein; CVD: cardiovascular disease; PAD: peripheral artery disease; PheWAS: phenome-wide association study; T2D: type 2 diabetes.

Planning the analysis: Drug target discovery or selection

A drug target may first be identified from a genome (GWAS) or exome-wide association study conducted using a clinical phenotype of interest. For example, a 2013 GWAS of CAD identified a lead variant in the IL6R gene (Deloukas et al., Reference Deloukas, Kanoni, Willenborg, Farrall, Assimes, Thompson, Ingelsson, Saleheen, Erdmann, Goldstein, Stirrups, König, Cazier, Johansson, Hall, Lee, Willer, Chambers, Esko, Folkersen, Goel, Grundberg, Havulinna, Ho, Hopewell, Eriksson, Kleber, Kristiansson, Lundmark, Lyytikäinen, Rafelt, Shungin, Strawbridge, Thorleifsson, Tikkanen, Van Zuydam, Voight, Waite, Zhang, Ziegler, Absher, Altshuler, Balmforth, Barroso, Braund, Burgdorf, Claudi-Boehm, Cox, Dimitriou, Do, Doney, El Mokhtari, Eriksson, Fischer, Fontanillas, Franco-Cereceda, Gigante, Groop, Gustafsson, Hager, Hallmans, Han, Hunt, Kang, Illig, Kessler, Knowles, Kolovou, Kuusisto, Langenberg, Langford, Leander, Lokki, Lundmark, McCarthy, Meisinger, Melander, Mihailov, Maouche, Morris, Müller-Nurasyid, Nikus, Peden, Rayner, Rasheed, Rosinger, Rubin, Rumpf, Schäfer, Sivananthan, Song, Stewart, Tan, Thorgeirsson, van der Schoot, Wagner, Wells, Wild, Yang, Amouyel, Arveiler, Basart, Boehnke, Boerwinkle, Brambilla, Cambien, Cupples, de Faire, Dehghan, Diemert, Epstein, Evans, Ferrario, Ferrières, Gauguier, Go, Goodall, Gudnason, Hazen, Holm, Iribarren, Jang, Kähönen, Kee, Kim, Klopp, Koenig, Kratzer, Kuulasmaa, Laakso, Laaksonen, Lee, Lind, Ouwehand, Parish, Park, Pedersen, Peters, Quertermous, Rader, Salomaa, Schadt, Shah, Sinisalo, Stark, Stefansson, Trégouët, Virtamo, Wallentin, Wareham, Zimmermann, Nieminen, Hengstenberg, Sandhu, Pastinen, Syvänen, Hovingh, Dedoussis, Franks, Lehtimäki, Metspalu, Zalloua, Siegbahn, Schreiber, Ripatti, Blankenberg, Perola, Clarke, Boehm, O’Donnell, Reilly, März, Collins, Kathiresan, Hamsten, Kooner, Thorsteinsdottir, Danesh, Palmer, Roberts, Watkins, Schunkert and Samani2013). This finding, in the context of independent human genetic evidence for a role of IL-6 signaling in the development of cardiovascular disease (Sarwar et al., Reference Sarwar, Adam and Butterworth2012), motivated further investigation of IL-6 signaling as a therapeutic target for CAD (Ridker and Rane, Reference Ridker and Rane2021). Several heuristics may aid in linking an identified genetic variant to the gene it causally influences, including the proximity of the variant to the gene and the functional consequence of the variant (Forgetta et al., Reference Forgetta, Jiang, Vulpescu, Hogan, Chen, Morris, Grinek, Benner, Jang, Hoang, Burtt, Flannick, McCarthy, Fauman, Greenwood, Maurano and Richards2022). With the increasing availability of exome data, a ‘gene-first’ approach utilizes the gene as the unit of analysis (Backman et al., Reference Backman, Li, Marcketta, Sun, Mbatchou, Kessler, Benner, Liu, Locke, Balasubramanian, Yadav, Banerjee, Gillies, Damask, Liu, Bai, Hawes, Maxwell, Gurski, Watanabe, Kosmicki, Rajagopal, Mighty, Jones, Mitnaul, Stahl, Coppola, Jorgenson, Habegger, Salerno, Shuldiner, Lotta, Overton, Cantor, Reid, Yancopoulos, Kang, Marchini, Baras, Abecasis and Ferreira2021). In these analyses, loss-of-function or deleterious missense variants are aggregated and tested for their associations with the outcome of interest. A causal link with the gene can more confidently be made in this setting. Multi-omics in silico data may also be integrated to help identify causal genes. One such approach prioritizes causal genes using machine learning methods to integrate multimodality functional data from proteomics, transcriptomics, and epigenomics across multiple tissue and cell types (https://genetics.opentargets.org/; Mountjoy et al., Reference Mountjoy, Schmidt, Carmona, Schwartzentruber, Peat, Miranda, Fumis, Hayhurst, Buniello, Karim, Wright, Hercules, Papa, Fauman, Barrett, Todd, Ochoa, Dunham and Ghoussaini2021). Additional methods for linking genetic variant to causal gene, and the respective strengths and weaknesses of these methods, are discussed at length elsewhere (Gallagher and Chen-Plotkin, Reference Gallagher and Chen-Plotkin2018).

The drug target may also be identified from preclinical hypothesis-driven studies, or from high-throughput in vitro or in vivo screens. Results from hypothesis-free scans utilizing libraries of genetic proxies for protein levels can also be used to prioritize a drug target (Folkersen et al., Reference Folkersen, Gustafsson, Wang, Hansen, Hedman, Schork, Page, Zhernakova, Wu, Peters, Eriksson, Bergen, Boutin, Bretherick, Enroth, Kalnapenkis, Gådin, Suur, Chen, Matic, Gale, Lee, Zhang, Quazi, Ala-Korpela, Choi, Claringbould, Danesh, Davey Smith, de Masi, Elmståhl, Engström, Fauman, Fernandez, Franke, Franks, Giedraitis, Haley, Hamsten, Ingason, Johansson, Joshi, Lind, Lindgren, Lubitz, Palmer, Macdonald-Dunlop, Magnusson, Melander, Michaelsson, Morris, Mägi, Nagle, Nilsson, Nilsson, Orho-Melander, Polasek, Prins, Pålsson, Qi, Sjögren, Sundström, Surendran, Võsa, Werge, Wernersson, Westra, Yang, Zhernakova, Ärnlöv, Fu, Smith, Esko, Hayward, Gyllensten, Landen, Siegbahn, Wilson, Wallentin, Butterworth, Holmes, Ingelsson and Mälarstig2020; Henry et al., Reference Henry, Gordillo-Marañón, Finan, Schmidt, Ferreira, Karra, Sundström, Lind, Arnlöv, Zannad, Mälarstig, Hingorani and Lumbers2022). Finally, an established drug target may be investigated for drug repurposing opportunities.

Approaches to the identification of a genetic proxy for use in drug target MR analyses

The first step in designing a drug target MR analysis is to select a strategy for the identification of a genetic proxy for a drug target. This involves several decisions regarding location of the genetic variants in relation to the protein-coding gene sequence, the functional consequence of the variant, and the choice of phenotype used to weigh the effects of the genetic proxy. Genetic variants are cis-acting when they are located within or close to the gene of interest. This distance from variant to gene is not standardized, with some studies using variants within 100 kb (Daghlas et al., Reference Daghlas, Karhunen, Ray, Zuber, Burgess, Tsao, Lynch, Lee, Voight, Chang, Baker, Damrauer, Howson, Vujkovic and Gill2021) upstream or downstream of the gene, and others using up to 1 Mb ranges (Pietzner et al., Reference Pietzner, Wheeler, Carrasco-Zanini, Cortes, Koprulu, Wörheide, Oerton, Cook, Stewart, Kerrison, Luan, Raffler, Arnold, Arlt, O’Rahilly, Kastenmüller, Gamazon, Hingorani, Scott, Wareham and Langenberg2021; Yang et al., Reference Yang, Farias, Ibanez, Suhy, Sadler, Fernandez, Wang, Bradley, Eiffert, Bahena, Budde, Li, Dube, Sung, Mihindukulasuriya, Morris, Fagan, Perrin, Benitez, Rhinn, Harari and Cruchaga2021). Efforts have been made to empirically define the optimal variant to gene distance for determination of cis versus trans function, but this is still a work in progress (Fauman and Hyde, Reference Fauman and Hyde2022). Trans-acting variants are positioned outside this genomic range, are not as confidently linked to the gene of interest and are thus considered to be less robust proxies for use in drug target MR analyses (Swerdlow et al., Reference Swerdlow, Kuchenbaecker, Shah, Sofat, Holmes, White, Mindell, Kivimaki, Brunner, Whittaker, Casas and Hingorani2016; Gill et al., Reference Gill, Georgakis, Walker, Schmidt, Gkatzionis, Freitag, Finan, Hingorani, Howson, Burgess, Swerdlow, Smith, Holmes, Dichgans, Zheng, Psaty and Davies2021). This is in part due to the potential for the variant to influence genes and biological pathways independent of the drug target of interest and hence violate the exclusion restriction condition (see Said et al. (Reference Said, Pazoki, Karhunen, Võsa, Ligthart, Bodinier, Koskeridis, Welsh, Alizadeh, Chasman, Sattar, Chadeau-Hyam, Evangelou, Jarvelin, Elliott, Tzoulaki and Dehghan2022) for an example of this phenomenon with MR analyses of C-reactive protein [CRP]). As previously outlined, plausibility that the genetic proxy for the drug target relates to function of the gene of interest may be enhanced by selecting damaging or predicted loss-of-function variants in the protein-coding sequence of the gene (Deboever et al., Reference Deboever, Tanigawa, Lindholm, McInnes, Lavertu, Ingelsson, Chang, Ashley, Bustamante, Daly and Rivas2018; Emdin et al., Reference Emdin, Khera, Chaffin, Klarin, Natarajan, Aragam, Haas, Bick, Zekavat, Nomura, Ardissino, Wilson, Schunkert, McPherson, Watkins, Elosua, Bown, Samani, Baber, Erdmann, Gupta, Danesh, Chasman, Ridker, Denny, Bastarache, Lichtman, D’Onofrio, Mattera, Spertus, Sheu, Taylor, Psaty, Rich, Post, Rotter, Chen, Krumholz, Saleheen, Gabriel and Kathiresan2018; Daghlas et al., Reference Daghlas and Gill2021). However, empiric studies have shown that reliable inference may still be obtained from MR analyses that only leverage intronic variants (Schmidt et al., Reference Schmidt, Finan, Gordillo-Marañón, Asselbergs, Freitag, Patel, Tyl, Chopade, Faraway, Zwierzyna and Hingorani2020).

The second consideration is what phenotype to use to weigh the effect of the genetic variant (Figure 3). One possibility is to use a phenotype of molecular function, such as gene expression or protein abundance (where the variants are known as protein quantitative trait loci, or pQTLs; Porcu et al., Reference Porcu, Rüeger, Lepik, Santoni, Reymond and Kutalik2019; Zheng et al., Reference Zheng, Haberland, Baird, Walker, Haycock, Hurle, Gutteridge, Erola, Liu, Luo, Robinson, Richardson, Staley, Elsworth, Burgess, Sun, Danesh, Runz, Maranville, Martin, Yarmolinsky, Laurin, Holmes, Liu, Estrada, Santos, McCarthy, Waterworth, Nelson, Smith, Butterworth, Hemani, Scott and Gaunt2020). Of these, pQTLs measured in the relevant tissue may be preferable given the proximity of protein levels to clinical phenotypes (Schmidt et al., Reference Schmidt, Finan, Gordillo-Marañón, Asselbergs, Freitag, Patel, Tyl, Chopade, Faraway, Zwierzyna and Hingorani2020; Gill et al., Reference Gill, Georgakis, Walker, Schmidt, Gkatzionis, Freitag, Finan, Hingorani, Howson, Burgess, Swerdlow, Smith, Holmes, Dichgans, Zheng, Psaty and Davies2021), and the fact that drug targets are typically proteins. Several large genome-wide association studies (GWAS) have been published that catalog pQTL associations in different tissues and have made their results publicly available (Yao et al., Reference Yao, Chen, Song, Keefe, Mendelson, Huan, Sun, Laser, Maranville, Wu, Ho, Courchesne, Lyass, Larson, Gieger, Graumann, Johnson, Danesh, Runz, Hwang, Liu, Butterworth, Suhre and Levy2018; Zheng et al., Reference Zheng, Haberland, Baird, Walker, Haycock, Hurle, Gutteridge, Erola, Liu, Luo, Robinson, Richardson, Staley, Elsworth, Burgess, Sun, Danesh, Runz, Maranville, Martin, Yarmolinsky, Laurin, Holmes, Liu, Estrada, Santos, McCarthy, Waterworth, Nelson, Smith, Butterworth, Hemani, Scott and Gaunt2020; Pietzner et al., Reference Pietzner, Wheeler, Carrasco-Zanini, Cortes, Koprulu, Wörheide, Oerton, Cook, Stewart, Kerrison, Luan, Raffler, Arnold, Arlt, O’Rahilly, Kastenmüller, Gamazon, Hingorani, Scott, Wareham and Langenberg2021; Suhre et al., Reference Suhre, McCarthy and Schwenk2021; Yang et al., Reference Yang, Farias, Ibanez, Suhy, Sadler, Fernandez, Wang, Bradley, Eiffert, Bahena, Budde, Li, Dube, Sung, Mihindukulasuriya, Morris, Fagan, Perrin, Benitez, Rhinn, Harari and Cruchaga2021). A key limitation in the analytic application of pQTLs is confounding due to variant effects that interfere with the aptamer assay used to measure protein levels (Suhre et al., Reference Suhre, McCarthy and Schwenk2021). These biases may be examined by using, when available, multiple independent pQTL datasets that utilize different assays for measurement of protein levels (Zheng et al., Reference Zheng, Haberland, Baird, Walker, Haycock, Hurle, Gutteridge, Erola, Liu, Luo, Robinson, Richardson, Staley, Elsworth, Burgess, Sun, Danesh, Runz, Maranville, Martin, Yarmolinsky, Laurin, Holmes, Liu, Estrada, Santos, McCarthy, Waterworth, Nelson, Smith, Butterworth, Hemani, Scott and Gaunt2020). Finally, molecular phenotypes may include measures of target engagement, such as the use of an acute phase reactant like CRP when examining effects of perturbing IL-6 signaling (Georgakis et al., Reference Georgakis, Malik, Li, Gill, Levin, Vy, Judy, Ritchie, Verma, Nadkarni, Damrauer, Theodoratou and Dichgans2021).

Figure 3. Different approaches to weighting the effect of a genetic variant on a drug target. The absence of an arrow between the ‘molecular biomarkers’ and ‘clinical risk factors and outcomes’ box demonstrates that a molecular biomarker does not necessarily have to be causal for a disease outcome.

An alternative or complementary strategy for weighting genetic variants for use in MR is to use disease risk factors or outcomes. An advantage of these approaches relative to molecular phenotypes is confirmation that the variant influences the clinical phenotype of interest. Examples of disease risk factors with relevance to drug targets include glycated hemoglobin for proxying effects of antidiabetic drugs (e.g., glucagon-like peptide 1 receptor agonists; Daghlas et al., Reference Daghlas, Karhunen, Ray, Zuber, Burgess, Tsao, Lynch, Lee, Voight, Chang, Baker, Damrauer, Howson, Vujkovic and Gill2021) and blood pressure for proxying effects of anti-hypertensive drugs (e.g., beta-blockers) (Gill et al., Reference Gill, Georgakis, Koskeridis, Jiang, Feng, Wei, Theodoratou, Elliott, Denny, Malik, Evangelou, Dehghan, Dichgans and Tzoulaki2019). Alternatively, genetic variants may be weighted by their association with a binary disease outcome such as hypertension (Burgess and Labrecque, Reference Burgess and Labrecque2018). The genetic variants must be strongly and reliably associated with this phenotype of interest to meet the first MR assumption of relevance (Davies et al., Reference Davies, Holmes and Davey Smith2018). In MR analyses, this statistical strength can be measured using the F-statistic (Davies et al., Reference Davies, Holmes and Davey Smith2018). If multiple variants are used, they should either be independently inherited, or specialized methods should be employed to account for their correlation (Yavorska and Burgess, Reference Yavorska and Burgess2017). Such methods can, for example, use correlation matrices to account for linkage disequilibrium between genetic variants, thus ensuring that each genetic variant provides ‘unique’ information for the analysis.

Several analytic approaches have been used to proxy inhibition of IL-6 signaling. One approach is to use the rs2228145 variant, which is a missense variant that reduces membrane-bound IL-6 receptor levels, increases soluble IL-6 receptor levels, which serves as a decoy receptor that negatively regulates IL-6 signaling (Sarwar et al., Reference Sarwar, Adam and Butterworth2012; Ferreira et al., Reference Ferreira, Freitag, Cutler, Howson, Rainbow, Smyth, Kaptoge, Clarke, Boreham, Coulson, Pekalski, Chen, Onengut-Gumuscu, Rich, Butterworth, Malarstig, Danesh and Todd2013). A second approach is to use independent cis variants associated with CRP, an established inflammatory biomarker of IL-6 signaling (Georgakis et al., Reference Georgakis, Malik, Gill, Franceschini, Sudlow and Dichgans2020). A third approach is to use pQTLs for soluble IL-6 receptor levels (Rosa et al., Reference Rosa, Chignon, Li, Boulanger, Arsenault, Bossé, Thériault and Mathieu2019). These approaches are not mutually exclusive and may be combined in complementary sensitivity analyses.

Phase 1 and 2: Using MR to identify measures of target engagement

Once a genetic proxy for perturbation of a drug target has been identified, the association of the proxy with circulating molecules may be characterized to understand the biology of perturbation of the drug target and to identify target engagement biomarkers (Stefaniak and Huber, Reference Stefaniak and Huber2020). Such biomarkers may be considered for further investigation as measurable outcomes in clinical studies. This genetic approach has been applied to characterize the molecular signature of inhibition of IL-6 signaling. For instance, the IL-6 receptor antagonist tocilizumab is known to raise circulating IL-6 and soluble IL-6 receptor levels while reducing serum fibrinogen levels (Georgakis et al., Reference Georgakis, Malik, Gill, Franceschini, Sudlow and Dichgans2020). An MR analysis using a cis genetic proxy for inhibition of IL-6 signaling recapitulated these same effects, validating use of this genetic proxy for testing the effects of perturbation of IL-6 signaling (Georgakis et al., Reference Georgakis, Malik, Gill, Franceschini, Sudlow and Dichgans2020). In a more novel application of MR for this aim, a genetic proxy for IL-6 signaling was tested for its effects on 40 circulating cytokines to characterize the inflammatory signature of IL-6 signaling (Rahman et al., Reference Rahman, McEwan, Ryan and Gill2022). Significant associations of genetically proxied inhibition of IL-6 signaling were identified for ten inflammatory molecules, providing novel insights into the molecular signature of pharmacologic inhibition of IL-6 signaling.

Phase 1 and2: Use of MR to assess for adverse effects of drug target perturbation

Genetic association data may be used in MR analyses for the investigation of on-target safety outcomes. On-target toxicity represents effects secondary to modulation of the drug target and can be investigated using MR. In contrast, off-target toxicity represents effects attributable to pleiotropic biochemical effects of the drug compound itself on proteins and pathways independent of the drug target of interest and cannot be investigated using MR (Rudmann, Reference Rudmann2013).

One approach, known as a ‘phenome-wide association study’ (PheWAS), uses MR to perform hypothesis-free scans for adverse effects of drug targets across the human phenome (Denny et al., Reference Denny, Ritchie, Basford, Pulley, Bastarache, Brown-Gentry, Wang, Masys, Roden and Crawford2010). Outcomes in PheWAS are often defined using International Classification of Disease (ICD) codes and therefore sample sizes may be smaller than those available from large-scale GWAS consortia. In the case of genetically proxied inhibition of IL-6 signaling, a PheWAS identified safety signals for increased risk of atopic dermatitis, cellulitis, urinary tract infections, and cholecystitis. In such cases, it may be unclear whether adverse effects of a drug target outweigh its benefit. One approach to address this question is to investigate the outcome of lifespan (Daghlas and Gill, Reference Daghlas and Gill2021). In the considered population, a net benefit in lifespan due to perturbation of a drug target suggests that adverse effects do not outweigh the clinical efficacy outcome and that the drug target may yield a mortality benefit in clinical trials (Daghlas and Gill, Reference Daghlas and Gill2021). Using the outcome of parental lifespan, an MR analysis that demonstrated a possible lifespan benefit for genetically proxied inhibition of IL-6 signaling in a general population (Rosa et al., Reference Rosa, Chignon, Li, Boulanger, Arsenault, Bossé, Thériault and Mathieu2019). This finding supports the notion that adverse effects of inhibition of IL-6 signaling (e.g., infection) are outweighed by the benefits of inhibiting this pathway (e.g., reduced risk of cardiovascular disease). It is important to appreciate that these analyses are typically performed using genetic associations with lifespan in the general population (Timmers et al., Reference Timmers, Mounier, Lall, Fischer, Ning, Feng, Bretherick, Clark, Shen, Esko, Kutalik, Wilson and Joshi2019) and therefore may not accurately represent effects anticipated in population subgroups targeted in clinical trials.

Pre-phase 3: Informing clinical trial design

Constructing a primary outcome

Investigators designing a clinical trial must select a primary efficacy outcome which is often a composite of related clinical outcomes (Andrade, Reference Andrade2015). MR may be leveraged to test the drug target across multiple related outcomes, and this information may be used to prioritize outcomes to include in a primary efficacy outcome in a clinical trial (Gill and Burgess, Reference Gill and Burgess2020). This is helpful because the inclusion of an outcome unaffected by the drug reduces statistical power and increases the odds of failure of a drug candidate in a phase 3 trial. For instance, the concordant effects of genetically proxied inhibition of IL-6 signaling on risk of stroke and CAD suggest that combining these clinical outcomes in a clinical trial could increase statistical power of a phase 3 clinical trial (Georgakis et al., Reference Georgakis, Malik, Gill, Franceschini, Sudlow and Dichgans2020). In contrast, the null associations with venous thromboembolism suggests that inclusion of this phenotype in an efficacy outcome would reduce the statistical power of a clinical trial (Georgakis et al., Reference Georgakis, Malik, Gill, Franceschini, Sudlow and Dichgans2020).

Investigating drug effects in population subgroups

Investigators and clinicians are typically interested in the effect of a drug in particular population subgroups. For instance, a clinical trial of an IL-6 inhibitor may enroll a patient population with evidence of elevated levels of IL-6 driven inflammation. Similarly, clinical trials typically stratify their enrolled population across covariates of interest to test for consistency of treatment effect in patient subgroups. MR analyses may be designed to mimic these conditions and to test for heterogeneous effects across subgroups. This application is demonstrated by an MR analysis of genetically proxied inhibition of IL-6 signaling that stratified across numerous covariates including sex, age, kidney function, body mass index, LDL-cholesterol, blood pressure, and hemoglobin A1C (Georgakis et al., Reference Georgakis, Malik, Richardson, Howson, Anderson, Burgess, Hovingh, Dichgans and Gill2022). The relative benefit of inhibition of IL-6 signaling was consistent across strata of all covariates except for LDL-cholesterol, where a greater magnitude of benefit was seen at levels of LDL-cholesterol greater than 160 mg/dL (Georgakis et al., Reference Georgakis, Malik, Richardson, Howson, Anderson, Burgess, Hovingh, Dichgans and Gill2022). A larger absolute benefit was observed in the population subgroup with highest baseline high-sensitivity CRP levels, suggesting that clinical trials should prioritize this population for IL-6 inhibition therapies. MR may also be used to identify population subgroups that are less likely to benefit from perturbation of a given drug target. This is illustrated by the null effect of genetically proxied inhibition of IL-6 signaling on risk of the cardioembolic stroke subtype relative to the small and large vessel ischemic stroke subtypes (Georgakis et al., Reference Georgakis, Malik, Gill, Franceschini, Sudlow and Dichgans2020).

Testing for drug–drug interactions

New drugs are often used in concert with other drugs, such as the combination of antiplatelet therapies with statins for the secondary prevention of ischemic stroke. Indeed, polypharmacy is becoming more common despite the absence of evidence for the safety of drug combinations (Oktora et al., Reference Oktora, Denig, Bos, Schuiling-Veninga and Hak2019). The potential consequences of such drug–drug interactions may be queried in appropriately designed MR analyses. One approach is to perform a 2 × 2 factorial analysis stratified across levels of genetic proxies for two drug targets (see the citation for alternative modeling approaches; Rees et al., Reference Rees, Foley and Burgess2020). This approach can determine whether genetically proxied drug effects on a clinical outcome are additive, or if they have supra-additive effects. An additive effect was identified in a 2 × 2 factorial analysis investigating interactions between genetically proxied inhibition of IL-6 signaling and genetically proxied HMGCR (the target of statins) inhibition (Georgakis et al., Reference Georgakis, Malik, Burgess and Dichgans2022).

Phase 3: Use of genetic proxies in MR to test clinical efficacy of a drug target

Once a genetic proxy for a drug target of interest has been selected, statistical associations of the genetic variants with the outcome of interest may be extracted from a GWAS. The effect of drug target perturbation, weighted by changes in the levels of a relevant biomarker, may then be estimated using conventional statistical methods (Burgess et al., Reference Burgess, Small and Thompson2017; Hemani et al., Reference Hemani, Zheng, Elsworth, Wade, Haberland, Baird, Laurin, Burgess, Bowden, Langdon, Tan, Yarmolinsky, Shihab, Timpson, Evans, Relton, Martin, Davey Smith, Gaunt and Haycock2018; Teumer, Reference Teumer2018). This approach was used in an MR analysis to support a protective effect of inhibition of IL-6 signaling on risk of the efficacy outcome of CAD (Sarwar et al., Reference Sarwar, Adam and Butterworth2012). Subsequent studies identified similar effects for additional cardiovascular outcomes including abdominal aortic aneurysm, peripheral artery disease (Levin et al., Reference Levin, Klarin, Georgakis, Lynch, Liao, Voight, O’Donnell, Chang, Assimes, Tsao and Damrauer2021), atrial fibrillation (Rosa et al., Reference Rosa, Chignon, Li, Boulanger, Arsenault, Bossé, Thériault and Mathieu2019), and ischemic stroke (Georgakis et al., Reference Georgakis, Malik, Gill, Franceschini, Sudlow and Dichgans2020). In contrast to the above studies, MR may be used to identify drug targets that are less likely to causally affect an outcome of interest. For example, MR analyses did not support causal effects of IL-6 signaling on pulmonary arterial hypertension, which was concordant with a null effect from a phase 2 clinical study (Toshner et al., Reference Toshner, Church, Harbaum, Rhodes, Villar Moreschi, Liley, Jones, Arora, Batai, Desai, Coghlan, Gibbs, Gor, Gräf, Harlow, Hernandez-Sanchez, Howard, Humbert, Karnes, Kiely, Kittles, Knightbridge, Lam, Lutz, Nichols, Pauciulo, Pepke-Zaba, Suntharalingam, Soubrier, Trembath, Schwantes-An, Wort, Wilkins, Gaine, Morrell and Corris2022). This result supports the redirection of resources away from further testing of IL-6 inhibition for this indication.

Post-phase 3: Using MR to identify repurposing opportunities for an established drug

There is great interest in drug repurposing (also referred to as drug repositioning), whereby a drug that has undergone safety and efficacy testing for one indication is proven to be effective for a separate indication (Glenn Begley et al., Reference Begley, Ashton, Baell, Bettess, Brown, Carter, Charman, Davis, Fisher, Frazer, Gautam, Jennings, Kearney, Keeffe, Kelly, Lopez, McGuckin, Parker, Rayner, Roberts, Rush and Sullivan2021). Such an approach circumvents several challenges and expenses in drug development outlined in the Introduction (Pushpakom et al., Reference Pushpakom, Iorio, Eyers, Escott, Hopper, Wells, Doig, Guilliams, Latimer, McNamee, Norris, Sanseau, Cavalla and Pirmohamed2018). MR may be used to provide evidence for these repurposing efforts (Gill and Vujkovic, Reference Gill and Vujkovic2022). This approach requires identification of a genetic proxy that is validated to influence the clinical outcomes for which a drug target has been approved (Gill and Burgess, Reference Gill and Burgess2020). A clinical outcome may then be selected for investigation, or a hypothesis-free PheWAS may be performed to identify novel repurposing opportunities.

As an example of a hypothesis-driven approach, genetically proxied inhibition of IL-6 signaling was investigated for its effects on the outcome of polymyalgia rheumatica (PMR), an inflammatory musculoskeletal disorder (Zhao and Gill, Reference Zhao and Gill2022). A protective effect of inhibition of IL-6 signaling was identified for this disease outcome, a finding which has been corroborated by clinical trial evidence for a beneficial effect of tocilizumab on disease activity in PMR (Devauchelle-Pensec et al., Reference Devauchelle-Pensec, Carvajal-Alegria, Dernis, Richez, Truchetet, Wendling, Toussirot, Perdriger, Gottenberg, Felten, Fautrel, Chiche, Hilliquin, Le Henaff, Dervieux, Direz, Chary-Valckenaere, Cornec, Guellec, Marhadour, Nowak and Saraux2022). This approach was also used to identify a protective effect of genetically proxied IL-6 signaling on risk of COVID-19 incidence and severity, a finding consistent with results from clinical trials of IL-6 receptor inhibitors (Bovijn et al., Reference Bovijn, Lindgren and Holmes2020; Larsson et al., Reference Larsson, Burgess and Gill2021; Rajasundaram et al., Reference Rajasundaram, Burgess and Gill2022). As an example of a hypothesis-free approach, an MR-PheWAS analysis using genetic data from the Million Veterans Program identified a potential repurposing opportunity for IL-6 receptor antagonists for prevention of aortic aneurysm (Cai et al., Reference Cai, Zhang, Ho, Link, Sun, Huang, Cai, Damrauer, Ahuja, Honerlaw, Huang, Costa, Schubert, Hong, Gagnon, Sun, Gaziano, Wilson, Cho, Tsao, O’Donnell and Liao2018), a finding replicated in the UK Biobank dataset (Georgakis et al., Reference Georgakis, Malik, Li, Gill, Levin, Vy, Judy, Ritchie, Verma, Nadkarni, Damrauer, Theodoratou and Dichgans2021).

Limitations and context

Despite the potential for MR to inform drug development efforts, the methodology has several limitations that must be considered when contextualizing any findings. The first set of limitations relate to interpretation of the effect size from an MR analysis. First, these numeric estimates reflect the consequence of lifelong perturbation of a drug target. In effect, the ‘time zero’ for the natural experiment is set either at gametogenesis or when the variant becomes biologically relevant, and so the magnitude of effect over this duration may not be predictive of the magnitude of benefit from a clinical intervention of shorter duration. This is illustrated by the larger magnitude of effect estimates for LDL-cholesterol lowering from MR analyses relative to those from clinical trials (Ference et al., Reference Ference, Ginsberg, Graham, Ray, Packard, Bruckert, Hegele, Krauss, Raal, Schunkert, Watts, Borén, Fazio, Horton, Masana, Nicholls, Nordestgaard, van de Sluis, Taskinen, Tokgözoglu, Landmesser, Laufs, Wiklund, Stock, Chapman and Catapano2017). Second, MR estimates may be biased by canalization, whereby lifelong genetic effects on a phenotype are buffered by compensatory developmental processes and hence may differ from shorter-term targeting of a protein later in life (Lawlor et al., Reference Lawlor, Harbord, Sterne, Timpson and Smith2008). Third, effect estimates in conventional MR analyses correspond to differences in exposure levels around the population mean and cannot inform the consequences of large changes in levels or function of the drug target. Exceptions to this principle include variants with large magnitudes of effect, such as protein-truncating variants, or the use of analyses that employ nonlinear statistical methodologies (Burgess et al., Reference Burgess, Davies and Thompson2014).

A second set of limitations correspond to the assumption of no bias due to pleiotropy, or the exclusion restriction condition. For instance, a genetic variant may be a cis pQTL for a drug target of interest and a trans pQTL for another protein. The association of this variant with a given outcome may be mediated through pathways independent of the protein of interest. Given the widespread pleiotropy in the human genome (Verbanck et al., Reference Verbanck, Chen, Neale and Do2018), the exclusion restriction assumption may be violated in many instances, although careful analysis may mitigate this bias. For instance, numerous statistical methods have been developed that recover consistent causal effect estimates despite varying degrees of pleiotropic effects of the genetic variants (Bowden et al., Reference Bowden, Del Greco, Minelli, Davey Smith, Sheehan and Thompson2017; Hemani et al., Reference Hemani, Bowden and Davey Smith2018). Additionally, cis variants are generally less likely to be pleiotropic than trans variants (Schmidt et al., Reference Schmidt, Finan, Gordillo-Marañón, Asselbergs, Freitag, Patel, Tyl, Chopade, Faraway, Zwierzyna and Hingorani2020). Finally, databases (Kamat et al., Reference Kamat, Blackshaw, Young, Surendran, Burgess, Danesh, Butterworth and Staley2019) of genetic associations may be used to test for pleiotropic associations of the genetic variants used as proxies.

In some cases, there may not be genetic variants available to proxy a drug target of interest. This can be due to a lack of sufficient human genetic variation in the gene region that influences the phenotype of interest within a given ancestry group, or there may not be variants at a locus that meet the assumptions for a valid MR analysis (e.g., at a highly pleiotropic locus). This limits the number of genes available for potential investigation using MR. Further, the selection of genes to be investigated is typically guided by the hypothesis or scientific question being addressed in the analysis. Additional limitations include lack of availability of a GWAS of the relevant phenotype or biomarker for that drug target, or lack of a GWAS of molecular data from the appropriate tissue or cell type. Another related limitation is that MR cannot necessarily instrument a drug that concurrently influences multiple parallel biological pathways. Rather, molecular mediators may be proxied individually, or in combination using a factorial MR approach (Yarmolinsky et al., Reference Yarmolinsky, Bull, Walker, Nounu and Smith2020).

Additionally, target phenotypes, disease outcomes, or disease subtypes may not yet be available as outcomes in sufficiently large genetic association datasets. For certain outcomes such as heart failure, clinical trials test interventions separately in patient subgroups, such as in patients with preserved, rather than reduced ejection fraction (Anker et al., Reference Anker, Butler, Filippatos, Ferreira, Bocchi, Bohm, Brunner-La Rocca, Choi, Chopra, Chuquiure-Valenzuela, Giannetti, Gomez-Mesa, Janssens, Januzzi, Gonzalez-Juanatey, Merkely, Nicholls, Perrone, Piña, Ponikowski, Senni, Sim, Spinar, Squire, Taddei, Tsutsui, Verma, Vinereanu, Zhang, Carson, Lam, Marx, Zeller, Sattar, Jamal, Schnaidt, Schnee, Brueckmann, Pocock, Zannad and Packer2021). In such cases, MR cannot yet be implemented to test hypotheses that parallel those investigated in clinical trials. Finally, in contrast to studies of disease susceptibility, data for genetic predictors of disease progression or disease outcomes are scarce (Paternoster et al., Reference Paternoster, Tilling and Davey Smith2017).

The conventional principles that guide interpretation of observational research also apply to MR analyses. Statistical power and precision of effect estimates should always be considered when interpreting a null result, particularly for outcomes with small sample sizes. Most genetic studies are performed using data from individuals of European ancestry and are therefore of unclear generalizability to individuals of different ancestries. This limitation may be addressed by the inclusion of diverse ancestry groups in future GWAS of clinical and molecular traits. When possible, independent replication and triangulation (Lawlor et al., Reference Lawlor, Tilling and Smith2016) of results with those from orthogonal research methodologies enhances confidence in any given finding. Finally, the strength of inference from an MR analysis is heavily influenced by the quality of the phenotype used in genetic association analyses. Modern statistical techniques and large sample sizes cannot overcome biases created by misdiagnosis, diagnostic or phenotypic heterogeneity, and ascertainment bias.

At this point, it is worth noting the similarities and differences between an MR finding and a genetic association at the same locus. For example, GWAS of CAD identify genetic variants in IL6R.(Deloukas et al., Reference Deloukas, Kanoni, Willenborg, Farrall, Assimes, Thompson, Ingelsson, Saleheen, Erdmann, Goldstein, Stirrups, König, Cazier, Johansson, Hall, Lee, Willer, Chambers, Esko, Folkersen, Goel, Grundberg, Havulinna, Ho, Hopewell, Eriksson, Kleber, Kristiansson, Lundmark, Lyytikäinen, Rafelt, Shungin, Strawbridge, Thorleifsson, Tikkanen, Van Zuydam, Voight, Waite, Zhang, Ziegler, Absher, Altshuler, Balmforth, Barroso, Braund, Burgdorf, Claudi-Boehm, Cox, Dimitriou, Do, Doney, El Mokhtari, Eriksson, Fischer, Fontanillas, Franco-Cereceda, Gigante, Groop, Gustafsson, Hager, Hallmans, Han, Hunt, Kang, Illig, Kessler, Knowles, Kolovou, Kuusisto, Langenberg, Langford, Leander, Lokki, Lundmark, McCarthy, Meisinger, Melander, Mihailov, Maouche, Morris, Müller-Nurasyid, Nikus, Peden, Rayner, Rasheed, Rosinger, Rubin, Rumpf, Schäfer, Sivananthan, Song, Stewart, Tan, Thorgeirsson, van der Schoot, Wagner, Wells, Wild, Yang, Amouyel, Arveiler, Basart, Boehnke, Boerwinkle, Brambilla, Cambien, Cupples, de Faire, Dehghan, Diemert, Epstein, Evans, Ferrario, Ferrières, Gauguier, Go, Goodall, Gudnason, Hazen, Holm, Iribarren, Jang, Kähönen, Kee, Kim, Klopp, Koenig, Kratzer, Kuulasmaa, Laakso, Laaksonen, Lee, Lind, Ouwehand, Parish, Park, Pedersen, Peters, Quertermous, Rader, Salomaa, Schadt, Shah, Sinisalo, Stark, Stefansson, Trégouët, Virtamo, Wallentin, Wareham, Zimmermann, Nieminen, Hengstenberg, Sandhu, Pastinen, Syvänen, Hovingh, Dedoussis, Franks, Lehtimäki, Metspalu, Zalloua, Siegbahn, Schreiber, Ripatti, Blankenberg, Perola, Clarke, Boehm, O’Donnell, Reilly, März, Collins, Kathiresan, Hamsten, Kooner, Thorsteinsdottir, Danesh, Palmer, Roberts, Watkins, Schunkert and Samani2013). Indeed, such an association could be used as evidence for a causal effect of IL-6 signaling on CAD risk. The MR paradigm offers several additional benefits. First, the MR paradigm explicitly formalizes the assumptions for causality and provides methods to test the plausibility of the assumptions. For example, numerous sensitivity analyses have been developed that provide results that are robust to inclusion of variants that affect the outcome through pathways unrelated to the exposure (Bowden et al., Reference Bowden, Smith and Burgess2015, Reference Bowden, Davey Smith, Haycock and Burgess2016). Second, MR can be used to produce effect estimates weighted by changes in levels of a clinical phenotype or biomarker. Third, MR can be used to aggregate the effects of multiple independent genetic variants and hence identify effects that are not genome-wide significant when using a single variant in a GWAS. Fourth, in hypothesis-driven MR the association of the variants with the exposure typically does not require as stringent of a statistical significance threshold as does a GWAS for a given locus (Davies et al., Reference Davies, Holmes and Davey Smith2018). Thus, findings from drug target MR analyses should be viewed as complementary to results from a genetic association at a given locus.

Conclusion

There is great potential for MR to aid in the drug development process. Ongoing clinical trials of inhibition of IL-6 signaling and for other drug targets prioritized in MR analyses will ultimately serve as tests for the utility of MR in drug development efforts and the advancement of human health.

Open peer review

To view the open peer review materials for this article, please visit http://doi.org/10.1017/pcm.2023.5.

Data availability statement

Data analysis was not performed for this study.

Acknowledgements

The figures were designed using open-access images from https://www.flaticon.com/.

Author contributions

I.D. and D.G. both made substantial contributions to the conception or design of the work; or the acquisition, analysis, or interpretation of data for the work. I.D. and D.G. both contributed to drafting the work or revising it critically for important intellectual content. I.D. and D.G. both provided final approval of the version to be published. I.D. and D.G. both agree to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Financial support

D.G. is supported by the British Heart Foundation Centre of Research Excellence (RE/18/4/34215) at Imperial College London.

Competing interest

I.D. declares no conflicts of interest. D.G. is employed part-time by Novo Nordisk.

References

Acosta, JN, Szejko, N and Falcone, GJ (2021) Mendelian randomization in stroke: A powerful approach to causal inference and drug target validation. Frontiers in Genetics 12, 683082. https://doi.org/10.3389/fgene.2021.683082.CrossRefGoogle ScholarPubMed
Akhtar, A (2015) The flaws and human harms of animal experimentation. Cambridge Quarterly of Healthcare Ethics 24(4), 407419. https://doi.org/10.1017/S0963180115000079.CrossRefGoogle ScholarPubMed
Andrade, C (2015) The primary outcome measure and its importance in clinical trials. Journal of Clinical Psychiatry 76(10), e1320e1323. https://doi.org/10.4088/JCP.15f10377.CrossRefGoogle ScholarPubMed
Anker, SD, Butler, J, Filippatos, G, Ferreira, JP, Bocchi, E, Bohm, M, Brunner-La Rocca, H-P, Choi, D-J, Chopra, V, Chuquiure-Valenzuela, E, Giannetti, N, Gomez-Mesa, JE, Janssens, S, Januzzi, JL, Gonzalez-Juanatey, JR, Merkely, B, Nicholls, SJ, Perrone, S, Piña, IL, Ponikowski, P, Senni, M, Sim, D, Spinar, J, Squire, I, Taddei, S, Tsutsui, H, Verma, S, Vinereanu, D, Zhang, J, Carson, P, Lam, CSP, Marx, N, Zeller, C, Sattar, N, Jamal, W, Schnaidt, S, Schnee, JM, Brueckmann, M, Pocock, SJ, Zannad, F and Packer, M, for the EMPEROR-Preserved Trial Investigators (2021) Empagliflozin in heart failure with a preserved ejection fraction. New England Journal of Medicine 385(16), 14511461. https://doi.org/10.1056/nejmoa2107038.CrossRefGoogle ScholarPubMed
Backman, JD, Li, AH, Marcketta, A, Sun, D, Mbatchou, J, Kessler, MD, Benner, C, Liu, D, Locke, AE, Balasubramanian, S, Yadav, A, Banerjee, N, Gillies, CE, Damask, A, Liu, S, Bai, X, Hawes, A, Maxwell, E, Gurski, L, Watanabe, K, Kosmicki, JA, Rajagopal, V, Mighty, J, Jones, M, Mitnaul, L, Stahl, E, Coppola, G, Jorgenson, E, Habegger, L, Salerno, WJ, Shuldiner, AR, Lotta, LA, Overton, JD, Cantor, MN, Reid, JG, Yancopoulos, G, Kang, HM, Marchini, J, Baras, A, Abecasis, GR, Ferreira, MAR (2021) Exome sequencing and analysis of 454,787 UK biobank participants. Nature 599(7886), 628634. https://doi.org/10.1038/s41586-021-04103-z.CrossRefGoogle ScholarPubMed
Bailey, J, Thew, M and Balls, M (2014) An analysis of the use of animal models in predicting human toxicology and drug safety. Alternatives to Laboratory Animals : ATLA 42(3), 181199. https://doi.org/10.1177/026119291404200306.CrossRefGoogle ScholarPubMed
Bovijn, J, Lindgren, CM and Holmes, M (2020) Genetic variants mimicking therapeutic inhibition of IL-6 receptor signaling and risk of COVID-19. The Lancet Rheumatology 2, e658e659. https://doi.org/10.1016/S2665.CrossRefGoogle ScholarPubMed
Bowden, J, Davey Smith, G, Haycock, PC and Burgess, S (2016) Consistent estimation in Mendelian randomization with some invalid instruments using a weighted median estimator. Genetic Epidemiology 40(4), 304314. https://doi.org/10.1002/gepi.21965.CrossRefGoogle ScholarPubMed
Bowden, J, Del Greco, MF, Minelli, C, Davey Smith, G, Sheehan, N and Thompson, J (2017) A framework for the investigation of pleiotropy in two-sample summary data Mendelian randomization. Statistics in Medicine 36(11), 17831802. https://doi.org/10.1002/sim.7221.CrossRefGoogle ScholarPubMed
Bowden, J, Smith, GD and Burgess, S (2015) Mendelian randomization with invalid instruments: Effect estimation and bias detection through egger regression. International Journal of Epidemiology 44(2), 512525. https://doi.org/10.1093/ije/dyv080.CrossRefGoogle ScholarPubMed
Burgess, S, Davey Smith, G, Davies, NM, Dudbridge, F, Gill, D, Glymour, MM, Hartwig, FP, Holmes, M, Minelli, C, Relton, CL and Theodoratou, E (2019) Guidelines for performing Mendelian randomization investigations. Wellcome Open Research 4, 186. https://doi.org/10.12688/wellcomeopenres.15555.1.CrossRefGoogle ScholarPubMed
Burgess, S, Davies, NM and Thompson, SG (2014) Instrumental variable analysis with a nonlinear exposure-outcome relationship. Epidemiology 25(6), 877885. https://doi.org/10.1097/EDE.0000000000000161.CrossRefGoogle ScholarPubMed
Burgess, S and Labrecque, JA (2018) Mendelian randomization with a binary exposure variable: Interpretation and presentation of causal estimates. European Journal of Epidemiology 33(10), 947952. https://doi.org/10.1007/s10654-018-0424-6.CrossRefGoogle ScholarPubMed
Burgess, S, Small, DS and Thompson, SG (2017) A review of instrumental variable estimators for Mendelian randomization. Statistical Methods in Medical Research 26(5), 23332355. https://doi.org/10.1177/0962280215597579.CrossRefGoogle ScholarPubMed
Cai, T, Zhang, Y, Ho, YL, Link, N, Sun, J, Huang, J, Cai, TA, Damrauer, S, Ahuja, Y, Honerlaw, J, Huang, J, Costa, L, Schubert, P, Hong, C, Gagnon, D, Sun, YV, Gaziano, JM, Wilson, P, Cho, K, Tsao, P, O’Donnell, CJ, Liao, KP (2018) Association of interleukin 6 receptor variant with cardiovascular disease effects of interleukin 6 receptor blocking therapy: A phenome - Wide association study. JAMA Cardiology 3(9), 849857. https://doi.org/10.1001/jamacardio.2018.2287.CrossRefGoogle ScholarPubMed
Cohen, JC, Boerwinkle, E, Mosley, TH and Hobbs, HH (2006) Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. New England Journal of Medicine 354(12), 12641272. https://doi.org/10.1056/NEJMoa054013.CrossRefGoogle ScholarPubMed
Daghlas, I and Gill, D (2021) Low-density lipoprotein cholesterol and lifespan: A Mendelian randomization study. British Journal of Clinical Pharmacology 87(10), 39163924. https://doi.org/10.1111/bcp.14811.CrossRefGoogle ScholarPubMed
Daghlas, I, Karhunen, V, Ray, D, Zuber, V, Burgess, S, Tsao, PS, Lynch, JA, Lee, KM, Voight, BF, Chang, KM, Baker, EH, Damrauer, SM, Howson, JMM, Vujkovic, M and Gill, D (2021) Genetic evidence for repurposing of GLP1R (glucagon-like peptide-1 receptor) agonists to prevent heart failure. Journal of the American Heart Association 10(13), e020331. https://doi.org/10.1161/JAHA.120.020331.CrossRefGoogle ScholarPubMed
Davies, NM, Holmes, M and Davey Smith, G (2018) Reading Mendelian randomisation studies: A guide, glossary, and checklist for clinicians. BMJ 362, k601. https://doi.org/10.1136/bmj.k601.CrossRefGoogle ScholarPubMed
Deboever, C, Tanigawa, Y, Lindholm, ME, McInnes, G, Lavertu, A, Ingelsson, E, Chang, C, Ashley, EA, Bustamante, CD, Daly, MJ and Rivas, MA (2018) Medical relevance of protein-truncating variants across 337,205 individuals in the UK biobank study. Nature Communications 9(1), 1612. https://doi.org/10.1038/s41467-018-03910-9.CrossRefGoogle ScholarPubMed
Deloukas, P, Kanoni, S, Willenborg, C, Farrall, M, Assimes, TL, Thompson, JR, Ingelsson, E, Saleheen, D, Erdmann, J, Goldstein, BA, Stirrups, K, König, IR, Cazier, JB, Johansson, A, Hall, AS, Lee, JY, Willer, CJ, Chambers, JC, Esko, T, Folkersen, L, Goel, A, Grundberg, E, Havulinna, AS, Ho, WK, Hopewell, JC, Eriksson, N, Kleber, ME, Kristiansson, K, Lundmark, P, Lyytikäinen, LP, Rafelt, S, Shungin, D, Strawbridge, RJ, Thorleifsson, G, Tikkanen, E, Van Zuydam, N, Voight, BF, Waite, LL, Zhang, W, Ziegler, A, Absher, D, Altshuler, D, Balmforth, AJ, Barroso, I, Braund, PS, Burgdorf, C, Claudi-Boehm, S, Cox, D, Dimitriou, M, Do, R, Doney, AS, El Mokhtari, N, Eriksson, P, Fischer, K, Fontanillas, P, Franco-Cereceda, A, Gigante, B, Groop, L, Gustafsson, S, Hager, J, Hallmans, G, Han, BG, Hunt, SE, Kang, HM, Illig, T, Kessler, T, Knowles, JW, Kolovou, G, Kuusisto, J, Langenberg, C, Langford, C, Leander, K, Lokki, ML, Lundmark, A, McCarthy, MI, Meisinger, C, Melander, O, Mihailov, E, Maouche, S, Morris, AD, Müller-Nurasyid, M, Nikus, K, Peden, JF, Rayner, NW, Rasheed, A, Rosinger, S, Rubin, D, Rumpf, MP, Schäfer, A, Sivananthan, M, Song, C, Stewart, AF, Tan, ST, Thorgeirsson, G, van der Schoot, CE, Wagner, PJ, Wells, GA, Wild, PS, Yang, TP, Amouyel, P, Arveiler, D, Basart, H, Boehnke, M, Boerwinkle, E, Brambilla, P, Cambien, F, Cupples, AL, de Faire, U, Dehghan, A, Diemert, P, Epstein, SE, Evans, A, Ferrario, MM, Ferrières, J, Gauguier, D, Go, AS, Goodall, AH, Gudnason, V, Hazen, SL, Holm, H, Iribarren, C, Jang, Y, Kähönen, M, Kee, F, Kim, HS, Klopp, N, Koenig, W, Kratzer, W, Kuulasmaa, K, Laakso, M, Laaksonen, R, Lee, JY, Lind, L, Ouwehand, WH, Parish, S, Park, JE, Pedersen, NL, Peters, A, Quertermous, T, Rader, DJ, Salomaa, V, Schadt, E, Shah, SH, Sinisalo, J, Stark, K, Stefansson, K, Trégouët, DA, Virtamo, J, Wallentin, L, Wareham, N, Zimmermann, ME, Nieminen, MS, Hengstenberg, C, Sandhu, MS, Pastinen, T, Syvänen, AC, Hovingh, GK, Dedoussis, G, Franks, PW, Lehtimäki, T, Metspalu, A, Zalloua, PA, Siegbahn, A, Schreiber, S, Ripatti, S, Blankenberg, SS, Perola, M, Clarke, R, Boehm, BO, O’Donnell, C, Reilly, MP, März, W, Collins, R, Kathiresan, S, Hamsten, A, Kooner, JS, Thorsteinsdottir, U, Danesh, J, Palmer, CN, Roberts, R, Watkins, H, Schunkert, H, Samani, NJ (2013) Large-scale association analysis identifies new risk loci for coronary artery disease. Nature Genetics 45(1), 2533. https://doi.org/10.1038/ng.2480.CrossRefGoogle ScholarPubMed
Denny, JC, Ritchie, MD, Basford, MA, Pulley, JM, Bastarache, L, Brown-Gentry, K, Wang, D, Masys, DR, Roden, DM and Crawford, DC (2010) PheWAS: Demonstrating the feasibility of a phenome-wide scan to discover gene-disease associations. Bioinformatics 26(9), 12051210. https://doi.org/10.1093/bioinformatics/btq126.CrossRefGoogle ScholarPubMed
Devauchelle-Pensec, V, Carvajal-Alegria, G, Dernis, E, Richez, C, Truchetet, ME, Wendling, D, Toussirot, E, Perdriger, A, Gottenberg, JE, Felten, R, Fautrel, BJ, Chiche, L, Hilliquin, P, Le Henaff, C, Dervieux, B, Direz, G, Chary-Valckenaere, I, Cornec, D, Guellec, D, Marhadour, T, Nowak, E, Saraux, A (2022) Effect of tocilizumab on disease activity in patients with active polymyalgia rheumatica receiving glucocorticoid therapy: A randomized clinical trial. JAMA 328(11), 10531062. https://doi.org/10.1001/jama.2022.15459.CrossRefGoogle ScholarPubMed
DiMasi, JA, Hansen, RW and Grabowski, HG (2003) The price of innovation: New estimates of drug development costs. Journal of Health Economics 22(2), 151185. https://doi.org/10.1016/S0167-6296(02)00126-1.CrossRefGoogle Scholar
Elliott, LT, Sharp, K, Alfaro-Almagro, F, Shi, S, Miller, KL, Douaud, G, Marchini, J and Smith, SM (2018) Genome-wide association studies of brain imaging phenotypes in UK biobank. Nature 562(7726), 210216. https://doi.org/10.1038/s41586-018-0571-7.CrossRefGoogle ScholarPubMed
Emdin, CA, Khera, AV, Chaffin, M, Klarin, D, Natarajan, P, Aragam, K, Haas, M, Bick, A, Zekavat, SM, Nomura, A, Ardissino, D, Wilson, JG, Schunkert, H, McPherson, R, Watkins, H, Elosua, R, Bown, MJ, Samani, NJ, Baber, U, Erdmann, J, Gupta, N, Danesh, J, Chasman, D, Ridker, P, Denny, J, Bastarache, L, Lichtman, JH, D’Onofrio, G, Mattera, J, Spertus, JA, Sheu, WH, Taylor, KD, Psaty, BM, Rich, SS, Post, W, Rotter, JI, Chen, YI, Krumholz, H, Saleheen, D, Gabriel, S, and Kathiresan, S (2018) Analysis of predicted loss-of-function variants in UK biobank identifies variants protective for disease. Nature Communications 9(1), 1613. https://doi.org/10.1038/s41467-018-03911-8.CrossRefGoogle ScholarPubMed
Fauman, EB and Hyde, C (2022) An optimal variant to gene distance window derived from an empirical definition of cis and trans protein QTLs. BMC Bioinformatics 23(1), 169. https://doi.org/10.1186/s12859-022-04706-x.CrossRefGoogle ScholarPubMed
Ference, BA, Ginsberg, HN, Graham, I, Ray, KK, Packard, CJ, Bruckert, E, Hegele, RA, Krauss, RM, Raal, FJ, Schunkert, H, Watts, GF, Borén, J, Fazio, S, Horton, JD, Masana, L, Nicholls, SJ, Nordestgaard, BG, van de Sluis, B, Taskinen, MR, Tokgözoglu, L, Landmesser, U, Laufs, U, Wiklund, O, Stock, JK, Chapman, MJ, and Catapano, AL (2017) Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement fromthe European atherosclerosis society consensus panel. European Heart Journal 38(32), 24592472. https://doi.org/10.1093/eurheartj/ehx144.CrossRefGoogle Scholar
Ferreira, RC, Freitag, DF, Cutler, AJ, Howson, JMM, Rainbow, DB, Smyth, DJ, Kaptoge, S, Clarke, P, Boreham, C, Coulson, RM, Pekalski, ML, Chen, WM, Onengut-Gumuscu, S, Rich, SS, Butterworth, AS, Malarstig, A, Danesh, J and Todd, JA (2013) Functional IL6R 358Ala allele impairs classical IL-6 receptor Signaling and influences risk of diverse inflammatory diseases. PLoS Genetics 9(4), e1003444. https://doi.org/10.1371/journal.pgen.1003444.CrossRefGoogle ScholarPubMed
Finan, C, Gaulton, A, Kruger, FA, Lumbers, RT, Shah, T, Engmann, J, Galver, L, Kelley, R, Karlsson, A, Santos, R, Overington, JP, Hingorani, AD and Casas, JP (2017) The druggable genome and support for target identification and validation in drug development. Science Translational Medicine 9(383), eaag1166. https://doi.org/10.1126/scitranslmed.aag1166.CrossRefGoogle ScholarPubMed
Folkersen, L, Gustafsson, S, Wang, Q, Hansen, DH, Hedman, ÅK, Schork, A, Page, K, Zhernakova, DV, Wu, Y, Peters, J, Eriksson, N, Bergen, SE, Boutin, TS, Bretherick, AD, Enroth, S, Kalnapenkis, A, Gådin, JR, Suur, BE, Chen, Y, Matic, L, Gale, JD, Lee, J, Zhang, W, Quazi, A, Ala-Korpela, M, Choi, SH, Claringbould, A, Danesh, J, Davey Smith, G, de Masi, F, Elmståhl, S, Engström, G, Fauman, E, Fernandez, C, Franke, L, Franks, PW, Giedraitis, V, Haley, C, Hamsten, A, Ingason, A, Johansson, Å, Joshi, PK, Lind, L, Lindgren, CM, Lubitz, S, Palmer, T, Macdonald-Dunlop, E, Magnusson, M, Melander, O, Michaelsson, K, Morris, AP, Mägi, R, Nagle, MW, Nilsson, PM, Nilsson, J, Orho-Melander, M, Polasek, O, Prins, B, Pålsson, E, Qi, T, Sjögren, M, Sundström, J, Surendran, P, Võsa, U, Werge, T, Wernersson, R, Westra, HJ, Yang, J, Zhernakova, A, Ärnlöv, J, Fu, J, Smith, JG, Esko, T, Hayward, C, Gyllensten, U, Landen, M, Siegbahn, A, Wilson, JF, Wallentin, L, Butterworth, AS, Holmes, MV, Ingelsson, E, and Mälarstig, A (2020) Genomic and drug target evaluation of 90 cardiovascular proteins in 30,931 individuals. Nature Metabolism 2(10), 11351148. https://doi.org/10.1038/s42255-020-00287-2.CrossRefGoogle ScholarPubMed
Forgetta, V, Jiang, L, Vulpescu, NA, Hogan, MS, Chen, S, Morris, JA, Grinek, S, Benner, C, Jang, DK, Hoang, Q, Burtt, N, Flannick, JA, McCarthy, MI, Fauman, E, Greenwood, CMT, Maurano, MT and Richards, JB (2022) An effector index to predict target genes at GWAS loci. Human Genetics 141(8), 14311447. https://doi.org/10.1007/s00439-022-02434-z.CrossRefGoogle ScholarPubMed
Fuchs, VR (2010) New priorities for future biomedical innovations. New England Journal of Medicine 363(8), 704706. https://doi.org/10.1056/NEJMp0906597.CrossRefGoogle ScholarPubMed
Gallagher, MD and Chen-Plotkin, AS (2018) The post-GWAS era: From association to function. American Journal of Human Genetics 102(5), 717730. https://doi.org/10.1016/j.ajhg.2018.04.002.CrossRefGoogle ScholarPubMed
Georgakis, MK, Malik, R, Burgess, S and Dichgans, M (2022) Additive effects of genetic interleukin-6 signaling downregulation and low-density lipoprotein cholesterol lowering on cardiovascular disease: A 2×2 factorial Mendelian randomization analysis. Journal of the American Heart Association 11(1), e023277. https://doi.org/10.1161/JAHA.121.023277.CrossRefGoogle ScholarPubMed
Georgakis, MK, Malik, R, Gill, D, Franceschini, N, Sudlow, CLM and Dichgans, M (2020) Interleukin-6 signaling effects on ischemic stroke and other cardiovascular outcomes: A Mendelian randomization study. Circulation: Genomic and Precision Medicine 13(3), E002872. https://doi.org/10.1161/CIRCGEN.119.002872.Google ScholarPubMed
Georgakis, MK, Malik, R, Li, X, Gill, D, Levin, MG, Vy, HMT, Judy, R, Ritchie, M, Verma, SS, Nadkarni, GN, Damrauer, SM, Theodoratou, E and Dichgans, M (2021) Genetically downregulated interleukin-6 signaling is associated with a favorable cardiometabolic profile: A phenome-wide association study. Circulation 143(11), 11771180. https://doi.org/10.1161/CIRCULATIONAHA.120.052604.CrossRefGoogle ScholarPubMed
Georgakis, MK, Malik, R, Richardson, TG, Howson, JMM, Anderson, CD, Burgess, S, Hovingh, GK, Dichgans, M and Gill, D (2022) Associations of genetically predicted IL-6 signaling with cardiovascular disease risk across population subgroups. BMC Medicine 20(1), 245. https://doi.org/10.1186/s12916-022-02446-6.CrossRefGoogle ScholarPubMed
Gill, D and Burgess, S (2020) Use of a genetic variant related to circulating FXa (activated factor X) levels to proxy the effect of FXa inhibition on cardiovascular outcomes. Circulation: Genomic and Precision Medicine 13, 551553. https://doi.org/10.1161/CIRCGEN.120.003061.Google ScholarPubMed
Gill, D, Georgakis, MK, Koskeridis, F, Jiang, L, Feng, Q, Wei, WQ, Theodoratou, E, Elliott, P, Denny, JC, Malik, R, Evangelou, E, Dehghan, A, Dichgans, M and Tzoulaki, I (2019) Use of genetic variants related to antihypertensive drugs to inform on efficacy and side effects. Circulation 140(4), 270279. https://doi.org/10.1161/CIRCULATIONAHA.118.038814.CrossRefGoogle ScholarPubMed
Gill, D, Georgakis, MK, Walker, VM, Schmidt, AF, Gkatzionis, A, Freitag, DF, Finan, C, Hingorani, AD, Howson, JMM, Burgess, S, Swerdlow, DI, Smith, GD, Holmes, M, Dichgans, M, Zheng, J, Psaty, BM and Davies, NM (2021) Mendelian randomization for studying the effects of perturbing drug targets [version 1; Peer review: Awaiting peer review]. Wellcome Open Research 6, 111. https://doi.org/10.12688/WELLCOMEOPENRES.16544.1.CrossRefGoogle ScholarPubMed
Gill, D and Vujkovic, M (2022) The potential of genetic data for prioritizing drug repurposing efforts. Neurology 99(7), 267268. https://doi.org/10.1212/WNL.0000000000200803.CrossRefGoogle ScholarPubMed
Begley, CG, Ashton, M, Baell, J, Bettess, M, Brown, MP, Carter, B, Charman, WN, Davis, C, Fisher, S, Frazer, I, Gautam, A, Jennings, MP, Kearney, P, Keeffe, E, Kelly, D, Lopez, AF, McGuckin, M, Parker, MW, Rayner, C, Roberts, B, Rush, JS Sullivan, M (2021) Drug repurposing: Misconceptions, challenges, and opportunities for academic researchers. Science Translational Medicine 13(612). https://doi.org/10.1126/scitranslmed.abd5524CrossRefGoogle ScholarPubMed
Hall, SS (2013) A gene of rare effect. Nature 496(7444), 152155. https://doi.org/10.1038/496152a.CrossRefGoogle ScholarPubMed
Hay, M, Thomas, DW, Craighead, JL, Economides, C and Rosenthal, J (2014) Clinical development success rates for investigational drugs. Nature Biotechnology 32(1), 4051. https://doi.org/10.1038/nbt.2786.CrossRefGoogle ScholarPubMed
Hemani, G, Bowden, J and Davey Smith, G (2018) Evaluating the potential role of pleiotropy in Mendelian randomization studies. Human Molecular Genetics 27(R2), R195R208. https://doi.org/10.1093/hmg/ddy163.CrossRefGoogle ScholarPubMed
Hemani, G, Zheng, J, Elsworth, B, Wade, KH, Haberland, V, Baird, D, Laurin, C, Burgess, S, Bowden, J, Langdon, R, Tan, VY, Yarmolinsky, J, Shihab, HA, Timpson, NJ, Evans, DM, Relton, C, Martin, RM, Davey Smith, G, Gaunt, TR and Haycock, PC (2018) The MR-Base platform supports systematic causal inference across the human phenome. https://doi.org/10.7554/eLife.34408.001.CrossRefGoogle Scholar
Henry, A, Gordillo-Marañón, M, Finan, C, Schmidt, AF, Ferreira, JP, Karra, R, Sundström, J, Lind, L, Arnlöv, J, Zannad, F, Mälarstig, A, Hingorani, AD and Lumbers, RT (2022) Therapeutic targets for heart failure identified using proteomics and Mendelian randomization. Circulation 145(16), 12051217. https://doi.org/10.1161/CIRCULATIONAHA.121.056663.CrossRefGoogle ScholarPubMed
Kamat, MA, Blackshaw, JA, Young, R, Surendran, P, Burgess, S, Danesh, J, Butterworth, AS and Staley, JR (2019) PhenoScanner V2: An expanded tool for searching human genotype-phenotype associations. Bioinformatics 35(22), 48514853. https://doi.org/10.1093/bioinformatics/btz469.CrossRefGoogle ScholarPubMed
Kang, S, Tanaka, T, Narazaki, M and Kishimoto, T (2019) Targeting interleukin-6 signaling in clinic. Immunity 50(4), 10071023. https://doi.org/10.1016/j.immuni.2019.03.026.CrossRefGoogle ScholarPubMed
Karatasakis, A, Danek, BA, Karacsonyi, J, Rangan, B, Roesle, MK, Knickelbine, T, Miedema, MD, Khalili, H, Ahmad, Z, Abdullah, S, Banerjee, S and Brilakis, ES (2017) Effect of PCSK9 inhibitors on clinical outcomes in patients with hypercholesterolemia: A meta-analysis of 35 randomized controlled trials. Journal of the American Heart Association 6(12), e006910. https://doi.org/10.1161/JAHA.117.006910.CrossRefGoogle ScholarPubMed
King, EA, Wade Davis, J and Degner, JF (2019) Are drug targets with genetic support twice as likely to be approved? Revised estimates of the impact of genetic support for drug mechanisms on the probability of drug approval. PLoS Genetics 15(12), e1008489. https://doi.org/10.1371/journal.pgen.1008489.CrossRefGoogle ScholarPubMed
Kurki, M, Karjalainen, J, Palta, P, Sipilä, T, Kristiansson, K, Donner, K, Reeve, M, Laivuori, H, Aavikko, M, Kaunisto, M, Loukola, A, Lahtela, E, Mattsson, H, Laiho, P, Della Briotta Parolo, P, Lehisto, A, Kanai, M, Mars, N, Rämö, J, Kiiskinen, T, Heyne, H, Veerapen, K, Rüeger, S, Lemmelä, S, Zhou, W, Ruotsalainen, S, Pärn, K, Hiekkalinna, T, Koskelainen, S, Paajanen, T, Llorens, V, Gracia-Tabuenca, J, Siirtola, H, Reis, K, Elnahas, A, Aalto-Setälä, K, Alasoo, K, Arvas, M, Auro, K, Biswas, S, Bizaki-Vallaskangas, A, Carpen, O, Chen, C, Dada, O, Ding, Z, Ehm, M, Eklund, K, Färkkilä, M, Finucane, H, Ganna, A, Ghazal, A, Graham, R, Green, E, Hakanen, A, Hautalahti, M, Hedman, Å, Hiltunen, M, Hinttala, R, Hovatta, I, Hu, X, Huertas-Vazquez, A, Huilaja, L, Hunkapiller, J, Jacob, H, Jensen, J, Joensuu, H, John, S, Julkunen, V, Jung, M, Junttila, J, Kaarniranta, K, Kähönen, M, Kajanne, R, Kallio, L, Kälviäinen, R, Kaprio, J, Kerimov, N, Kettunen, J, Kilpeläinen, E, Kilpi, T, Klinger, K, Kosma, V, Kuopio, T, Kurra, V, Laisk, T, Laukkanen, J, Lawless, N, Liu, A, Longerich, S, Mägi, R, Mäkelä, J, Mäkitie, A, Malarstig, A, Mannermaa, A, Maranville, J, Matakidou, A, Meretoja, T, Mozaffari, S, Niemi, M, Niemi, M, Niiranen, T, O’Donnell, C, Obeidat, M, Okafo, G, Ollila, H, Palomäki, A, Palotie, T, Partanen, J, Paul, D, Pelkonen, M, Pendergrass, R, Petrovski, S, Pitkäranta, A, Platt, A, Pulford, D, Punkka, E, Pussinen, P, Raghavan, N, Rahimov, F, Rajpal, D, Renaud, N, Riley-Gillis, B, Rodosthenous, R, Saarentaus, E, Salminen, A, Salminen, E, Salomaa, V, Schleutker, J, Serpi, R, Shen, H, Siegel, R, Silander, K, Siltanen, S, Soini, S, Soininen, H, Sul, J, Tachmazidou, I, Tasanen, K, Tienari, P, Toppila-Salmi, S, Tukiainen, T, Tuomi, T, Turunen, J, Ulirsch, J, Vaura, F, Virolainen, P, Waring, J, Waterworth, D, Yang, R, Nelis, M, Reigo, A, Metspalu, A, Milani, L, Esko, T, Fox, C, Havulinna, A, Perola, M, Ripatti, S, Jalanko, A, Laitinen, T, Mäkelä, T, Plenge, R, McCarthy, M, Runz, H, Daly, M, and Palotie, A (2022) FinnGen: Unique genetic insights from combining isolated population and national health register data. MedRxiv. https://doi.org/10.1101/2022.03.03.22271360.Google Scholar
Larsson, SC, Burgess, S and Gill, D (2021) Genetically proxied interleukin-6 receptor inhibition: Opposing associations with COVID-19 and pneumonia. European Respiratory Journal 57(1), 2003545. https://doi.org/10.1183/13993003.03545-2020.CrossRefGoogle ScholarPubMed
Lawlor, DA, Harbord, RM, Sterne, JAC, Timpson, N and Smith, GD (2008) Mendelian randomization: Using genes as instruments for making causal inferences in epidemiology. Statistics in Medicine 27(8), 11331163. https://doi.org/10.1002/sim.3034.CrossRefGoogle ScholarPubMed
Lawlor, DA, Tilling, K and Smith, GD (2016) Triangulation in aetiological epidemiology. International Journal of Epidemiology 45(6), 18661886. https://doi.org/10.1093/ije/dyw314.Google ScholarPubMed
Levin, MG, Klarin, D, Georgakis, MK, Lynch, J, Liao, KP, Voight, BF, O’Donnell, CJ, Chang, KM, Assimes, TL, Tsao, PS and Damrauer, SM (2021) A missense variant in the IL-6 receptor and protection from peripheral artery disease. Circulation Research 129, 968970. https://doi.org/10.1161/CIRCRESAHA.121.319589.CrossRefGoogle ScholarPubMed
Lichtenberg, FR (2019) The impact of pharmaceutical innovation on the burden of disease in Canada, 2000–2016. SSM - Population Health 8, 100457. https://doi.org/10.1016/j.ssmph.2019.100457.CrossRefGoogle ScholarPubMed
Mountjoy, E, Schmidt, EM, Carmona, M, Schwartzentruber, J, Peat, G, Miranda, A, Fumis, L, Hayhurst, J, Buniello, A, Karim, MA, Wright, D, Hercules, A, Papa, E, Fauman, EB, Barrett, JC, Todd, JA, Ochoa, D, Dunham, I and Ghoussaini, M (2021) An open approach to systematically prioritize causal variants and genes at all published human GWAS trait-associated loci. Nature Genetics 53(11), 15271533. https://doi.org/10.1038/s41588-021-00945-5.CrossRefGoogle ScholarPubMed
Nelson, MR, Tipney, H, Painter, JL, Shen, J, Nicoletti, P, Shen, Y, Floratos, A, Sham, PC, Li, MJ, Wang, J, Cardon, LR, Whittaker, JC and Sanseau, P (2015) The support of human genetic evidence for approved drug indications. Nature Genetics 47(8), 856860. https://doi.org/10.1038/ng.3314.CrossRefGoogle ScholarPubMed
Oktora, MP, Denig, P, Bos, JHJ, Schuiling-Veninga, CCM and Hak, E (2019) Trends in polypharmacy and dispensed drugs among adults in the Netherlands as compared to the United States. PLoS One 14(3), e0214240. https://doi.org/10.1371/journal.pone.0214240.CrossRefGoogle ScholarPubMed
Paglialunga, S, Morimoto, BH, Clark, M and Friedrichs, GS (2019) Translatability of the S7A core battery respiratory safety pharmacology studies: Preclinical respiratory and related clinical adverse events. Journal of Pharmacological and Toxicological Methods 99, 106596. https://doi.org/10.1016/j.vascn.2019.106596.CrossRefGoogle ScholarPubMed
Paternoster, L, Tilling, K and Davey Smith, G (2017) Genetic epidemiology and Mendelian randomization for informing disease therapeutics: Conceptual and methodological challenges. PLoS Genetics 13(10), e1006944. https://doi.org/10.1371/journal.pgen.1006944.CrossRefGoogle ScholarPubMed
Perel, P, Roberts, I, Sena, E, Wheble, P, Briscoe, C, Sandercock, P, Macleod, M, Mignini, LE, Jayaram, P and Khan, KS (2007) Comparison of treatment effects between animal experiments and clinical trials: Systematic review. British Medical Journal 334(7586), 197200. https://doi.org/10.1136/bmj.39048.407928.BE.CrossRefGoogle ScholarPubMed
Pietzner, M, Wheeler, E, Carrasco-Zanini, J, Cortes, A, Koprulu, M, Wörheide, MA, Oerton, E, Cook, J, Stewart, ID, Kerrison, ND, Luan, J, Raffler, J, Arnold, M, Arlt, W, O’Rahilly, S, Kastenmüller, G, Gamazon, ER, Hingorani, AD, Scott, RA, Wareham, NJ, and Langenberg, C (2021) Mapping the proteo-genomic convergence of human diseases. Science 374(6569), eabj1541. https://doi.org/10.1126/science.abj1541.CrossRefGoogle ScholarPubMed
Porcu, E, Rüeger, S, Lepik, K, Santoni, FA, Reymond, A, and Kutalik, Z (2019) Mendelian randomization integrating GWAS and eQTL data reveals genetic determinants of complex and clinical traits. Nature Communications 10(1), 3300. https://doi.org/10.1038/s41467-019-10936-0.CrossRefGoogle ScholarPubMed
Price, AL, Patterson, NJ, Plenge, RM, Weinblatt, ME, Shadick, NA and Reich, D (2006) Principal components analysis corrects for stratification in genome-wide association studies. Nature Genetics 38(8), 904909. https://doi.org/10.1038/ng1847.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 (2018) Drug repurposing: Progress, challenges and recommendations. Nature Reviews Drug Discovery 18(1), 4158. https://doi.org/10.1038/nrd.2018.168.CrossRefGoogle ScholarPubMed
Rahman, RP, McEwan, L, Ryan, DK and Gill, D (2022) Leveraging genetic data to investigate the effects of interleukin-6 receptor signalling on levels of 40 circulating cytokines. British Journal of Clinical Pharmacology 88(3), 13731378. https://doi.org/10.1111/bcp.15079.CrossRefGoogle ScholarPubMed
Rajasundaram, S, Burgess, S and Gill, D (2022) Treatment of severe covid-19 with interleukin 6 receptor inhibition. BMJ Medicine 1(1), e000144. https://doi.org/10.1136/bmjmed-2022-000144.CrossRefGoogle Scholar
Rees, JMB, Foley, CN and Burgess, S (2020) Factorial Mendelian randomization: Using genetic variants to assess interactions. International Journal of Epidemiology 49(4), 11471158. https://doi.org/10.1093/ije/dyz161.CrossRefGoogle ScholarPubMed
Ridker, PM and Rane, M (2021) Interleukin-6 signaling and anti-interleukin-6 therapeutics in cardiovascular disease. Circulation Research 128(11), 17281746. https://doi.org/10.1161/CIRCRESAHA.121.319077.CrossRefGoogle ScholarPubMed
Rosa, M, Chignon, A, Li, Z, Boulanger, MC, Arsenault, BJ, Bossé, Y, Thériault, S and Mathieu, P (2019) A Mendelian randomization study of IL6 signaling in cardiovascular diseases, immune-related disorders and longevity. NPJ Genomic Medicine 4(1), 23. https://doi.org/10.1038/s41525-019-0097-4.CrossRefGoogle ScholarPubMed
Rudmann, DG (2013) On-target and off-target-based toxicologic effects. Toxicologic Pathology 41(2), 310314. https://doi.org/10.1177/0192623312464311.CrossRefGoogle ScholarPubMed
Said, S, Pazoki, R, Karhunen, V, Võsa, U, Ligthart, S, Bodinier, B, Koskeridis, F, Welsh, P, Alizadeh, BZ, Chasman, DI, Sattar, N, Chadeau-Hyam, M, Evangelou, E, Jarvelin, MR, Elliott, P, Tzoulaki, I and Dehghan, A (2022) Genetic analysis of over half a million people characterises C-reactive protein loci. Nature Communications 13(1), 2198. https://doi.org/10.1038/s41467-022-29650-5.CrossRefGoogle ScholarPubMed
Santos, R, Ursu, O, Gaulton, A, Bento, AP, Donadi, RS, Bologa, CG, Karlsson, A, Al-Lazikani, B, Hersey, A, Oprea, TI and Overington, JP (2016) A comprehensive map of molecular drug targets. Nature Reviews Drug Discovery 16(1), 1934. https://doi.org/10.1038/nrd.2016.230.CrossRefGoogle ScholarPubMed
Sarwar, N, Adam, D and Butterworth, S (2012) Interleukin-6 receptor pathways in coronary heart disease: A collaborative meta-analysis of 82 studies IL6R genetics consortium and emerging risk factors collaboration. The Lancet 379, 12051213. https://doi.org/10.1016/S0140.Google Scholar
Schlander, M, Hernandez-Villafuerte, K, Cheng, CY, Mestre-Ferrandiz, J and Baumann, M (2021) How much does it cost to research and develop a new drug? A systematic review and assessment. PharmacoEconomics 39(11), 12431269. https://doi.org/10.1007/s40273-021-01065-y.CrossRefGoogle ScholarPubMed
Schmidt, AF, Finan, C, Gordillo-Marañón, M, Asselbergs, FW, Freitag, DF, Patel, RS, Tyl, B, Chopade, S, Faraway, R, Zwierzyna, M and Hingorani, AD (2020) Genetic drug target validation using Mendelian randomisation. Nature Communications 11(1), 3255. https://doi.org/10.1038/s41467-020-16969-0.CrossRefGoogle ScholarPubMed
Shapiro, MD, Tavori, H and Fazio, S (2018) PCSK9 from basic science discoveries to clinical trials. Circulation Research 122(10), 14201438. https://doi.org/10.1161/CIRCRESAHA.118.311227.CrossRefGoogle ScholarPubMed
Smith, GD and Ebrahim, S (2003) “Mendelian randomization”: Can genetic epidemiology contribute to understanding environmental determinants of disease? International Journal of Epidemiology 32(1), 122. https://doi.org/10.1093/ije/dyg070.CrossRefGoogle ScholarPubMed
Smith, GD and Ebrahim, S (2004) Mendelian randomization: Prospects, potentials, and limitations. International Journal of Epidemiology 33(1), 3042. https://doi.org/10.1093/ije/dyh132.CrossRefGoogle ScholarPubMed
Stefaniak, J and Huber, KVM (2020) Importance of quantifying drug-target engagement in cells. ACS Medicinal Chemistry Letters 11(4), 403406. https://doi.org/10.1021/acsmedchemlett.9b00570.CrossRefGoogle ScholarPubMed
Sudlow, C, Gallacher, J, Allen, N, Beral, V, Burton, P, Danesh, J, Downey, P, Elliott, P, Green, J, Landray, M, Liu, B, Matthews, P, Ong, G, Pell, J, Silman, A, Young, A, Sprosen, T, Peakman, T and Collins, R (2015) UK biobank: An open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Medicine 12(3), e1001779. https://doi.org/10.1371/journal.pmed.1001779.CrossRefGoogle ScholarPubMed
Suhre, K, McCarthy, MI and Schwenk, JM (2021) Genetics meets proteomics: Perspectives for large population-based studies. Nature Reviews Genetics 22(1), 1937. https://doi.org/10.1038/s41576-020-0268-2.CrossRefGoogle ScholarPubMed
Swerdlow, DI, Kuchenbaecker, KB, Shah, S, Sofat, R, Holmes, M, White, J, Mindell, JS, Kivimaki, M, Brunner, EJ, Whittaker, JC, Casas, JP and Hingorani, AD (2016) Selecting instruments for Mendelian randomization in the wake of genome-wide association studies. International Journal of Epidemiology 45(5), 16001616. https://doi.org/10.1093/ije/dyw088.CrossRefGoogle ScholarPubMed
Teumer, A (2018) Common methods for performing Mendelian randomization. Frontiers in Cardiovascular Medicine 5, 51. https://doi.org/10.3389/fcvm.2018.00051.CrossRefGoogle ScholarPubMed
Timmers, PR, Mounier, N, Lall, K, Fischer, K, Ning, Z, Feng, X, Bretherick, AD, Clark, DW, Shen, X, Esko, T, Kutalik, Z, Wilson, JF, and Joshi, PK (2019) Genomics of 1 million parent lifespans implicates novel pathways and common diseases and distinguishes survival chances. eLife 8, e39856. https://doi.org/10.7554/eLife.39856.CrossRefGoogle ScholarPubMed
Toshner, M, Church, C, Harbaum, L, Rhodes, C, Villar Moreschi, SS, Liley, J, Jones, R, Arora, A, Batai, K, Desai, AA, Coghlan, JG, Gibbs, JSR, Gor, D, Gräf, S, Harlow, L, Hernandez-Sanchez, J, Howard, LS, Humbert, M, Karnes, J, Kiely, DG, Kittles, R, Knightbridge, E, Lam, B, Lutz, KA, Nichols, WC, Pauciulo, MW, Pepke-Zaba, J, Suntharalingam, J, Soubrier, F, Trembath, RC, Schwantes-An, TL, Wort, SJ, Wilkins, MR, Gaine, S, Morrell, NW, and Corris, PA (2022) Mendelian randomisation and experimental medicine approaches to interleukin-6 as a drug target in pulmonary arterial hypertension. European Respiratory Journal 59(3), 2002463. https://doi.org/10.1183/13993003.02463-2020.CrossRefGoogle ScholarPubMed
van Norman, GA (2019) Limitations of animal studies for predicting toxicity in clinical trials: Is it time to rethink our current approach? JACC: Basic to Translational Science 4(7), 845854. https://doi.org/10.1016/j.jacbts.2019.10.008.Google ScholarPubMed
Verbanck, M, Chen, CY, Neale, B and Do, R (2018) Detection of widespread horizontal pleiotropy in causal relationships inferred from Mendelian randomization between complex traits and diseases. Nature Genetics 50(5), 693698. https://doi.org/10.1038/s41588-018-0099-7.CrossRefGoogle ScholarPubMed
Yang, C, Farias, FHG, Ibanez, L, Suhy, A, Sadler, B, Fernandez, MV, Wang, F, Bradley, JL, Eiffert, B, Bahena, JA, Budde, JP, Li, Z, Dube, U, Sung, YJ, Mihindukulasuriya, KA, Morris, JC, Fagan, AM, Perrin, RJ, Benitez, BA, Rhinn, H, Harari, O, and Cruchaga, C (2021) Genomic atlas of the proteome from brain, CSF and plasma prioritizes proteins implicated in neurological disorders. Nature Neuroscience 24(9), 13021312. https://doi.org/10.1038/s41593-021-00886-6.CrossRefGoogle ScholarPubMed
Yao, C, Chen, G, Song, C, Keefe, J, Mendelson, M, Huan, T, Sun, BB, Laser, A, Maranville, JC, Wu, H, Ho, JE, Courchesne, P, Lyass, A, Larson, MG, Gieger, C, Graumann, J, Johnson, AD, Danesh, J, Runz, H, Hwang, SJ, Liu, C, Butterworth, AS, Suhre, K, and Levy, D (2018) Genome‐wide mapping of plasma protein QTLs identifies putatively causal genes and pathways for cardiovascular disease. Nature Communications 9(1), 3268. https://doi.org/10.1038/s41467-018-05512-x.CrossRefGoogle Scholar
Yarmolinsky, J, Bull, CJ, Walker, VM, Nounu, A and Smith, GD (2020) Mendelian randomization applied to pharmaceutical use: The case of metformin and lung cancer. International Journal of Epidemiology 49(4), 14101411. https://doi.org/10.1093/ije/dyaa059.CrossRefGoogle ScholarPubMed
Yavorska, OO and Burgess, S (2017) Mendelian randomization: An R package for performing Mendelian randomization analyses using summarized data. International Journal of Epidemiology 46(6), 17341739. https://doi.org/10.1093/ije/dyx034.CrossRefGoogle Scholar
Zhao, SS and Gill, D (2022) Genetically proxied IL-6 receptor inhibition and risk of polymyalgia rheumatica. Annals of the Rheumatic Diseases, 88(10), 14801482. https://doi.org/10.1136/annrheumdis-2022-222578.CrossRefGoogle Scholar
Zheng, J, Haberland, V, Baird, D, Walker, V, Haycock, PC, Hurle, MR, Gutteridge, A, Erola, P, Liu, Y, Luo, S, Robinson, J, Richardson, TG, Staley, JR, Elsworth, B, Burgess, S, Sun, BB, Danesh, J, Runz, H, Maranville, JC, Martin, HM, Yarmolinsky, J, Laurin, C, Holmes, MV, Liu, JZ, Estrada, K, Santos, R, McCarthy, L, Waterworth, D, Nelson, MR, Smith, GD, Butterworth, AS, Hemani, G, Scott, RA, and Gaunt, TR (2020) Phenome-wide Mendelian randomization mapping the influence of the plasma proteome on complex diseases. Nature Genetics 52(10), 11221131. https://doi.org/10.1038/s41588-020-0682-6.CrossRefGoogle ScholarPubMed
Zuber, V, Grinberg, NF, Gill, D, Manipur, I, Slob, EAW, Patel, A, Wallace, C and Burgess, S (2022) Combining evidence from Mendelian randomization and colocalization: Review and comparison of approaches. American Journal of Human Genetics 109(5), 767782. https://doi.org/10.1016/j.ajhg.2022.04.001.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Structure and assumptions of Mendelian randomization analyses (A) and application to the example of genetically proxied inhibition of interleukin 6 (IL-6) signaling (B).

Figure 1

Figure 2. Applications of Mendelian randomization (MR) to each phase of drug development. PheWAS, phenome-wide association study.

Figure 2

Table 1. Selected examples of MR analyses cited in this Review that investigated effects of genetically proxied inhibition of IL-6 signaling on a diverse range of outcomes

Figure 3

Figure 3. Different approaches to weighting the effect of a genetic variant on a drug target. The absence of an arrow between the ‘molecular biomarkers’ and ‘clinical risk factors and outcomes’ box demonstrates that a molecular biomarker does not necessarily have to be causal for a disease outcome.

Author comment: Mendelian randomization as a tool to inform drug development using human genetics — R0/PR1

Comments

No accompanying comment.

Review: Mendelian randomization as a tool to inform drug development using human genetics — R0/PR2

Comments

Comments to Author: This article gives and overview how Mendelian Randomization (MR) can be utilized to inform the different stages of drug development. Throughout the review the authors illustrate how MR applications using genetic inhibition of interleukin-6 as an exemplar.

1. The main MR examples in the manuscript use interleukin-6 (IL6) but in the preamble PCSK9 is used as an example – what was the rationale for this? Is the information on PCSK9 needed?

2. How were the published genetic studies for IL-6 that are included in Table 1 selected? Was this a done systematically?

3. On line 185 the authors state that “Additional methods for linking genetic variant to causal gene, and the respective strengths and weaknesses of these methods, have been discussed at length elsewhere”. It would be helpful to include a brief description of the methods or at least summarize the approaches in a table.

4. On lines 248- 250 the authors state that “If multiple variants are used, they should be independently inherited, or methods designed for the use of correlated genetic variants should be employed.” Although a reference is given it would be helpful to provide a brief summary of the methods should be provided as the target audience for this article will be non-experts.

5. More details on how lifespan can be used to demonstrate net benefits is needed. An example of the findings in relation to IL6 should be given rather than just citing a reference with minimal detail.

6. I would have liked to see included in the review with recommendations on how the representation of different ancestries can be improved – not just that these studies are lacking and so generalizability is limited to Europeans only.

Review: Mendelian randomization as a tool to inform drug development using human genetics — R0/PR3

Conflict of interest statement

Reviewer declares none.

Comments

Comments to Author: The review concisely explains the concept of Mendelian randomization (MR) and places it into a drug discovery context with a series of exemplars focusing on IL6 signalling.

The review lays out some of the decision making process during the design of MR experiments, particularly in the selection of the genetic instrument. For example the variants should generally by cis to the gene, and ideally in the coding region so that the causal link is strongly established.

The review considers in detail some of the key elements that are needed to plan a successful MR study, including a clearly defined modifiable exposure of interest, the outcome of interest, the genetic instrument, and the statistical analysis required to detect a true signal.

Overall this is an excellent review and I agree with the authors that growing international population genetic resources will certainly increase the use of MR so that it becomes a pivotal tool in most drug development efforts.

Minor comments that might be addressed by the authors:

1) One area I would like to have seen addressed a little more thoroughly, was a comparison of MR with conventional genetic association methodology. I understand that MR is considered a more powerful method for studying the causal effect of an exposure on an outcome because it can provide a more unbiased estimate of the causal effect and is less prone to confounding compared to observational studies, but it would be good to contrast this to more general use of genetic association. In short how do an MR finding and a genetic association at the same locus differ?

2) Another area for discussion is the suitability of a locus for MR? Is it likely that all genes could potentially be investigated using MR? What are the likely key limitations and would it be possible to estimate roughly how many genes might be investigated? Which disease traits are particularly suited to MR and conversely are there disease traits that are unsuited to a MR approach?

Recommendation: Mendelian randomization as a tool to inform drug development using human genetics — R0/PR4

Comments

Comments to Author: As you will see from their comments, the reviewers were positive about your review but raise a few minor questions and suggestions that will hopefully improve the final paper when addressed.

Decision: Mendelian randomization as a tool to inform drug development using human genetics — R0/PR5

Comments

No accompanying comment.

Author comment: Mendelian randomization as a tool to inform drug development using human genetics — R1/PR6

Comments

January 8th, 2023

Dr. Damian Smedley

Editor, Cambridge Prisms: Precision Medicine

Dear Dr. Smedley,

We would like to express our gratitude to the Reviewers for their insightful and constructive comments. We have carefully revised the manuscript as detailed below, and now find it much improved. We look forward to hearing from you.

Sincerely,

Iyas Daghlas, MD

University of California, San Francisco

Eighth Floor, 400 Parnassus Ave, San Francisco, CA 94143

Telephone: +1-573-823-3483

Email: iyas.daghlas@ucsf.edu

Review: Mendelian randomization as a tool to inform drug development using human genetics — R1/PR7

Comments

Comments to Author: This is an excellent review that serves as an informative introduction to the use of MR. I will not reiterate my previous comments, but note that my comments about the need for comparison and contrast between MR and GWAS findings are fully addressed.

Review: Mendelian randomization as a tool to inform drug development using human genetics — R1/PR8

Comments

Comments to Author: I have no further comments

Recommendation: Mendelian randomization as a tool to inform drug development using human genetics — R1/PR9

Comments

No accompanying comment.

Decision: Mendelian randomization as a tool to inform drug development using human genetics — R1/PR10

Comments

No accompanying comment.