Skip to main content
The last 10 years of research in stroke genetics have seen rapid progress in our understanding of the genetic architecture of stroke. The first genome-wide association study (GWAS) of stroke was published barely 17 years ago,1 and in the last 10 years alone, no less than 17 additional GWAS have been reported.2–4 Coupled with advances in multiomics investigations and statistical genetics methodologies, the potential for clinical applications is finally emerging. This article will focus on: (1) a brief summary of the main discoveries in stroke genetics, as well as methodological developments that enabled these in the past 10 years; (2) anticipated perspectives for the next 10 years for expanding our understanding of stroke genomics and deriving clinical applications (Figure).
Figure. Overview of discoveries and perspectives for future developments in stroke genomics. Summary of the number of independent genetic risk loci identified in genome-wide association study (GWAS) of main stroke types and related phenotypes (A) and progress and perspectives in stroke genomics (B). A, Numbers represent the numbers of independent genetic risk loci, based on the latest largest single-trait GWAS, described in the article (for intracerebral hemorrhage (ICH) a larger candidate gene study confirming a genome-wide significant association of APOE (Apolipoprotein E) epsilon 2 and 4 alleles with lobar hemorrhage is also accounted for). NIHSS indicates National Institutes of Health Stroke Scale; and mRS, modified Rankin Scale. *Worldwide distribution of stroke types according to Global Burden of Disease Study.5 †This category is too broad to provide exact numbers but usually less than a handful per condition if any (eg, cervical artery dissection, moyamoya disease, early onset, etc).

ADVANCES IN STROKE GENETICS AND GENOMICS IN THE PAST 10 YEARS

Over the past decade, major technological breakthroughs, methodological innovations, and large international consortia pioneering data sharing in an open science spirit have enabled major discoveries in genomics of stroke and complex diseases at large.

Complex Stroke Genomics

Increasingly large cross-ancestry GWAS meta-analyses have discovered over 100 independent genetic risk loci harboring common single nucleotide variants associated with stroke risk. The largest stroke GWAS to date comprised 110 000 patients with stroke, with additional 90 000 patients with stroke to follow-up significant findings. Most associations were observed for any stroke and ischemic stroke,3,4 with so far few genome-wide significant associations for intracerebral hemorrhage, due to a smaller sample size.6,7 Extensive efforts have been made to identify genetic associations for individual ischemic stroke subtypes,4,8 but numbers remain considerably smaller (<15 000 for small vessel, large artery, or cardioembolic stroke). Moreover, thanks to worldwide international collaborations, the contribution of non-European data sets to stroke GWAS has considerably increased in recent years, representing about a third of patients (most of whom are of East-Asian ancestry). This has enabled a notable gain in power and made it possible to compare risk variants and effect sizes of genetic associations across ancestries. Seventeen genetic risk loci have been identified for intracranial aneurysms, the leading cause of subarachnoid hemorrhage.9 In parallel, over 60 loci were found to be associated with magnetic resonance imaging-based endophenotypes of stroke, especially magnetic resonance imaging markers of cerebral small vessel disease.10,11 Recent efforts also revealed genetic variants modulating acute12 or long-term outcomes after stroke.13 Although substantial shared genetic variation with vascular risk factors (particularly blood pressure) was observed, many stroke risk loci point to novel pathways.4
Whole exome sequencing14 and whole genome sequencing15,16 studies of stroke, enabling the detection of rare stroke risk variants, respectively, within genes and at the much broader genome-wide level (enabling to capture variants that regulate gene expression), are still scarce and underpowered. These will likely provide important novel insight once larger samples become available, especially if rare deleterious or protective variants with large effect sizes can be identified. A few studies also explored other types of genetic variants, such as structural variants (copy number variants, mitochondrial DNA copy number, etc)17,18 in relation with stroke, but these remain largely unexplored. Increasingly scalable long-read sequencing technologies will open new perspectives for deciphering the impact of such variants with greater accuracy.19

METHODOLOGICAL DEVELOPMENTS IN GENOMICS OF COMPLEX DISEASES

High-quality standards for GWAS, increasingly large resources and systematic sharing of GWAS summary statistics have considerably enhanced the yield of genomic research. Powerful methods have been developed that leverage GWAS data to explore shared genetic variation across traits, establish causal inferences, and enable increasingly informative risk prediction. Two-sample Mendelian randomization, which consists of using genetic risk variants as instruments for a given exposure or to mimic drug effects, is a powerful tool to seek evidence for causal relationships and rule out reverse causation.11 Mendelian randomization provided for instance evidence for subtype specificity of stroke risk factors (eg, association of genetic liability to thrombosis with all ischemic stroke subtypes except small vessel stroke),4 or for a stronger protective effect of β-blockers than calcium channel blockers on risk of cervical artery dissection,20 leading cause of stroke in young adults. Polygenic scores (PGS), which summarize the effects of many genetic risk variants, were shown to predict stroke4,21 and other cardiovascular diseases.22 Combining stroke PGS across ancestries improved predictive performances and transportability to non-European populations.4

Genetics of Rare Causes of Stroke

Developments in next-generation sequencing technologies have also enabled the study of rare genetic variants underpinning Mendelian (monogenic) strokes on a scale never possible before, yielding important insights. First, rare genetic variants hitherto believed to cause Mendelian strokes in a quasi-deterministic fashion are in fact much more common than anticipated on a population scale.23 A study of 5 monogenic cerebral small vessel disease genes in 200 000 persons reported a prevalence of putative pathogenic variants of 1:200.24 Across 18 genes causing Mendelian stroke the aggregate prevalence of pathogenic variants varied between 0.7% and 2.8% depending on genetic ancestry,25 suggesting variable penetrance and the need for additional factors (including common genetic variants) for strokes to manifest clinically. The identification of genes (eg, COL4A1/2, HTRA1) harboring both common variants associated with complex stroke and rare mutations causing monogenic stroke further adds to the blurring distinction between monogenic and multifactorial stroke.3,11 Another major discovery is the recognition that somatic mutations can lead to vascular malformations causing stroke, such as arterio-venous malformations,26 paving the way for novel therapeutics.27

PERSPECTIVES IN STROKE GENOMICS AND PRECISION MEDICINE OVER THE NEXT 10 YEARS

Despite major advances, many opportunities and challenges remain. The nature of genetic associations, whereby thousands of variants with individually weak effects contribute to disease risk, makes it imperative to achieve large sample sizes. This is also true for rare genetic variants with large effect sizes. The number of identified loci is directly proportional to the discovery sample size, with no apparent plateau reached so far. Fortunately, rapid development in sequencing technology makes whole genome sequencing increasingly affordable, with predictions that the aspirational goal of the $100 genome will be reached soon.28 Compounding the need for large sample size is the need for increased diversity in genetic studies. Major efforts were undertaken to maximize diversity in stroke genomics; however, there remain large gaps. Diversity is important for generalizability and equity of genetic risk prediction and genomics-driven drug discovery and also a major opportunity for enhancing genetic discoveries and fine-mapping genetic risk loci. There is also a major need for larger genomic studies of ischemic stroke subtypes, to decipher more specific molecular and cellular mechanisms, and of stroke-related phenotypes such as stroke recovery, to help unravel neuroprotective pathways.

From Genomics to Multiomics

Translating genetic loci to therapies requires precise knowledge of both causal genes and cell-specific directionality of expression effects. Combining genomics with other omics technologies (epigenomics, transcriptomics, proteomics, etc) that are increasingly affordable creates an opportunity to make rapid progress toward defining molecular mechanisms and causal pathways underlying genetic associations.29 Single-cell transcriptomics and its spatial profiling can enhance this potential by enabling mechanisms to be mapped to specific cell (sub)types, and have for instance shown enrichment of genetic risk loci for perivascular space burden, an imaging marker of cerebral small vessel disease, in brain vascular endothelial cells based on a human single-cell atlas of fetal gene expression.10 Recent work also revealed that many genes differentially expressed in vascular brain cells of Alzheimer disease patients are directly linked to Alzheimer disease-associated GWAS loci, highlighting the strong interconnection of vascular and neurodegenerative processes.30 Various bioinformatics approaches facilitate the integration of genomic with other omic data (transcriptome- and proteome-wide association studies, colocalization analyses, Mendelian randomization, etc), leveraging increasingly rich resources of tissue- and cell-specific multiomics.4,29 These are beginning to yield major results for a variety of complex diseases, such as a cerebrospinal fluid-based proteogenomic study that identified 42 putative causal proteins for Alzheimer disease, a third of which point to drug repositioning opportunities, also shedding new light on how independent Alzheimer disease GWAS loci are connected to each other.31 Artificial intelligence-based approaches are also growingly proposed for multidimensional integration of omics,32 although this field is still at a preliminary stage for brain diseases, with a growing interest in expanding that integration further to the combination of omics and imaging dimensions, with even greater complexity, as outlined for the Alzheimer precision medicine initiative.33

Toward Precision Medicine and Prevention

As sequencing becomes more affordable and rare mutations causing Mendelian forms of stroke are more common than previously suspected, genetic testing may eventually be more frequently included in the clinical work-up of patients with stroke. However, the added value of systematic mutation screening in sporadic patients with stroke is still unknown and requires additional research. PGS also have the potential to improve disease prevention,34 and to enable predictive and prognostic enrichment of clinical trials, by selecting individuals who are most likely to develop the disease and respond to treatment.35 The clinical utility of PGS in the specific setting of stroke prevention remains to be determined, for example, to discriminate individuals at high risk of ischemic versus hemorrhagic stroke to guide antithrombotic drug prescription in situations of clinical equipoise. Implementing PGS will require addressing ethical and societal challenges and gaining acceptance by physicians.36 Whether PGS will ultimately have broad applications in the clinic,37 or whether their use will mostly remain restricted to a research setting is still unclear. Recent pharmacogenomic studies also show that clinically relevant adverse drug reactions can be significantly reduced by testing for genetic variants modifying drug metabolism, demonstrating feasibility across diverse health care systems.38 However, the implementation of pharmacogenetic testing in clinical practice is slow, due to some conflicting findings, a lack of stroke-specific comparative effectiveness trials, and practical aspects including cost-effectiveness. As an example, despite encouraging results supporting the use of pharmacogenetics to guide antiplatelet (clopidogrel) and anticoagulant (warfarin) treatment, clinical implementation remains sporadic.39 Overall, such developments will require genomics savvy health care workforce and public health specialists,40 with an urgent need for curricula in precision medicine and public health.

Genomics-Driven Drug Discovery

Providing genetic evidence for drug effects was retrospectively estimated to increase the success rate of clinical trials by >2-fold.41 Currently available genetic evidence supported 63% of drugs approved by the Food and Drug Administration in the past decade.42 However, genetics drove drug discovery only in a small fraction of these cases.43 Several lipid-lowering therapies derived from genetic findings, such as PCSK9 (proprotein convertase subtilisin/kexin type 9) and ANGPTL3 (angiopoietin like 3) inhibitors and gene-editing/-silencing therapies in advanced development are of direct relevance for stroke prevention.43 The trajectory from discovery of genetic associations to drug development is long (25 years on average) and complex. Robust evidence for causal genes and downstream effectors is required. Gene-editing/-silencing experiments in cellular and animal models are typically used, but remain challenging.44 Resource-intensive complementary approaches such as massively parallel reporter assays or Clustered Regularly Interspaced Short Palindromic Repeats-mediated gene-editing of induced pluripotent stem cell lines (including in 3-dimensional models such as organoids), may eventually play a crucial role in larger scale screening of putative causal variants and genes at genetic risk loci for stroke and related disorders, as was recently successfully implemented for autism spectrum disorder.45 In combination with drug databases, GWAS can also provide guidance for repositioning of drugs for stroke that are already commercialized or tested in clinical trials for other indications. Such evidence was recently shown for F11 inhibitors and PROC (protein C) agonists, currently in phase II/III trials for stroke, and drugs targeting LAMC1 (laminin subunit gamma 1) and GP1BA (glycoprotein Ib platelet subunit alpha).4 Finally, the convergence of genomic breakthroughs with the advent of next-generation programable therapies (RNA based and other) will likely accelerate genomics-driven drug discovery.
The past 10 years have been transformative to the field of stroke genetics, yet the full promise of precision medicine has not reached clinical practices. Translation of genetic findings into improved care is arguably the most important challenge for the next 10 years. Many of the genetic associations already identified likely hold the key to new avenues for prevention and treatment of stroke, yet the investment necessary to translate these findings is considerable. As rapid progress continues to be made in stroke genetics, the urgency of translational opportunities only becomes more pressing.

REFERENCES

1.
Kubo M, Hata J, Ninomiya T, Matsuda K, Yonemoto K, Nakano T, Matsushita T, Yamazaki K, Ohnishi Y, Saito S, et al. A nonsynonymous SNP in PRKCH (protein kinase C eta) increases the risk of cerebral infarction. Nat Genet. 2007;39:212–217. doi: 10.1038/ng1945
2.
Surakka I, Wu KH, Hornsby W, Wolford BN, Shen F, Zhou W, Huffman JE, Pandit A, Hu Y, Brumpton B, et al; TOPMed Stroke Working Group. Multi-ancestry meta-analysis identifies 5 novel loci for ischemic stroke and reveals heterogeneity of effects between sexes and ancestries. Cell genomics. 2023;3:100345. doi: 10.1016/j.xgen.2023.100345
3.
Debette S, Markus HS. Stroke genetics: discovery, insight into mechanisms, and clinical perspectives. Circ Res. 2022;130:1095–1111. doi: 10.1161/CIRCRESAHA.122.319950
4.
Mishra A, Malik R, Hachiya T, Jürgenson T, Namba S, Posner DC, Kamanu FK, Koido M, Le Grand Q, Shi M, et al; COMPASS Consortium. Stroke genetics informs drug discovery and risk prediction across ancestries. Nature. 2022;611:115–123. doi: 10.1038/s41586-022-05165-3
5.
Krishnamurthi RV, Ikeda T, Feigin VL. Global, regional and country-specific burden of ischaemic stroke, intracerebral haemorrhage and subarachnoid haemorrhage: a systematic analysis of the global burden of disease study 2017. Neuroepidemiology. 2020;54:171–179. doi: 10.1159/000506396
6.
Biffi A, Sonni A, Anderson CD, Kissela B, Jagiella JM, Schmidt H, Jimenez-Conde J, Hansen BM, Fernandez-Cadenas I, Cortellini L, et al; International Stroke Genetics Consortium. Variants at APOE influence risk of deep and lobar intracerebral hemorrhage. Ann Neurol. 2010;68:934–943. doi: 10.1002/ana.22134
7.
Woo D, Falcone GJ, Devan WJ, Brown WM, Biffi A, Howard TD, Anderson CD, Brouwers HB, Valant V, Battey TWK, et al; International Stroke Genetics Consortium. Meta-analysis of genome-wide association studies identifies 1q22 as a susceptibility locus for intracerebral hemorrhage. Am J Hum Genet. 2014;94:511–521. doi: 10.1016/j.ajhg.2014.02.012
8.
Traylor M, Persyn E, Tomppo L, Klasson S, Abedi V, Bakker MK, Torres N, Li L, Bell S, Rutten-Jacobs L, et al; Helsinki Stroke, Study Dutch Parelsnoer Institute-Cerebrovascular Accident (CVA) Study Group. Genetic basis of lacunar stroke: a pooled analysis of individual patient data and genome-wide association studies. Lancet Neurol. 2021;20:351–361. doi: 10.1016/S1474-4422(21)00031-4
9.
Bakker MK, van der Spek RAA, van Rheenen W, Morel S, Bourcier R, Hostettler IC, Alg VS, van Eijk KR, Koido M, Akiyama M, et al; HUNT All-In Stroke. Genome-wide association study of intracranial aneurysms identifies 17 risk loci and genetic overlap with clinical risk factors. Nat Genet. 2020;52:1303–1313. doi: 10.1038/s41588-020-00725-7
10.
Duperron MG, Knol MJ, Le Grand Q, Evans TE, Mishra A, Tsuchida A, Roshchupkin G, Konuma T, Trégouët DA, Romero JR, et al; CHARGE Consortium. Genomics of perivascular space burden unravels early mechanisms of cerebral small vessel disease. Nat Med. 2023;29:950–962. doi: 10.1038/s41591-023-02268-w
11.
Bordes C, Sargurupremraj M, Mishra A, Debette S. Genetics of common cerebral small vessel disease. Nat Rev Neurol. 2022;18:84–101. doi: 10.1038/s41582-021-00592-8
12.
Ibanez L, Heitsch L, Carrera C, Farias FHG, Del Aguila JL, Dhar R, Budde J, Bergmann K, Bradley J, Harari O, et al. Multi-ancestry gwas reveals excitotoxicity associated with outcome after ischaemic stroke. Brain. 2022;145:2394–2406. doi: 10.1093/brain/awac080
13.
Söderholm M, Pedersen A, Lorentzen E, Stanne TM, Bevan S, Olsson M, Cole JW, Fernandez-Cadenas I, Hankey GJ, Jimenez-Conde J, et al; International Stroke Genetics Consortium, the NINDS-SiGN Consortium, and the Genetics of Ischaemic Stroke Functional Outcome (GISCOME) Network. Genome-wide association meta-analysis of functional outcome after ischemic stroke. Neurology. 2019;92:e1271–e1283. doi: 10.1212/WNL.0000000000007138
14.
Xie Y, Acosta JN, Ye Y, Demarais ZS, Conlon CJ, Chen M, Zhao H, Falcone GJ. Whole-exome sequencing analyses support a role of vitamin d metabolism in ischemic stroke. Stroke. 2023;54:800–809. doi: 10.1161/STROKEAHA.122.040883
15.
Hu Y, Haessler JW, Manansala R, Wiggins KL, Moscati A, Beiser A, Heard-Costa NL, Sarnowski C, Raffield LM, Chung J, et al; Trans-Omics for Precision Medicine (TOPMed) Stroke Working Group, the NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium. Whole-genome sequencing association analyses of stroke and its subtypes in ancestrally diverse populations from trans-omics for precision medicine project. Stroke. 2022;53:875–885. doi: 10.1161/STROKEAHA.120.031792
16.
Cheng S, Xu Z, Bian S, Chen X, Shi Y, Li Y, Duan Y, Liu Y, Lin J, Jiang Y, et al. The STROMICS genome study: deep whole-genome sequencing and analysis of 10k Chinese patients with ischemic stroke reveal complex genetic and phenotypic interplay. Cell Discovery. 2023;9:75. doi: 10.1038/s41421-023-00582-8
17.
Chong MR, Narula S, Morton R, Judge C, Akhabir L, Cawte N, Pathan N, Lali R, Mohammadi-Shemirani P, Shoamanesh A, et al. Mitochondrial DNA copy number as a marker and mediator of stroke prognosis: observational and Mendelian randomization analyses. Neurology. 2022;98:e470–e482. doi: 10.1212/WNL.0000000000013165
18.
Pfeiffer D, Chen B, Schlicht K, Ginsbach P, Abboud S, Bersano A, Bevan S, Brandt T, Caso V, Debette S, et al; CADISP; GISCOME; SiGN Studies; and ISGC. Genetic imbalance is associated with functional outcome after ischemic stroke. Stroke. 2019;50:298–304. doi: 10.1161/STROKEAHA.118.021856
19.
Liao WW, Asri M, Ebler J, Doerr D, Haukness M, Hickey G, Lu S, Lucas JK, Monlong J, Abel HJ, et al. A draft human pangenome reference. Nature. 2023;617:312–324. doi: 10.1038/s41586-023-05896-x
20.
Le Grand Q, Ecker Ferreira L, Metso TM, Schilling S, Tatlisumak T, Grond-Ginsbach C, Engelter ST, Lyrer P, Majersik JJ, Worrall BB, et al; CADISP Consortium. Genetic insights on the relation of vascular risk factors and cervical artery dissection. J Am Coll Cardiol. 2023;82:1411–1423. doi: 10.1016/j.jacc.2023.07.021
21.
Abraham G, Malik R, Yonova-Doing E, Salim A, Wang T, Danesh J, Butterworth AS, Howson JMM, Inouye M, Dichgans M. Genomic risk score offers predictive performance comparable to clinical risk factors for ischaemic stroke. Nat Commun. 2019;10:5819. doi: 10.1038/s41467-019-13848-1
22.
Khera AV, Chaffin M, Aragam KG, Haas ME, Roselli C, Choi SH, Natarajan P, Lander ES, Lubitz SA, Ellinor PT, et al. Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nat Genet. 2018;50:1219–1224. doi: 10.1038/s41588-018-0183-z
23.
Rutten JW, Dauwerse HG, Gravesteijn G, van Belzen MJ, van der Grond J, Polke JM, Bernal-Quiros M, Lesnik Oberstein SA. Archetypal notch3 mutations frequent in public exome: implications for cadasil. Ann Clin Transl Neurol. 2016;3:844–853. doi: 10.1002/acn3.344
24.
Ferguson AC, Thrippleton S, Henshall D, Whittaker E, Conway B, MacLeod M, Malik R, Rawlik K, Tenesa A, Sudlow C, et al. Frequency and phenotype associations of rare variants in 5 monogenic cerebral small vessel disease genes in 200,000 UK Biobank participants. Neurol Genet. 2022;8:e200015. doi: 10.1212/NXG.0000000000200015
25.
Grami N, Chong M, Lali R, Mohammadi-Shemirani P, Henshall DE, Rannikmäe K, Paré G. Global assessment of Mendelian stroke genetic prevalence in 101 635 individuals from 7 ethnic groups. Stroke. 2020;51:1290–1293. doi: 10.1161/STROKEAHA.119.028840
26.
Nikolaev SI, Vetiska S, Bonilla X, Boudreau E, Jauhiainen S, Rezai Jahromi B, Khyzha N, DiStefano PV, Suutarinen S, Kiehl TR, et al. Somatic activating kras mutations in arteriovenous malformations of the brain. N Engl J Med. 2018;378:250–261. doi: 10.1056/NEJMoa1709449
27.
Nguyen HL, Boon LM, Vikkula M. Trametinib as a promising therapeutic option in alleviating vascular defects in an endothelial kras-induced mouse model. Hum Mol Genet. 2023;32:276–289. doi: 10.1093/hmg/ddac169
28.
Singh S. The hundred-dollar genome: a health care cart before the genomic horse. CMAJ. 2018;190:E514. doi: 10.1503/cmaj.69259
29.
Montaner J, Ramiro L, Simats A, Tiedt S, Makris K, Jickling GC, Debette S, Sanchez JC, Bustamante A. Multilevel omics for the discovery of biomarkers and therapeutic targets for stroke. Nat Rev Neurol. 2020;16:247–264. doi: 10.1038/s41582-020-0350-6
30.
Sun N, Akay LA, Murdock MH, Park Y, Galiana-Melendez F, Bubnys A, Galani K, Mathys H, Jiang X, Ng AP, et al. Single-nucleus multiregion transcriptomic analysis of brain vasculature in alzheimer’s disease. Nat Neurosci. 2023;26:970–982. doi: 10.1038/s41593-023-01334-3
31.
Cruchaga C, Western D, Timsina J, Wang L, Wang C, Yang C, Ali M, Beric A, Gorijala P, Kohlfeld P, et al. Proteogenomic analysis of human cerebrospinal fluid identifies neurologically relevant regulation and informs causal proteins for alzheimer’s disease. Research Square. 2023;rs.3.rs-2814616. doi: 10.21203/rs.3.rs-2814616/v1
32.
Reel PS, Reel S, Pearson E, Trucco E, Jefferson E. Using machine learning approaches for multi-omics data analysis: a review. Biotechnol Adv. 2021;49:107739. doi: 10.1016/j.biotechadv.2021.107739
33.
Hampel H, Nisticò R, Seyfried NT, Levey AI, Modeste E, Lemercier P, Baldacci F, Toschi N, Garaci F, Perry G, et al; Alzheimer Precision Medicine Initiative (APMI). Omics sciences for systems biology in alzheimer’s disease: state-of-the-art of the evidence. Ageing Res Rev. 2021;69:101346. doi: 10.1016/j.arr.2021.101346
34.
O’Sullivan JW, Raghavan S, Marquez-Luna C, Luzum JA, Damrauer SM, Ashley EA, O’Donnell CJ, Willer CJ, Natarajan P; American Heart Association Council on Genomic and Precision Medicine; Council on Clinical Cardiology; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Radiology and Intervention; Council on Lifestyle and Cardiometabolic Health; and Council on Peripheral Vascular Disease. Polygenic risk scores for cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2022;146:e93–e118. doi: 10.1161/CIR.0000000000001077
35.
Fahed AC, Philippakis AA, Khera AV. The potential of polygenic scores to improve cost and efficiency of clinical trials. Nat Commun. 2022;13:2922. doi: 10.1038/s41467-022-30675-z
36.
Polygenic Risk Score Task Force of the International Common Disease Alliance. Responsible use of polygenic risk scores in the clinic: potential benefits, risks and gaps. Nat Med. 2021;27:1876–1884. doi: 10.1038/s41591-021-01549-6
37.
Hao L, Kraft P, Berriz GF, Hynes ED, Koch C, Korategere VKP, Parpattedar SS, Steeves M, Yu W, Antwi AA, et al. Development of a clinical polygenic risk score assay and reporting workflow. Nat Med. 2022;28:1006–1013. doi: 10.1038/s41591-022-01767-6
38.
Swen JJ, van der Wouden CH, Manson LE, Abdullah-Koolmees H, Blagec K, Blagus T, Böhringer S, Cambon-Thomsen A, Cecchin E, Cheung KC, et al; Ubiquitous Pharmacogenomics Consortium. A 12-gene pharmacogenetic panel to prevent adverse drug reactions: an open-label, multicentre, controlled, cluster-randomised crossover implementation study. Lancet. 2023;401:347–356. doi: 10.1016/S0140-6736(22)01841-4
39.
Ross S, Krebs K, Paré G, Milani L. Pharmacogenomics in stroke and cardiovascular disease: state of the art. Stroke. 2023;54:270–278. doi: 10.1161/STROKEAHA.122.037717
40.
We need a genomics-savvy healthcare workforce. Nat Med. 2023;29:1877–1878. doi: 10.1038/s41591-023-02522-1
41.
King EA, Davis JW, Degner JF. 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 Genet. 2019;15:e1008489. doi: 10.1371/journal.pgen.1008489
42.
Rusina PV, Falaguera MJ, Romero JMR, McDonagh EM, Dunham I, Ochoa D. Genetic support for FDA-approved drugs over the past decade. Nat Rev Drug Discov. 2023;22:864. doi: 10.1038/d41573-023-00158-x
43.
Trajanoska K, Bhérer C, Taliun D, Zhou S, Richards JB, Mooser V. From target discovery to clinical drug development with human genetics. Nature. 2023;620:737–745. doi: 10.1038/s41586-023-06388-8
44.
Gupta RM. Causal gene confusion: the complicated edn1/phactr1 locus for coronary artery disease. Arterioscler Thromb Vasc Biol. 2022;42:610–612. doi: 10.1161/ATVBAHA.122.317539
45.
Li C, Fleck JS, Martins-Costa C, Burkard TR, Themann J, Stuempflen M, Peer AM, Vertesy A, Littleboy JB, Esk C, et al. Single-cell brain organoid screening identifies developmental defects in autism. Nature. 2023;621:373–380. doi: 10.1038/s41586-023-06473-y

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Stroke
Stroke
Pages: 2163 - 2168
PubMed: 38511336

Versions

You are viewing the most recent version of this article.

History

Published online: 21 March 2024
Published in print: August 2024

Permissions

Request permissions for this article.

Keywords

  1. genetics
  2. genomics
  3. multiomics
  4. precision medicine
  5. stroke

Subjects

Authors

Affiliations

University of Bordeaux, INSERM, Bordeaux Population Health, France (S.D.).
Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux University Hospital, France (S.D.).
Guillaume Paré, MD, MSc https://orcid.org/0000-0002-6795-4760
Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada (G.P.).

Notes

The American Heart Association celebrates its 100th anniversary in 2024. This article is part of a series across the entire AHA Journal portfolio written by international thought leaders on the past, present, and future of cardiovascular and cerebrovascular research and care. To explore the full Centennial Collection, visit https://www.ahajournals.org/centennial.
The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association.
For Sources of Funding and Disclosures, see pages 2166–2167.
Correspondence to: Stéphanie Debette, MD, PhD, University of Bordeaux, INSERM, Bordeaux Population Health, UM1219, 146, rue Léo Saignat, F-33000 Bordeaux, France. Email [email protected]

Disclosures

Dr Paré has received research funding from Bayer and honoraria from Amgen, Bayer, Illumina, Novartis and Sanofi. The other author reports no conflicts.

Sources of Funding

Dr Debette is supported by a grant overseen by the French National Research Agency (Agence National de la Recherche, ANR) as part of the Investment for the Future Programme ANR-18-RHUS-0002, by the Precision and Global Vascular Brain Health Institute, ANR-23-IAHU-0001, funded by the France 2030 investment plan (Instituts Hospitalo-Universitaires vague 3, initiative), and by the European Union’s Horizon 2020 research and innovation program under grant agreement 754517.

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this journal for 24 hours

Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Figures

Tables

Media

Share

Share

Share article link

Share

Comment Response