Hostname: page-component-76fb5796d-45l2p Total loading time: 0 Render date: 2024-04-29T05:57:46.886Z Has data issue: false hasContentIssue false

Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift?

Published online by Cambridge University Press:  20 November 2023

Saad Javed
Affiliation:
1National Heart and Lung Institute, Imperial College London, UK 2Cardiovascular Research Centre, Cardiovascular Magnetic Resonance Unit & Inherited Cardiac Conditions Care Group, Royal Brompton and Harefield Hospitals, Part of Guy’s and St Thomas’ NHS Foundation Trust, London, UK
Brian P. Halliday*
Affiliation:
1National Heart and Lung Institute, Imperial College London, UK 2Cardiovascular Research Centre, Cardiovascular Magnetic Resonance Unit & Inherited Cardiac Conditions Care Group, Royal Brompton and Harefield Hospitals, Part of Guy’s and St Thomas’ NHS Foundation Trust, London, UK
*
Corresponding author: Brian P. Halliday; Email: b.halliday@imperial.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Precision medicine for cardiomyopathies holds great promise to improve patient outcomes costs by shifting the focus to patient-specific treatment decisions, maximising the use of therapies most likely to lead to benefit and minimising unnecessary intervention. Dilated cardiomyopathy (DCM), characterised by left ventricular dilatation and impairment, is a major cause of heart failure globally. Advances in genomic medicine have increased our understanding of the genetic architecture of DCM. Understanding the functional implications of genetic variation to reveal genotype-specific disease mechanisms is the subject of intense investigation, with advanced cardiac imaging and mutliomics approaches playing important roles. This may lead to increasing use of novel, targeted therapy. Individualised treatment and risk stratification is however made more complex by the modifying effects of common genetic variation and acquired environmental factors that help explain the variable expressivity of rare genetic variants and gene elusive disease. The next frontier must be expanding work into early disease to understand the mechanisms that drive disease expression, so that the focus can be placed on disease prevention rather than management of later symptomatic disease. Overcoming these challenges holds the key to enabling a paradigm shift in care from the management of symptomatic heart failure to prevention of disease.

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

Advances in the understanding of the molecular mechanisms that cause dilated cardiomyopathy offer the opportunity to personalise care and improve the outcomes of patients with this heterogeneous family of disease. Comprehensive characterisation of the disease with genetic testing and advanced imaging will play a key role. Precision therapies that target the primary disease mechanism will offer new hope for disease prevention in genetically susceptible individuals at risk of developing highly penetrant, malignant forms of the condition as well as effective treatments of early asymptomatic disease.

Introduction

Heart failure is a looming global health crisis with a predicted lifetime risk of 25 to 45% that is rapidly reaching epidemic proportions (Huffman et al., Reference Huffman, Berry, Ning, Dyer, Garside, Cai, Daviglus and Lloyd-Jones2013; Benjamin et al., Reference Benjamin, Muntner, Alonso, Bittencourt, Callaway, Carson, Chamberlain, Chang, Cheng, Das, Delling, Djousse, Elkind, Ferguson, Fornage, Jordan, Khan, Kissela, Knutson, Kwan, Lackland, Lewis, Lichtman, Longenecker, Loop, Lutsey, Martin, Matsushita, Moran, Mussolino, O’Flaherty, Pandey, Perak, Rosamond, Roth, Sampson, Satou, Schroeder, Shah, Spartano, Stokes, Tirschwell, Tsao, Turakhia, VanWagner, Wilkins, Wong and Virani2019). Despite the already high risk of developing heart failure, current projections indicate that the prevalence of the condition will surge by 46%, and treatment expenditure will increase by a staggering 127% by 2030 (Heidenreich et al., Reference Heidenreich, Albert, Allen, Bluemke, Butler, Fonarow, Ikonomidis, Khavjou, Konstam, Maddox, Nichol, Pham, Pina and Trogdon2013; Huffman et al., Reference Huffman, Berry, Ning, Dyer, Garside, Cai, Daviglus and Lloyd-Jones2013). These sobering statistics call for a radical shift in our current approach to managing the disease.

Dilated cardiomyopathy (DCM) is a myocardial disorder characterised by left ventricular (LV) dilatation accompanied by systolic dysfunction, in the absence of abnormal loading conditions or coronary artery disease (Yancy et al., Reference Yancy, Jessup, Bozkurt, Butler, Casey, Drazner, Fonarow, Geraci, Horwich, Januzzi, Johnson, Kasper, Levy, Masoudi, McBride, McMurray, Mitchell, Peterson, Riegel, Sam, Stevenson, Tang, Tsai and Wilkoff2013; Pinto et al., Reference Pinto, Elliott, Arbustini, Adler, Anastasakis, Bohm, Duboc, Gimeno, de Groote, Imazio, Heymans, Klingel, Komajda, Limongelli, Linhart, Mogensen, Moon, Pieper, Seferovic, Schueler, Zamorano, Caforio and Charron2016; Heidenreich et al., Reference Heidenreich, Bozkurt, Aguilar, Allen, Byun, Colvin, Deswal, Drazner, Dunlay, Evers, Fang, Fedson, Fonarow, Hayek, Hernandez, Khazanie, Kittleson, Lee, Link, Milano, Nnacheta, Sandhu, Stevenson, Vardeny, Vest and Yancy2022). Its prevalence is around 1 in 220 people and it represents the leading indication for heart transplantation (Japp et al., Reference Japp, Gulati, Cook, Cowie and Prasad2016; Chambers et al., Reference Chambers, Cherikh, Goldfarb, Hayes, Kucheryavaya, Toll, Khush, Levvey, Meiser, Rossano, Stehlik and Lung2018). DCM arises from a range of genetic and acquired factors, often occurring simultaneously. There is a significant overlap between intrinsic and extrinsic causes (Figure 1). Conditions that were previously considered as separate aetiologies, such as peripartum cardiomyopathy, cardiomyopathy following anthracycline chemotherapy and alcohol-related cardiomyopathy have been shown to have similar genetic backgrounds (Ware et al., Reference Ware, Li, Mazaika, Yasso, DeSouza, Cappola, Tsai, Hilfiker-Kleiner, Kamiya, Mazzarotto, Cook, Halder, Prasad, Pisarcik, Hanley-Yanez, Alharethi, Damp, Hsich, Elkayam, Sheppard, Kealey, Alexis, Ramani, Safirstein, Boehmer, Pauly, Wittstein, Thohan, Zucker, Liu, Gorcsan, McNamara, Seidman, Seidman, Arany and Imac and Investigators2016; Ware et al., Reference Ware, Amor-Salamanca, Tayal, Govind, Serrano, Salazar-Mendiguchia, Garcia-Pinilla, Pascual-Figal, Nunez, Guzzo-Merello, Gonzalez-Vioque, Bardaji, Manito, Lopez-Garrido, Padron-Barthe, Edwards, Whiffin, Walsh, Buchan, Midwinter, Wilk, Prasad, Pantazis, Baski, O’Regan, Alonso-Pulpon, Cook, Lara-Pezzi, Barton and Garcia-Pavia2018; Garcia-Pavia et al., Reference Garcia-Pavia, Kim, Alejandra Restrepo-Cordoba, Lunde, Wakimoto, Smith, Toepfer, Getz, Gorham, Patel, Ito, Willcox, Arany, Li, Owens, Govind, Nunez, Mazaika, Bayes-Genis, Walsh, Finkelman, Lupon, Whiffin, Serrano, Midwinter, Wilk, Bardaji, Ingold, Buchan, Tayal, Pascual-Figal, de Marvao, Ahmad, Garcia-Pinilla, Pantazis, Dominguez, Baksi, O’Regan, Rosen, Prasad, Lara-Pezzi, Provencio, Lyon, Alonso-Pulpon, Cook, SR, PJR, Aplenc, Seidman, Ky, Ware and Seidman2019). It is therefore perhaps best to consider DCM as a family of related disease that require comprehensive geno- and phenotyping to fully understand (Yancy et al., Reference Yancy, Jessup, Bozkurt, Butler, Casey, Drazner, Fonarow, Geraci, Horwich, Januzzi, Johnson, Kasper, Levy, Masoudi, McBride, McMurray, Mitchell, Peterson, Riegel, Sam, Stevenson, Tang, Tsai and Wilkoff2013; Japp et al., Reference Japp, Gulati, Cook, Cowie and Prasad2016; Pinto et al., Reference Pinto, Elliott, Arbustini, Adler, Anastasakis, Bohm, Duboc, Gimeno, de Groote, Imazio, Heymans, Klingel, Komajda, Limongelli, Linhart, Mogensen, Moon, Pieper, Seferovic, Schueler, Zamorano, Caforio and Charron2016).

Figure 1. Dilated cardiomyopathy (DCM) – a family of diseases. Selected syndromic causes of dilated cardiomyopathy include Barth syndrome, haemochromatosis, Kearns–Sayre syndrome, and Carvajal syndrome (adapted with permission from Halliday, Reference Halliday2022).

Present treatment strategies centre on the management of symptomatic heart failure using guideline-directed heart failure management (GDMT) – a combination of beta-blockers, angiotensin-converting enzyme inhibitors, mineralocorticoid antagonists and SGLT-2 inhibitors (Japp et al., Reference Japp, Gulati, Cook, Cowie and Prasad2016; McDonagh et al., Reference McDonagh, Metra, Adamo, Gardner, Baumbach, Bohm, Burri, Butler, Celutkiene, Chioncel, Cleland, Coats, Crespo-Leiro, Farmakis, Gilard, Heymans, Hoes, Jaarsma, Jankowska, Lainscak, Lam, Lyon, McMurray, Mebazaa, Mindham, Muneretto, Francesco Piepoli, Price, Rosano, Ruschitzka, Kathrine Skibelund and Group2021; Heidenreich et al., Reference Heidenreich, Bozkurt, Aguilar, Allen, Byun, Colvin, Deswal, Drazner, Dunlay, Evers, Fang, Fedson, Fonarow, Hayek, Hernandez, Khazanie, Kittleson, Lee, Link, Milano, Nnacheta, Sandhu, Stevenson, Vardeny, Vest and Yancy2022). This easily generalisable approach has dramatically improved the outcome of patients with heart failure reduced ejection fraction over the last 40 years (Vaduganathan et al., Reference Vaduganathan, Claggett, Jhund, Cunningham, Pedro Ferreira, Zannad, Packer, Fonarow, McMurray and Solomon2020). However, it places little focus on early treatment of asymptomatic myocardial dysfunction before the onset of heart failure and instead predominantly targets the neurohormonal consequences of the heart failure syndrome. The question of the “right time” to start these treatments is unclear and typically these agents are commenced when patients develop symptoms, late in the disease pathway. An increasing number of asymptomatic individuals with mild disease or genetically susceptible individuals are being identified through screening strategies. The incorporation of genetic information into DCM care offers opportunities for early precision intervention in individuals at risk before they develop symptoms (Figure 2). Whilst current GDMT will undoubtedly continue to form a mainstay of symptomatic heart failure, precision medicine offers a revolutionary solution that could not only offer additional targeted therapies for those with more advanced disease but perhaps, more importantly, offer targeted therapies to prevent and slow disease expression much earlier in the disease course. Currently, there is limited evidence focusing on the treatment of early DCM. This is likely to be related to low event rates, a long latent period in the development of overt disease and variability in the natural history of different gene mutations. However, with increasing numbers of individuals being identified at risk, it is important that streamlined clinical trials across large populations, using pragmatic end-points set out to address this important issue.

Figure 2. Precision therapies for genotype-positive, phenotype-negative (G+ P−) individuals would likely involve genotype-specific therapies, and lifestyle interventions. Treatments that could be introduced at an early disease stage include anti-fibrotic agents, and therapies to target cardiac metabolism (such as SGLT2 inhibitors) whereas advanced disease therapies include antiarrhythmics for those at the highest risk, ICD therapy and guideline-directed heart failure therapy (GDMT) (HF, heart failure; LVSD, left ventricular systolic dysfunction).

The fundamental premise is that a nuanced understanding of an individual’s disease through advanced cardiac imaging, genetics, and biomarkers enables a more personalised understanding of the mechanism and guides a more refined and targeted therapeutic approach. But is this all just a pipedream, or are we really on the brink of a paradigm shift in DCM care? In this article, we examine the present state and future promise of precision medicine in DCM to guide novel treatments using precision phenotyping of disease. We provide an overview of existing knowledge regarding the genetic origins of DCM and contemporary approaches to individualised management.

The genetic architecture of DCM

Using currently available next-generation sequencing panels, a causative rare genetic variant in about 20–30% of cases of DCM (Tayal et al., Reference Tayal, Ware, Lakdawala, Heymans and Prasad2021). The yield may be higher in select populations, such as those referred for advanced heart failure therapies, younger patients or those with a high burden of ventricular arrhythmia or conduction disease (Herman et al., Reference Herman, Lam, Taylor, Wang, Teekakirikul, Christodoulou, Conner, SR, McDonough, Sparks, Teodorescu, Cirino, Banner, Pennell, Graw, Merlo, Di Lenarda, Sinagra, Bos, Ackerman, Mitchell, Murry, Lakdawala, Ho, Barton, Cook, Mestroni, Seidman and Seidman2012; Lukas Laws et al., Reference Lukas Laws, Lancaster, Ben Shoemaker, Stevenson, Hung, Wells, Marshall Brinkley, Hughes, Anderson, Roden and Stevenson2022). Several different genes encoding a range of proteins with diverse functions have been implicated in DCM (Jordan et al., Reference Jordan, Peterson, Ai, Asatryan, Bronicki, Brown, Celeghin, Edwards, Fan, Ingles, James, Jarinova, Johnson, Judge, Lahrouchi, Lekanne Deprez, Lumbers, Mazzarotto, Medeiros Domingo, Miller, Morales, Murray, Peters, Pilichou, Protonotarios, Semsarian, Shah, Syrris, Thaxton, van Tintelen, Walsh, Wang, Ware and Hershberger2021). These are most commonly autosomal with X-linked and mitochondrial variants also uncommonly identified. Autosomal dominant inheritance is the norm and the most common genes implicated are those coding for sarcomeric proteins, including titin (TTN) and beta-myosin (MYH7). Other notable genes include those coding for cytoskeletal (FLNC), nuclear envelope (LMNA) and desmosomal (DSP) proteins. In some cases, the identification of a causative variant may influence treatment decisions, particularly those associated with a more malignant prognosis. However, the greatest value of genetic testing comes in predicting the risk of asymptomatic relatives by enabling the identification of those at high risk of developing the disease in the future. In gene elusive disease, all first-degree family relatives typically remain under clinical surveillance until the age of 60 years. If a causative genetic variant is identified, cascade screening will be able to identify the 50% of relatives who are carriers and who have an elevated risk of developing disease. Those who do not carry the variant can be discharged from follow-up.

A landmark study found rare truncating variants in the titin (TTNtv) gene in 25% of patients with advanced or familial DCM (Herman et al., Reference Herman, Lam, Taylor, Wang, Teekakirikul, Christodoulou, Conner, SR, McDonough, Sparks, Teodorescu, Cirino, Banner, Pennell, Graw, Merlo, Di Lenarda, Sinagra, Bos, Ackerman, Mitchell, Murry, Lakdawala, Ho, Barton, Cook, Mestroni, Seidman and Seidman2012). More recent studies found TTNtv in 13% of nonfamilial cases of DCM and ~ 0.5% of the general population (McNally and Mestroni, Reference McNally and Mestroni2017; Tayal and Prasad, Reference Tayal and Prasad2018; Schultheiss et al., Reference Schultheiss, Fairweather, Caforio, Escher, Hershberger, Lipshultz, Liu, Matsumori, Mazzanti, McMurray and Priori2019; Verdonschot et al., Reference Verdonschot, Hazebroek, Ware, Prasad and Heymans2019). These variants are associated with incomplete penetrance and variable expression (Japp et al., Reference Japp, Gulati, Cook, Cowie and Prasad2016) that may be attributed to additional gene-modifying factors, environmental factors, or the penetrance of disease later in life (Japp et al., Reference Japp, Gulati, Cook, Cowie and Prasad2016). Penetrance is likely to vary significantly between asymptomatic carriers with a family history of DCM and those found to carry a TTNtv as a secondary finding on testing performed for another reason. Nevertheless, subtle markers of reduced cardiac function have been found in carriers in the general population (Schafer et al., Reference Schafer, de Marvao, Adami, Fiedler, Ng, Khin, Rackham, van Heesch, Pua, Kui, Walsh, Tayal, Prasad, Dawes, Ko, Sim, Chan, Chin, Mazzarotto, Barton, Kreuchwig, de Kleijn, Totman, Biffi, Tee, Rueckert, Schneider, Faber, Regitz-Zagrosek, Seidman, Seidman, Linke, Kovalik, O’Regan, Ware, Hubner and Cook2016), suggesting they may be more susceptible to extrinsic insults, such as alcohol and cardiotoxic chemotherapy.

Recent research has emphasised the importance of common genetic variation or polygenic risk in determining the risk of developing DCM (Pirruccello et al., Reference Pirruccello, Bick, Wang, Chaffin, Friedman, Yao, Guo, Venkatesh, Taylor, Post, Rich, Lima, Rotter, Philippakis, Lubitz, Ellinor, Khera, Kathiresan and Aragam2020; Tadros et al., Reference Tadros, Francis, Xu, Vermeer, Harper, Huurman, Kelu Bisabu, Walsh, Hoorntje, Te Rijdt, Buchan, van Velzen, van Slegtenhorst, Vermeulen, Offerhaus, Bai, de Marvao, Lahrouchi, Beekman, Karper, Veldink, Kayvanpour, Pantazis, Baksi, Whiffin, Mazzarotto, Sloane, Suzuki, Schneider-Luftman, Elliott, Richard, Ader, Villard, Lichtner, Meitinger, Tanck, van Tintelen, Thain, McCarty, Hegele, Roberts, Amyot, Dube, Cadrin-Tourigny, Giraldeau, L’Allier, Garceau, Tardif, Boekholdt, Lumbers, Asselbergs, Barton, Cook, Prasad, O’Regan, van der Velden, Verweij, Talajic, Lettre, Pinto, Meder, Charron, de Boer, Christiaans, Michels, Wilde, Watkins, Matthews, Ware and Bezzina2021). Many patients without a rare variant cause are likely to have high polygenic risk contributing to the development of contractile dysfunction along with extrinsic factors. Polygenic risk is also likely to influence the penetrance of rare genetic variants, helping to explain the variable expressivity and incomplete penetrance commonly seen across families with a pathogenic variant. A future precision approach to determining disease risk in families with DCM is likely to integrate data on phenotype, rare and common genetic variation and the interaction with extrinsic insults.

Autosomal recessive transmission has also been described. This is of particular relevance in younger individuals and childhood cardiomyopathies. For example, biallelic loss of function mutations in the nebulin-related anchoring protein gene (NRAP) have been identified in some individuals with severe sporadic DCM and have been proposed to cause low-penetrant recessive disease (Iuso et al., Reference Iuso, Wiersma, Schuller, Pode-Shakked, Marek-Yagel, Grigat, Schwarzmayr, Berutti, Alhaddad, Kanon, Grzeschik, Okun, Perles, Salem, Barel, Vardi, Rubinshtein, Tirosh, Dubnov-Raz, Messias, Terrile, Barshack, Volkov, Avivi, Eyal, Mastantuono, Kumbar, Abudi, Braunisch, Strom, Meitinger, Hoffmann, Prokisch, Haack, Brundel, Haas, Sibon and Anikster2018; Koskenvuo et al., Reference Koskenvuo, Saarinen, Ahonen, Tommiska, Weckstrom, Seppala, Tuupanen, Kangas-Kontio, Schleit, Helio, Hathaway, Gummesson, Dahlberg, Ojala, Vepsalainen, Kytola, Muona, Sistonen, Salmenpera, Gentile, Paananen, Myllykangas, Alastalo and Helio2021). Several syndromic causes of DCM have been identified. These include dystrophinopathies such as Duchenne and Becker’s muscular dystrophy, and other eponymous syndromes including Barth syndrome (Hershberger et al., Reference Hershberger, Cowan, Morales and Siegfried2009; Hershberger et al., Reference Hershberger, Hedges and Morales2013). A raised creatinine kinase level and characteristic sub-epicardial fibrosis in the lateral wall are typical in patients with dystrophinopathies and cardiac manifestations may predate neuromuscular symptoms in Becker’s muscular dystrophy (Del Rio-Pertuz et al., Reference Del Rio-Pertuz, Morataya, Parmar, Dubay and Argueta-Sosa2022). Rare metabolic disorders, particularly inborn errors of metabolism, have also been associated with DCM (Guertl et al., Reference Guertl, Noehammer and Hoefler2000; Cox, Reference Cox2007). Broadly, these can be grouped into disorders of amino acid/organic acid metabolism, disorders of fatty acid metabolism, glycogen and lysosomal storage disorder and mitochondrial disorders (Guertl et al., Reference Guertl, Noehammer and Hoefler2000).

Stratifying arrhythmic risk in genetic DCM

The traditional approach to stratifying the risk of major ventricular arrhythmia in DCM relies on a combination of symptoms and left ventricular ejection fraction (LVEF). However, this “cause-agnostic” approach does not fully encapsulate the heterogeneity of DCM and a growing body of data support an increased risk of SCD with specific genotypes. This has begun to influence international guidelines on the selection of patients for implantable cardioverter defibrillators (ICDs) that recommend lower thresholds for such devices in patients with LMNA, FLNC, PLN or RBM20 variants and other high-risk features beyond LVEF (Zeppenfeld et al., Reference Zeppenfeld, Tfelt-Hansen, de Riva, Winkel, Behr, Blom, Charron, Corrado, Dagres, de Chillou, Eckardt, Friede, Haugaa, Hocini, Lambiase, Marijon, Merino, Peichl, Priori, Reichlin, Schulz-Menger, Sticherling, Tzeis, Verstrael, Volterrani and Group2022). This represents a wider trend in recent guidelines that attempt to risk stratify patients according to genotype and phenotype to make personalised decisions about their care, attempting to break down the traditional grouping of “non-ischemic cardiomyopathy” (Table 1).

Table 1. Genes with definite/strong association with DCM and their functional and phenotypic implications (14, 17)

Is genotype-specific therapy the answer?

Discovering a monogenic cause for DCM provides direct insight into the molecular mechanisms that drive disease. This creates the possibility of using precision therapies directed at the primary molecular basis of disease. Such approaches are not only relevant to those with symptomatic heart failure where they may be used alongside GDMT, but perhaps more importantly for asymptomatic individuals with early markers of disease or those with genetic susceptibility to developing disease later in life. Evidence-based treatments for the latter groups are currently lacking. Targeted therapy for disease prevention must be a priority. Strategies targeting the primary disease mechanisms may take different main approaches.

The immediately downstream molecular consequences of the variant represent attractive targets. Most genes associated with cardiomyopathy serve important functions within the cardiomyocyte, with their respective proteins carrying out specific functions. Disruptions in the function of these proteins, either through loss or gain of function, result in intracellular changes in signal transduction, prompting the cardiomyocyte to undergo adaptive changes (Reichart et al., Reference Reichart, Magnussen, Zeller and Blankenberg2019). Given the heterogeneity of DCM, downstream targeting of these processes would require a wide variety of agents to target the products of different genes implicated in the pathogenesis. One such target that was recently investigated in phase II and III trials was the heightened cardiac activity of ERK1/2, JNK, and p38 MAP kinases downstream from variants in LMNA associated with DCM (Muchir et al., Reference Muchir, Wu, Choi, Iwata, Morrow, Homma and Worman2012). Much hope arose from animal studies that demonstrated a reduction in adverse remodelling following the administration of a p38 inhibitor (Wu et al., Reference Wu, Muchir, Shan, Bonne and Worman2011; Laurini et al., Reference Laurini, Martinelli, Lanzicher, Puzzi, Borin, Chen, Long, Lee, Mestroni, Taylor, Sbaizero and Pricl2018). Unfortunately, these results were not translated into the phase III trial that was recently stopped due to futility.

Another example comes from the use of myosin modulators in sarcomeric DCM. Sarcomeric dysfunction is the primary mechanism of DCM in patients with TTNtv or relevant variants in MYH7. This is the opposite functional consequence of sarcomeric variants causing hypertrophic cardiomyopathy (HCM) that are associated with sarcomeric over-action. In the same way that promise has arisen from the use of myosin inhibitors, there is excitement about the potential use of agents such as danicamtiv and omecamtiv mecarbil that increase actin-myosin cross-bridging in sarcomeric DCM (Voors et al., Reference Voors, Tamby, Cleland, Koren, Forgosh, Gupta, Lund, Camacho, Karra, Swart, Pellicori, Wagner, Hershberger, Prasad, Anderson, Anto, Bell, Edelberg, Fang, Henze, Kelly, Kurio, Li, Wells, Yang, Teichman, Del Rio and Solomon2020; Teerlink et al., Reference Teerlink, Diaz, Felker, McMurray, Metra, Solomon, Adams, Anand, Arias-Mendoza, Biering-Sorensen, Bohm, Bonderman, Cleland, Corbalan, Crespo-Leiro, Dahlstrom, Echeverria, Fang, Filippatos, Fonseca, Goncalvesova, Goudev, Howlett, Lanfear, Li, Lund, Macdonald, Mareev, Momomura, O’Meara, Parkhomenko, Ponikowski, Ramires, Serpytis, Sliwa, Spinar, Suter, Tomcsanyi, Vandekerckhove, Vinereanu, Voors, Yilmaz, Zannad, Sharpsten, Legg, Varin, Honarpour, Abbasi, Malik and Kurtz2021). Similarly, emerging data suggest that TTNtv are associated with modifications in cardiac metabolism and energy utilisation (Verdonschot et al., Reference Verdonschot, Hazebroek, Derks, Barandiaran Aizpurua, Merken, Wang, Bierau, van den Wijngaard, Schalla, Abdul Hamid, van Bilsen, van Empel, Knackstedt, Brunner-La Rocca, Brunner, Krapels and Heymans2018; Ware and Cook, Reference Ware and Cook2018; Zhou et al., Reference Zhou, Ng, Ko, Fiedler, Khin, Lim, Sahib, Wu, Chothani, Schafer, Bay, Sinha, Cook and Yen2019). In particular, an upregulation in the transcription of important mitochondrial machinery may represent a compensatory response to sarcomeric dysfunction (Ware and Cook, Reference Ware and Cook2018). Targeting early mitochondrial dysfunction may therefore be a promising target for future investigation.

An example of precision therapy from current clinical practice is the use of sodium channel blockers such as flecainide or quinidine for DCM associated with SCN5A variants that result in an increased sodium current (Peters et al., Reference Peters, Thompson, Perrin, James, Zentner, Kalman, Vandenberg and Fatkin2022). A recent systematic review has shown such cardiomyopathies, typically associated with a high burden of ventricular arrhythmias, to be responsive to sodium channel blockers (Peters et al., Reference Peters, Thompson, Perrin, James, Zentner, Kalman, Vandenberg and Fatkin2022). It may be argued that such phenotypes are a primary electrical disease rather than a true cardiomyopathy. Nevertheless, the reversibility with widely available therapies emphasises the importance of achieving a genetic diagnosis, avoiding other unnecessary invasive procedures (Figure 3).

Figure 3. Cellular locations of some of the proteins with their respective genes associated with dilated cardiomyopathy.

Arguably, the most definitive treatment approaches are those that directly target the genetic variant (Verdonschot et al., Reference Verdonschot, Hazebroek, Ware, Prasad and Heymans2019). Various methods are currently being investigated to accomplish this objective, including: (1) gene editing – the use of CRISPR/Cas9 to directly edit the genetic sequence and restore normal protein function, (2) gene replacement therapy for cardiomyopathies associated with loss of function variants where the wild type gene is expressed, primarily through gene transfer techniques with viral vectors, (3) gene silencing therapy, primarily using small interfering RNA molecules to reduce the expression of abnormal functioning protein as a result of missense variants, and (4) exon skipping, involving the use of anti-sense oligonucleotides to mask exons during transcription and restoring the reading frame (Carrier et al., Reference Carrier, Mearini, Stathopoulou and Cuello2015; Gramlich et al., Reference Gramlich, Pane, Zhou, Chen, Murgia, Schotterl, Goedel, Metzger, Brade, Parrotta, Schaller, Gerull, Thierfelder, Aartsma-Rus, Labeit, Atherton, McGaughran, Harvey, Sinnecker, Mann, Laugwitz, Gawaz and Moretti2015; Prondzynski et al., Reference Prondzynski, Kramer, Laufer, Shibamiya, Pless, Flenner, Muller, Munch, Redwood, Hansen, Patten, Eschenhagen, Mearini and Carrier2017; Ma et al., Reference Ma, Zhang, Itzhaki, Zhang, Chen, Haddad, Kitani, Wilson, Tian, Shrestha, Wu, Lam, Sayed and Wu2018).

Much of the early progress in this area has been in Duchenne muscular dystrophy (DMD) where both exon skipping and gene editing have been used to restore dystrophin production in experimental models (Amoasii et al., Reference Amoasii, Hildyard, Li, Sanchez-Ortiz, Mireault, Caballero, Harron, Stathopoulou, Massey, Shelton, Bassel-Duby, Piercy and Olson2018). Early work has also demonstrated the potential of similar approaches in DCM associated with TTN, another similarly large gene with areas of redundant sequence (Gramlich et al., Reference Gramlich, Pane, Zhou, Chen, Murgia, Schotterl, Goedel, Metzger, Brade, Parrotta, Schaller, Gerull, Thierfelder, Aartsma-Rus, Labeit, Atherton, McGaughran, Harvey, Sinnecker, Mann, Laugwitz, Gawaz and Moretti2015; Romano et al., Reference Romano, Ghahremani, Zimmerman, Legere, Thakar, Ladha, Pettinato and Hinson2022). Pre-clinical studies in mice with the well-described PLN R14 gene deletion, have also used anti-sense oligonucleotides to decrease phospholamban activity, prevent cardiac dysfunction, and improve survival (Grote Beverborg et al., Reference Grote Beverborg, Spater, Knoll, Hidalgo, Yeh, Elbeck, Sillje, Eijgenraam, Siga, Zurek, Palmer, Pehrsson, Albery, Bomer, Hoes, Boogerd, Frisk, van Rooij, Damle, Louch, Wang, Fritsche-Danielson, Chien, Hansson, Mullick, de Boer and van der Meer2021; Deiman et al., Reference Deiman, Bomer, van der Meer and Grote Beverborg2022). This particular founder variant is associated with a malignant form of DCM, commonly encountered in the Netherlands.

Although substantial progress has been made in demonstrating the feasibility and potential of genome editing in cellular and murine models, numerous unanswered questions remain prior to advancing to human trials involving currently available techniques. Key considerations include ensuring the safety of viral delivery and accurately targeting the vector to the intended site with appropriate dosage (Colella et al., Reference Colella, Ronzitti and Mingozzi2018).

Precision therapy in gene elusive disease

Whilst genetic therapies hold great promise for those with monogenic disease, the majority will have little relevance for the majority of patients with DCM without a rare variant genetic cause. This group of patients are likely to have a diverse range of disease mechanisms including activation of fibroinflammatory pathways and metabolic dysfunction, driven by extrinsic causes including toxic insults, inflammatory or metabolic disease as well as genetic susceptibility related to common genetic variation (Reichart et al., Reference Reichart, Lindberg, Maatz, Miranda, Viveiros, Shvetsov, Gartner, Nadelmann, Lee, Kanemaru, Ruiz-Orera, Strohmenger, DeLaughter, Patone, Zhang, Woehler, Lippert, Kim, Adami, Gorham, Barnett, Brown, Buchan, Chowdhury, Constantinou, Cranley, Felkin, Fox, Ghauri, Gummert, Kanda, Li, Mach, McDonough, Samari, Shahriaran, Yapp, Stanasiuk, Theotokis, Theis, van den Bogaerdt, Wakimoto, Ware, Worth, Barton, Lee, Teichmann, Milting, Noseda, Oudit, Heinig, Seidman, Hubner and Seidman2022). Characterising these mechanisms in individual patients using precision phenotyping may help guide targeted therapy. The integration of advanced cardiac imaging and biomarkers offer huge potential to individualise management.

Myocardial fibrosis

In DCM the balance between extracellular matrix (ECM) synthesis and degradation is disrupted (Piek et al., Reference Piek, de Boer and Sillje2016). This leads to the formation myocardial fibrosis. Fibrosis is initiated by the activation and differentiation of fibroblasts into myofibroblasts, triggered by transforming growth factor (TGF-β) (Khalil et al., Reference Khalil, Kanisicak, Prasad, Correll, Fu, Schips, Vagnozzi, Liu, Huynh, Lee, Karch and Molkentin2017). Myofibroblasts produce higher levels of ECM proteins, contributing to the development of fibrosis (Nagaraju et al., Reference Nagaraju, Robinson, Abdesselem, Trenson, Dries, Gilbert, Janssens, Van Cleemput, Rega, Meyns, Roderick, Driesen and Sipido2019). Fibrosis leads to reduction in compliance of the diseased myocardium and acts as a substrate for arrhythmias (de Jong S et al., Reference de Jong, van Veen, van Rijen and de Bakker2011; Ellims et al., Reference Ellims, Shaw, Stub, Iles, Hare, Slavin, Kaye and Taylor2014). It is recognised as a key disease mechanism across a spectrum of DCM and is thought to represent a modifiable target for treatment, particularly in early disease before replacement fibrosis or scar has developed (Halliday and Prasad, Reference Halliday and Prasad2019).

Fibrosis is likely to be driven via multiple different pathways. Neurohormonal activation as part of the heart failure syndrome with upregulation of angiotensin II and aldosterone is likely to play an important role (Halliday and Prasad, Reference Halliday and Prasad2019). Myocardial inflammation and immune activation are also tightly linked to fibrotic pathways and are likely to play an important role in a subset of patients (Halliday and Prasad, Reference Halliday and Prasad2019). Upregulation of fibrosis also appears to be an early feature of specific genotypes including FLNC, DSP and LMNA (Augusto et al., Reference Augusto, Eiros, Nakou, Moura-Ferreira, Treibel, Captur, Akhtar, Protonotarios, Gossios, Savvatis, Syrris, Mohiddin, Moon, Elliott and Lopes2020). Targeting patients in these groups with anti-fibrotic agents may therefore be fruitful.

Mineralocorticoid receptor antagonists, which are an important part of GDMT show promise as potential antifibrotic drugs for patients with DCM (Izawa et al., Reference Izawa, Murohara, Nagata, Isobe, Asano, Amano, Ichihara, Kato, Ohshima, Murase, Iino, Obata, Noda, Okumura and Yokota2005; Al-Khatib et al., Reference Al-Khatib, Stevenson, Ackerman, Bryant, Callans, Curtis, Deal, Dickfeld, Field, Fonarow, Gillis, Granger, Hammill, Hlatky, Joglar, Kay, Matlock, Myerburg and Page2018; McDonagh et al., Reference McDonagh, Metra, Adamo, Gardner, Baumbach, Bohm, Burri, Butler, Celutkiene, Chioncel, Cleland, Coats, Crespo-Leiro, Farmakis, Gilard, Heymans, Hoes, Jaarsma, Jankowska, Lainscak, Lam, Lyon, McMurray, Mebazaa, Mindham, Muneretto, Francesco Piepoli, Price, Rosano, Ruschitzka, Kathrine Skibelund and Group2021). These medications can influence remodelling, reduce biomarkers associated with collagen biosynthesis, and improve patient outcomes (Sharma et al., Reference Sharma, Pokharel, van Brakel, van Berlo, Cleutjens, Schroen, Andre, Crijns, Gabius, Maessen and Pinto2004; Besler et al., Reference Besler, Lang, Urban, Rommel, Roeder, Fengler, Blazek, Kandolf, Klingel, Thiele, Linke, Schuler, Adams and Lurz2017). Evidence also suggests that antifibrotic agents used in other diseases, such as pirfenidone, may hold some promise in the treatment of heart failure (Lewis et al., Reference Lewis, Dodd, Clayton, Bedson, Eccleson, Schelbert, Naish, Jimenez, Williams, Cunnington, Ahmed, Cooper, Rajavarma, Russell, McDonagh, Williamson and Miller2021).

Cardiac metabolism

A key characteristic of DCM and heart failure is reduced oxidative metabolism and a shift from fatty acid oxidation to increased glucose utilisation (Heggermont et al., Reference Heggermont, Papageorgiou, Heymans and van Bilsen2016). Whether this is adaptive or maladaptive remains a topic of debate. Other important metabolic changes include increased ketone metabolism that is thought to represent a therapeutic target. Regardless of the cause, a myocardial energy deficit appears to be an important pathway in perpetuating the progression of the disease (Heggermont et al., Reference Heggermont, Papageorgiou, Heymans and van Bilsen2016; Sacchetto et al., Reference Sacchetto, Sequeira, Bertero, Dudek, Maack and Calore2019).

It appears likely that the myocardial energetic phenotype and impact of impaired myocardial energetics will differ across the spectrum of DCM. This may be influenced by co-morbidities such as diabetes mellitus as well as age that are associated with impairment of energetics (Chowdhary et al., Reference Chowdhary, Javed, Thirunavukarasu, Jex, Kotha, Kellman, Swoboda, Greenwood, Plein and Levelt2022). Genotype-specific differences are also likely to exist. In recent studies, the impact of DCM-causing TTNtv was explored in rats, revealing a correlation with impaired autophagy, reduced oxygen consumption rate, increased production of reactive oxygen species (ROS), and elevated ubiquitination of mitochondrial proteins in cardiomyocytes (Sacchetto et al., Reference Sacchetto, Sequeira, Bertero, Dudek, Maack and Calore2019; Zhou et al., Reference Zhou, Ng, Ko, Fiedler, Khin, Lim, Sahib, Wu, Chothani, Schafer, Bay, Sinha, Cook and Yen2019). This is supported by data from human myocardial tissue demonstrating important changes in the transcription of proteins relevant to mitochondrial function in carriers of TTNtv (Verdonschot et al., Reference Verdonschot, Hazebroek, Derks, Barandiaran Aizpurua, Merken, Wang, Bierau, van den Wijngaard, Schalla, Abdul Hamid, van Bilsen, van Empel, Knackstedt, Brunner-La Rocca, Brunner, Krapels and Heymans2018; Reichart et al., Reference Reichart, Lindberg, Maatz, Miranda, Viveiros, Shvetsov, Gartner, Nadelmann, Lee, Kanemaru, Ruiz-Orera, Strohmenger, DeLaughter, Patone, Zhang, Woehler, Lippert, Kim, Adami, Gorham, Barnett, Brown, Buchan, Chowdhury, Constantinou, Cranley, Felkin, Fox, Ghauri, Gummert, Kanda, Li, Mach, McDonough, Samari, Shahriaran, Yapp, Stanasiuk, Theotokis, Theis, van den Bogaerdt, Wakimoto, Ware, Worth, Barton, Lee, Teichmann, Milting, Noseda, Oudit, Heinig, Seidman, Hubner and Seidman2022). Additionally, an aberrant signalling pathway involving ERK1/2 was associated with altered mitochondrial shape, distribution, fragmentation, and degeneration in a mouse model of LMNA DCM (Galata et al., Reference Galata, Kloukina, Kostavasili, Varela, Davos, Makridakis, Bonne and Capetanaki2018).

There are many possible metabolic modulators that could be studied in a targeted fashion. There is interest in the use of the antioxidant and cofactor for mitochondrial electron transport, coenzyme Q10. Phase III trial data in heart failure with reduced ejection fraction was promising, however larger, more robust trials are required before routine clinical use (Mortensen et al., Reference Mortensen, Rosenfeldt, Kumar, Dolliner, Filipiak, Pella, Alehagen, Steurer, Littarru and Investigators2014). A mitochondrial-targeted form of coenzyme Q10, MitoQ, has also gained interest following convincing experimental data (Goh et al., Reference Goh, He, Song, Jinno, Rogers, Sethu, Halade, Rajasekaran, Liu, Prabhu, Darley-Usmar, Wende and Zhou2019). Whether some forms of DCM, such as those related to TTNtv, may gain more benefit from such therapies is unclear. It is also possible that such therapies will improve cardiac function through other pathways, such as by reducing endothelial dysfunction and reducing afterload (Roura and Bayes-Genis, Reference Roura and Bayes-Genis2009; Giannitsi et al., Reference Giannitsi, Bougiakli, Bechlioulis and Naka2019). Trimetazidine inhibits the protein thiolase I, responsible for the final step of beta-oxidation in the mitochondria. This results in a shift in substrate utilisation towards glucose metabolism (Tuunanen et al., Reference Tuunanen, Engblom, Naum, Nagren, Scheinin, Hesse, Juhani Airaksinen, Nuutila, Iozzo, Ukkonen, Opie and Knuuti2008). Perhexilene reduces fatty acid oxidation by inhibiting carnitine palmitoyltransferase-1 and similarly promotes a switch to glucose utilisation (Beadle et al., Reference Beadle, Williams, Kuehl, Bowater, Abozguia, Leyva, Yousef, Wagenmakers, Thies, Horowitz and Frenneaux2015). Early phase data have suggested that such agents may improve myocardial energetics and LV systolic function, however, later phase data are still lacking and concerns regarding the long-term safety of perhexiline remain (Tuunanen et al., Reference Tuunanen, Engblom, Naum, Nagren, Scheinin, Hesse, Juhani Airaksinen, Nuutila, Iozzo, Ukkonen, Opie and Knuuti2008; Zhang et al., Reference Zhang, Lu, Jiang, Zhang, Sun, Zou and Ge2012; Beadle et al., Reference Beadle, Williams, Kuehl, Bowater, Abozguia, Leyva, Yousef, Wagenmakers, Thies, Horowitz and Frenneaux2015; Fan et al., Reference Fan, Niu and Ma2018). Debate continues whether downregulating fatty oxidation is truly beneficial (Watson et al., Reference Watson, Green, Lewis, Arvidsson, De Maria, Arheden, Heiberg, Clarke, Rodgers, Valkovic, Neubauer, Herring and Rider2023). Much therefore remains to be understood about the role of personalised metabolic therapy.

Given the likely variable impact of fibrosis, immune activation and metabolic dysfunction across the spectrum of DCM, it is essential that we have accessible non-invasive methods to assess the role of these mechanisms in individual cases to guide precision and targeted therapies. Cardiac imaging as well as circulating biomarkers have the potential to play an important role.

Cardiac imaging

Whilst echocardiography (TTE) serves as the initial modality for diagnosing patients with heart failure with reduced ejection fraction, it is unable to reliably discriminate the cause of left ventricular dysfunction. Much data supports the use of cardiac magnetic resonance (CMR) imaging as a valuable tool for discriminating between ischaemic and non-ischaemic aetiologies and refining the cause and mechanism of non-ischaemic LV dysfunction (Japp et al., Reference Japp, Gulati, Cook, Cowie and Prasad2016; Halliday, Reference Halliday2022). It does so through detailed tissue characterisation using late gadolinium enhancement (LGE) imaging and parametric mapping (Japp et al., Reference Japp, Gulati, Cook, Cowie and Prasad2016; Halliday, Reference Halliday2022; Merlo et al., Reference Merlo, Gagno, Baritussio, Bauce, Biagini, Canepa, Cipriani, Castelletti, Dellegrottaglie, Guaricci, Imazio, Limongelli, Musumeci, Parisi, Pica, Pontone, Todiere, Torlasco, Basso, Sinagra, Filardi, Indolfi, Autore and Barison2023). This insight currently provides important information that guides selection of patients for ICDs and may also help individualise other treatment decisions in the future.

LGE represents replacement myocardial fibrosis and is present in around one-third to a half of cases (Kuruvilla et al., Reference Kuruvilla, Adenaw, Katwal, Lipinski, Kramer and Salerno2014; Di Marco et al., Reference Di Marco, Anguera, Schmitt, Klem, Neilan, White, Sramko, Masci, Barison, McKenna, Mordi, Haugaa, Leyva, Rodriguez Capitan, Satoh, Nabeta, Dallaglio, Campbell, Sabate and Cequier2017). LGE presence has been found to be a predictor of mortality, hospitalisation, and sudden cardiac death (SCD). Furthermore, the presence, extent, and patterns of LGE may provide additional valuable predictive information regarding malignant ventricular arrhythmias (VAs) or left ventricular (LV) reverse remodelling (Kuruvilla et al., Reference Kuruvilla, Adenaw, Katwal, Lipinski, Kramer and Salerno2014). The presence of LGE is now included in guidelines for primary prevention ICD implantation (McDonagh et al., Reference McDonagh, Metra, Adamo, Gardner, Baumbach, Bohm, Burri, Butler, Celutkiene, Chioncel, Cleland, Coats, Crespo-Leiro, Farmakis, Gilard, Heymans, Hoes, Jaarsma, Jankowska, Lainscak, Lam, Lyon, McMurray, Mebazaa, Mindham, Muneretto, Francesco Piepoli, Price, Rosano, Ruschitzka, Kathrine Skibelund and Group2021; Zeppenfeld et al., Reference Zeppenfeld, Tfelt-Hansen, de Riva, Winkel, Behr, Blom, Charron, Corrado, Dagres, de Chillou, Eckardt, Friede, Haugaa, Hocini, Lambiase, Marijon, Merino, Peichl, Priori, Reichlin, Schulz-Menger, Sticherling, Tzeis, Verstrael, Volterrani and Group2022). The pattern of myocardial fibrosis on CMR may also point towards particularly genetic aetiologies. Variants in desmoplakin (DSP) and filamin C (FLNC) have been shown to be associated with ring-like patterns of myocardial fibrosis which has been associated with worse outcomes (Augusto et al., Reference Augusto, Eiros, Nakou, Moura-Ferreira, Treibel, Captur, Akhtar, Protonotarios, Gossios, Savvatis, Syrris, Mohiddin, Moon, Elliott and Lopes2020). Parametric mapping with CMR also offers the ability to quantify interstitial changes, including fibrosis and oedema. Another exciting emerging fibrosis imaging technique is 68-gallium-labelled fibroblast activation protein inhibitor (FAPI) positron emission tomography (PET). This nuclear technique offers the potential to image fibrosis activity, anticipate fibrotic remodelling and prevent clinical disease before it occurs using targeted anti-fibrotic therapies.

31Phosphorus magnetic resonance offers the unique ability to study myocardial energetics in vivo. Studies have confirmed that DCM is characterised by a decrease in the ratio of phosphocreatine to adenosine triphosphate, a marker of impaired energetics (Stoll et al., Reference Stoll, Clarke, Levelt, Liu, Myerson, Robson, Neubauer and Rodgers2016). This has been shown to improve with reverse remodelling and predict outcome (Neubauer et al., Reference Neubauer, Horn, Cramer, Harre, Newell, Peters, Pabst, Ertl, Hahn, Ingwall and Kochsiek1997). This technique offers the ability to characterise the metabolic phenotype of individual patients and perhaps identify those who may gain most benefit from targeted metabolic therapies.

Diffusion tensor CMR enables comprehensive evaluation of cardiac microstructure revealing intricate details of myocardial wall mechanics, including the rotational torsion of myocardial sheetlets. This emerging technique may offer unique insight into the response to therapies targeting the sarcomere (Nielles-Vallespin et al., Reference Nielles-Vallespin, Khalique, Ferreira, de Silva, Scott, Kilner, McGill, Giannakidis, Gatehouse, Ennis, Aliotta, Al-Khalil, Kellman, Mazilu, Balaban, Firmin, Arai and Pennell2017).

Blood biomarkers

Circulating biomarkers provide the opportunity to characterise metabolic derangement, collagen turnover as well inflammatory and immune activation (Rubis et al., Reference Rubis, Dziewiecka, Wisniowska-Smialek, Banys, Urbanczyk-Zawadzka, Krupinski, Mielnik, Karabinowska and Garlitski2022). This has the potential to guide therapy decisions. One potential disadvantage is that many are not cardiac-specific. For example, circulating serum biomarkers of fibrosis reflect collagen turnover not only in the heart but also in various organs such as vessels, liver, and bone. Nevertheless, the carboxy-terminal propeptide of procollagen type I (PICP) and the amino-terminal propeptide of procollagen type III (PIIINP) have been correlated cardiac fibrosis observed on histology (Izawa et al., Reference Izawa, Murohara, Nagata, Isobe, Asano, Amano, Ichihara, Kato, Ohshima, Murase, Iino, Obata, Noda, Okumura and Yokota2005; Lopez et al., Reference Lopez, Gonzalez and Diez2010; Rubis et al., Reference Rubis, Dziewiecka, Wisniowska-Smialek, Banys, Urbanczyk-Zawadzka, Krupinski, Mielnik, Karabinowska and Garlitski2022) and elevated levels of these peptides predict an unfavourable outcome in patients with HF (Martos et al., Reference Martos, Baugh, Ledwidge, O’Loughlin, Murphy, Conlon, Patle, Donnelly and McDonald2009; Sweeney et al., Reference Sweeney, Corden and Cook2020; Cleland et al., Reference Cleland, Ferreira, Mariottoni, Pellicori, Cuthbert, Verdonschot, Petutschnigg, Ahmed, Cosmi, Brunner La Rocca, Mamas, Clark, Edelmann, Pieske, Khan, McDonald, Rouet, Staessen, Mujaj, Gonzalez, Diez, Hazebroek, Heymans, Latini, Grojean, Pizard, Girerd, Rossignol, Collier and Zannad2021). There has been interest in using markers to select patients who may benefit the most from anti-fibrotic therapy (Cleland et al., Reference Cleland, Ferreira, Mariottoni, Pellicori, Cuthbert, Verdonschot, Petutschnigg, Ahmed, Cosmi, Brunner La Rocca, Mamas, Clark, Edelmann, Pieske, Khan, McDonald, Rouet, Staessen, Mujaj, Gonzalez, Diez, Hazebroek, Heymans, Latini, Grojean, Pizard, Girerd, Rossignol, Collier and Zannad2021; Raafs et al., Reference Raafs, Verdonschot, Henkens, Adriaans, Wang, Derks, Abdul Hamid, Knackstedt, van Empel, Diez, Brunner-La Rocca, Brunner, Gonzalez, Bekkers, Heymans and Hazebroek2021). Galectin-3 is another marker of fibro-inflammatory activity and has been identified as a prognostic marker due to its association with worse outcomes in DCM (Sharma et al., Reference Sharma, Pokharel, van Brakel, van Berlo, Cleutjens, Schroen, Andre, Crijns, Gabius, Maessen and Pinto2004; Besler et al., Reference Besler, Lang, Urban, Rommel, Roeder, Fengler, Blazek, Kandolf, Klingel, Thiele, Linke, Schuler, Adams and Lurz2017). It appears likely that fibrosis plays an important role in driving early disease in particular phenotypes. The extent to which biomarkers will be able to guide therapy prior to the emergence of symptomatic DCM is unknown. One advantage of using them in susceptible individuals or those with early disease is that extra-cardiac causes of fibrosis are less likely to be relevant in this younger, less co-morbid group. Additionally, other markers such has high-sensitivity troponin T (hsTnT) and N-terminal prohormone brain natriuretic peptide (nt-proBNP) may have an important role in predicting disease progression (Chmielewski et al., Reference Chmielewski, Michalak, Kowalik, Franaszczyk, Sobieszczanska-Malek, Truszkowska, Stepien-Wojno, Biernacka, Foss-Nieradko, Lewandowski, Oreziak, Bilinska, Kusmierczyk, Tesson, Grzybowski, Zielinski, Ploski and Bilinska2020; Suresh et al., Reference Suresh, Martens and Tang2022). Both, for example, have been associated with the risk of malignant ventricular arrythmias in LMNA mutation carriers (Figure 4).

Figure 4. Precision medicine for dilated cardiomyopathy.

Precision phenotyping in DCM

Another key challenge is integrating these multidimensional data in a simple, accessible way to create a ground truth for the patient we see in clinic. Several studies have used unbiased clustering analysis known as phenomapping, in patients with various forms of heart failure including DCM, to help define subgroups of patients (Shah et al., Reference Shah, Katz, Selvaraj, Burke, Yancy, Gheorghiade, Bonow, Huang and Deo2015; Verdonschot et al., Reference Verdonschot, Merlo, Dominguez, Wang, Henkens, Adriaens, Hazebroek, Mase, Escobar, Cobas-Paz, Derks, van den Wijngaard, Krapels, Brunner, Sinagra, Garcia-Pavia and Heymans2020; Tayal et al., Reference Tayal, Gregson, Buchan, Whiffin, Halliday, Lota, Roberts, Baksi, Voges, Jarman, Baruah, Frenneaux, Cleland, Barton, Pennell, Ware, Cook and Prasad2022). The heterogenous aetiology of DCM makes it imminently suitable for this form of classification. Tayal et colleagues used a machine-learning based approach to cluster patients based on clinical, imaging, genetic and circulating characteristics and identified distinct subclasses of DCM with shared and distinct disease mechanisms (Tayal et al., Reference Tayal, Gregson, Buchan, Whiffin, Halliday, Lota, Roberts, Baksi, Voges, Jarman, Baruah, Frenneaux, Cleland, Barton, Pennell, Ware, Cook and Prasad2022). Verdonschot and colleagues used a similar approach incorporating transcriptomics to identify distinct transcriptomic profiles, including, pro-fibrotic, pro-inflammatory and metabolic subtypes (Verdonschot et al., Reference Verdonschot, Merlo, Dominguez, Wang, Henkens, Adriaens, Hazebroek, Mase, Escobar, Cobas-Paz, Derks, van den Wijngaard, Krapels, Brunner, Sinagra, Garcia-Pavia and Heymans2020). Both groups then used common clinical variables to discriminate between the groups so that this approach could be translated more easily into clinical practice.

By untangling the upstream causes and downstream active processes unique to each patient, such approaches may illuminate the targets for therapeutic intervention. The heterogeneity of DCM necessitates a personalised approach, with treatment strategies designed to benefit the individual patient subgroups that emerge from thorough phenotypic characterisation.

Co-morbidities and lifestyle

In the individualised treatment of individuals with DCM, it is important to also manage comorbidities such as coronary artery disease, hypertension, diabetes, thyroid disease, anaemia, and obesity (Reichart et al., Reference Reichart, Magnussen, Zeller and Blankenberg2019; Verdonschot et al., Reference Verdonschot, Hazebroek, Ware, Prasad and Heymans2019; Zeppenfeld et al., Reference Zeppenfeld, Tfelt-Hansen, de Riva, Winkel, Behr, Blom, Charron, Corrado, Dagres, de Chillou, Eckardt, Friede, Haugaa, Hocini, Lambiase, Marijon, Merino, Peichl, Priori, Reichlin, Schulz-Menger, Sticherling, Tzeis, Verstrael, Volterrani and Group2022). It is likely that such co-morbidities interact with intrinsic susceptibility to develop contractile impairment. Whether more intensive treatment and stricter control of these issues improves outcomes remains unclear. Special attention should also be paid to the impact of alcohol and cardiotoxic chemotherapy, such as anthracyclines (Ware et al., Reference Ware, Amor-Salamanca, Tayal, Govind, Serrano, Salazar-Mendiguchia, Garcia-Pinilla, Pascual-Figal, Nunez, Guzzo-Merello, Gonzalez-Vioque, Bardaji, Manito, Lopez-Garrido, Padron-Barthe, Edwards, Whiffin, Walsh, Buchan, Midwinter, Wilk, Prasad, Pantazis, Baski, O’Regan, Alonso-Pulpon, Cook, Lara-Pezzi, Barton and Garcia-Pavia2018; Andersson et al., Reference Andersson, Schou, Gustafsson and Torp-Pedersen2022; Tayal et al., Reference Tayal, Verdonschot, Hazebroek, Howard, Gregson, Newsome, Gulati, Pua, Halliday, Lota, Buchan, Whiffin, Kanapeckaite, Baruah, Jarman, O’Regan, Barton, Ware, Pennell, Adriaans, Bekkers, Donovan, Frenneaux, Cooper, Januzzi, Cleland, Cook, Deo, Heymans and Prasad2022). Whilst it is clear that excessive amounts of alcohol may be harmful, it is debatable whether low or moderate levels of consumption lead to adverse remodelling and unclear whether abstinence should be recommended (Andersson et al., Reference Andersson, Schou, Gustafsson and Torp-Pedersen2022). It is possible that specific genotypes may lead to increased susceptibility to cardiotoxins (Ware et al. 2018). Individualised exercise prescription is another important factor to consider. Patients with symptomatic DCM or features of increased risk should avoid engaging in high-intensity or competitive sports (Pelliccia et al., Reference Pelliccia, Solberg, Papadakis, Adami, Biffi, Caselli, La Gerche, Niebauer, Pressler, Schmied, Serratosa, Halle, Van Buuren, Borjesson, Carre, Panhuyzen-Goedkoop, Heidbuchel, Olivotto, Corrado, Sinagra and Sharma2019). There is particular concern for those with high-risk genotypes.

Conclusion

A precision medicine approach holds great promise for revolutionising our approach to patients with the heterogeneous family of diseases that make up DCM. By integrating findings from clinical data, genetic testing, advanced imaging and circulating biomarkers, clinicians can gain a detailed understanding of each patient’s disease that can help individualise treatment via a shared decision-making approach.

However, significant challenges remain. Integrating the breadth of available genomic and phenotypic data to predict individual risk remains a challenge. Whilst many disease-specific treatments are under investigation, some remain years away from clinical routine. Whilst disease mechanisms have been well characterised in advanced disease, at what stage these occur in the natural history of DCM and whether early targeted intervention will delay the onset of overt disease remains to be determined. Despite these hurdles, the incorporation of genomic and phenotypic data hold the potential to establish a novel clinical framework for evidence-based and personalised care in DCM.

Open peer review

To view the open peer review materials for this article, please visit http://doi.org/10.1017/pcm.2023.24.

Data availability statement

Data sharing not applicable – no new data generated.

Author contribution

Both authors contributed to the literature search, data analysis and manuscript preparation.

Financial support

B.P.H. is supported by a BHF Intermediate Clinical Research Fellowship awarded to B.P.H. (FS/ICRF/21/26019) and the Rosetrees Trust.

Competing interest

B.P.H. has served on an advisory board for Astra Zeneca. S.J. declares no competing interest.

References

Al-Khatib, SM, Stevenson, WG, Ackerman, MJ, Bryant, WJ, Callans, DJ, Curtis, AB, Deal, BJ, Dickfeld, T, Field, ME, Fonarow, GC, Gillis, AM, Granger, CB, Hammill, SC, Hlatky, MA, Joglar, JA, Kay, GN, Matlock, DD, Myerburg, RJ and Page, RL (2018) 2017 AHA/ACC/HRS guideline for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death: Executive summary: A report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines and the Heart Rhythm Society. Heart Rhythm 15(10), e190e252. https://doi.org/10.1016/j.hrthm.2017.10.035.CrossRefGoogle Scholar
Amoasii, L, Hildyard, JCW, Li, H, Sanchez-Ortiz, E, Mireault, A, Caballero, D, Harron, R, Stathopoulou, TR, Massey, C, Shelton, JM, Bassel-Duby, R, Piercy, RJ and Olson, EN (2018) Gene editing restores dystrophin expression in a canine model of Duchenne muscular dystrophy. Science 362(6410), 8691. https://doi.org/10.1126/science.aau1549.CrossRefGoogle Scholar
Andersson, C, Schou, M, Gustafsson, F and Torp-Pedersen, C (2022) Alcohol intake in patients with cardiomyopathy and heart failure: Consensus and controversy. Circulation: Heart Failure 15(8), e009459. https://doi.org/10.1161/CIRCHEARTFAILURE.121.009459.Google ScholarPubMed
Augusto, JB, Eiros, R, Nakou, E, Moura-Ferreira, S, Treibel, TA, Captur, G, Akhtar, MM, Protonotarios, A, Gossios, TD, Savvatis, K, Syrris, P, Mohiddin, S, Moon, JC, Elliott, PM and Lopes, LR (2020) Dilated cardiomyopathy and arrhythmogenic left ventricular cardiomyopathy: A comprehensive genotype-imaging phenotype study. European Heart Journal Cardiovascular Imaging 21(3), 326336. https://doi.org/10.1093/ehjci/jez188.Google ScholarPubMed
Beadle, RM, Williams, LK, Kuehl, M, Bowater, S, Abozguia, K, Leyva, F, Yousef, Z, Wagenmakers, AJ, Thies, F, Horowitz, J and Frenneaux, MP (2015) Improvement in cardiac energetics by perhexiline in heart failure due to dilated cardiomyopathy. JACC Heart Fail 3(3), 202211. https://doi.org/10.1016/j.jchf.2014.09.009.CrossRefGoogle ScholarPubMed
Benjamin, EJ, Muntner, P, Alonso, A, Bittencourt, MS, Callaway, CW, Carson, AP, Chamberlain, AM, Chang, AR, Cheng, S, Das, SR, Delling, FN, Djousse, L, Elkind, MSV, Ferguson, JF, Fornage, M, Jordan, LC, Khan, SS, Kissela, BM, Knutson, KL, Kwan, TW, Lackland, DT, Lewis, TT, Lichtman, JH, Longenecker, CT, Loop, MS, Lutsey, PL, Martin, SS, Matsushita, K, Moran, AE, Mussolino, ME, O’Flaherty, M, Pandey, A, Perak, AM, Rosamond, WD, Roth, GA, Sampson, UKA, Satou, GM, Schroeder, EB, Shah, SH, Spartano, NL, Stokes, A, Tirschwell, DL, Tsao, CW, Turakhia, MP, VanWagner, LB, Wilkins, JT, Wong, SS, Virani, SS and American Heart Association Council on E, Prevention Statistics C and Stroke Statistics S (2019) Heart disease and stroke Statistics-2019 update: A report from the American Heart Association. Circulation 139(10), e56e528. https://doi.org/10.1161/CIR.0000000000000659.CrossRefGoogle ScholarPubMed
Besler, C, Lang, D, Urban, D, Rommel, K-P, Roeder, M, Fengler, K, Blazek, S, Kandolf, R, Klingel, K, Thiele, H, Linke, A, Schuler, G, Adams, V and Lurz, P (2017) Plasma and cardiac Galectin-3 in patients with heart failure reflects both inflammation and fibrosis. Circulation: Heart Failure 10(3), e003804. https://doi.org/10.1161/CIRCHEARTFAILURE.116.003804.Google ScholarPubMed
Carrier, L, Mearini, G, Stathopoulou, K and Cuello, F (2015) Cardiac myosin-binding protein C (MYBPC3) in cardiac pathophysiology. Gene 573(2), 188197. https://doi.org/10.1016/j.gene.2015.09.008.CrossRefGoogle ScholarPubMed
Chambers, DC, Cherikh, WS, Goldfarb, SB, Hayes, D Jr, Kucheryavaya, AY, Toll, AE, Khush, KK, Levvey, BJ, Meiser, B, Rossano, JW, Stehlik, J, International Society for H and Lung, T (2018) The international thoracic organ transplant registry of the International Society for Heart and Lung Transplantation: Thirty-fifth adult lung and heart-lung transplant report-2018. Focus theme: Multiorgan Transplantation. The Journal of Heart and Lung Transplantation 37(10), 11691183. https://doi.org/10.1016/j.healun.2018.07.020.CrossRefGoogle ScholarPubMed
Chmielewski, P, Michalak, E, Kowalik, I, Franaszczyk, M, Sobieszczanska-Malek, M, Truszkowska, G, Stepien-Wojno, M, Biernacka, EK, Foss-Nieradko, B, Lewandowski, M, Oreziak, A, Bilinska, M, Kusmierczyk, M, Tesson, F, Grzybowski, J, Zielinski, T, Ploski, R and Bilinska, ZT (2020) Can circulating cardiac biomarkers be helpful in the assessment of LMNA mutation carriers? Journal of Clinical Medicine 9(5), 1443. https://doi.org/10.3390/jcm9051443.CrossRefGoogle ScholarPubMed
Chowdhary, A, Javed, W, Thirunavukarasu, S, Jex, N, Kotha, S, Kellman, P, Swoboda, P, Greenwood, JP, Plein, S and Levelt, E (2022) Cardiac adaptations to acute hemodynamic stress in function, perfusion, and energetics in type 2 diabetes with overweight and obesity. Diabetes Care 45(12), e176e178. https://doi.org/10.2337/dc22-0887.CrossRefGoogle ScholarPubMed
Cleland, JGF, Ferreira, JP, Mariottoni, B, Pellicori, P, Cuthbert, J, Verdonschot, JAJ, Petutschnigg, J, Ahmed, FZ, Cosmi, F, Brunner La Rocca, HP, Mamas, MA, Clark, AL, Edelmann, F, Pieske, B, Khan, J, McDonald, K, Rouet, P, Staessen, JA, Mujaj, B, Gonzalez, A, Diez, J, Hazebroek, M, Heymans, S, Latini, R, Grojean, S, Pizard, A, Girerd, N, Rossignol, P, Collier, TJ, Zannad, F and Committees HT and Investigators (2021) The effect of spironolactone on cardiovascular function and markers of fibrosis in people at increased risk of developing heart failure: The heart ‘OMics’ in AGEing (HOMAGE) randomized clinical trial. European Heart Journal 42(6), 684696. https://doi.org/10.1093/eurheartj/ehaa758.CrossRefGoogle ScholarPubMed
Colella, P, Ronzitti, G and Mingozzi, F (2018) Emerging issues in AAV-mediated in vivo gene therapy. Molecular Therapy - Methods & Clinical Development 8, 87104. https://doi.org/10.1016/j.omtm.2017.11.007.CrossRefGoogle ScholarPubMed
Cox, GF (2007) Diagnostic approaches to pediatric cardiomyopathy of metabolic genetic etiologies and their relation to therapy. Progress in Pediatric Cardiology 24(1), 1525. https://doi.org/10.1016/j.ppedcard.2007.08.013.CrossRefGoogle ScholarPubMed
de Jong, S, van Veen, TA, van Rijen, HV and de Bakker, JM (2011) Fibrosis and cardiac arrhythmias. Journal of Cardiovascular Pharmacology 57(6), 630638. https://doi.org/10.1097/FJC.0b013e318207a35f.CrossRefGoogle ScholarPubMed
Deiman, FE, Bomer, N, van der Meer, P and Grote Beverborg, N (2022) Review: Precision medicine approaches for genetic cardiomyopathy: Targeting Phospholamban R14del. Current Heart Failure Reports 19(4), 170179. https://doi.org/10.1007/s11897-022-00558-x.CrossRefGoogle ScholarPubMed
Del Rio-Pertuz, G, Morataya, C, Parmar, K, Dubay, S and Argueta-Sosa, E (2022) Dilated cardiomyopathy as the initial presentation of Becker muscular dystrophy: A systematic review of published cases. Orphanet Journal of Rare Diseases 17(1), 194. https://doi.org/10.1186/s13023-022-02346-1.CrossRefGoogle ScholarPubMed
Di Marco, A, Anguera, I, Schmitt, M, Klem, I, Neilan, TG, White, JA, Sramko, M, Masci, PG, Barison, A, McKenna, P, Mordi, I, Haugaa, KH, Leyva, F, Rodriguez Capitan, J, Satoh, H, Nabeta, T, Dallaglio, PD, Campbell, NG, Sabate, X and Cequier, A (2017) Late gadolinium enhancement and the risk for ventricular arrhythmias or sudden death in dilated cardiomyopathy: Systematic review and meta-analysis. JACC Heart Failure 5(1), 2838. https://doi.org/10.1016/j.jchf.2016.09.017.CrossRefGoogle ScholarPubMed
Ellims, AH, Shaw, JA, Stub, D, Iles, LM, Hare, JL, Slavin, GS, Kaye, DM and Taylor, AJ (2014) Diffuse myocardial fibrosis evaluated by post-contrast t1 mapping correlates with left ventricular stiffness. Journal of the American College of Cardiology 63(11), 11121118. https://doi.org/10.1016/j.jacc.2013.10.084.CrossRefGoogle ScholarPubMed
Fan, Q, Niu, Z and Ma, L (2018) Meta-analysis of trimetazidine treatment for cardiomyopathy. Bioscience Reports 38(3), BSR20171583. https://doi.org/10.1042/BSR20171583.CrossRefGoogle ScholarPubMed
Galata, Z, Kloukina, I, Kostavasili, I, Varela, A, Davos, CH, Makridakis, M, Bonne, G and Capetanaki, Y (2018) Amelioration of desmin network defects by alphaB-crystallin overexpression confers cardioprotection in a mouse model of dilated cardiomyopathy caused by LMNA gene mutation. Journal of Molecular and Cellular Cardiology 125, 7386. https://doi.org/10.1016/j.yjmcc.2018.10.017.CrossRefGoogle Scholar
Garcia-Pavia, P, Kim, Y, Alejandra Restrepo-Cordoba, M, Lunde, IG, Wakimoto, H, Smith, AM, Toepfer, CN, Getz, K, Gorham, J, Patel, P, Ito, K, Willcox, JA, Arany, Z, Li, J, Owens, AT, Govind, R, Nunez, B, Mazaika, E, Bayes-Genis, A, Walsh, R, Finkelman, B, Lupon, J, Whiffin, N, Serrano, I, Midwinter, W, Wilk, A, Bardaji, A, Ingold, N, Buchan, R, Tayal, U, Pascual-Figal, DA, de Marvao, A, Ahmad, M, Garcia-Pinilla, JM, Pantazis, A, Dominguez, F, Baksi, AJ, O’Regan, DP, Rosen, SD, Prasad, SK, Lara-Pezzi, E, Provencio, M, Lyon, AR, Alonso-Pulpon, L, Cook, SA, SR, DP, PJR, B, Aplenc, R, Seidman, JG, Ky, B, Ware, JS and Seidman, CE (2019) Genetic variants associated with cancer therapy-induced cardiomyopathy. Circulation 140, 31. https://doi.org/10.1161/CIRCULATIONAHA.118.037934.CrossRefGoogle ScholarPubMed
Giannitsi, S, Bougiakli, M, Bechlioulis, A and Naka, K (2019) Endothelial dysfunction and heart failure: A review of the existing bibliography with emphasis on flow mediated dilation. JRSM Cardiovascular Disease 8, 2048004019843047. https://doi.org/10.1177/2048004019843047.CrossRefGoogle ScholarPubMed
Goh, KY, He, L, Song, J, Jinno, M, Rogers, AJ, Sethu, P, Halade, GV, Rajasekaran, NS, Liu, X, Prabhu, SD, Darley-Usmar, V, Wende, AR and Zhou, L (2019) Mitoquinone ameliorates pressure overload-induced cardiac fibrosis and left ventricular dysfunction in mice. Redox Biology 21, 101100. https://doi.org/10.1016/j.redox.2019.101100.CrossRefGoogle ScholarPubMed
Gramlich, M, Pane, LS, Zhou, Q, Chen, Z, Murgia, M, Schotterl, S, Goedel, A, Metzger, K, Brade, T, Parrotta, E, Schaller, M, Gerull, B, Thierfelder, L, Aartsma-Rus, A, Labeit, S, Atherton, JJ, McGaughran, J, Harvey, RP, Sinnecker, D, Mann, M, Laugwitz, KL, Gawaz, MP and Moretti, A (2015) Antisense-mediated exon skipping: A therapeutic strategy for titin-based dilated cardiomyopathy. EMBO Molecular Medicine 7(5), 562576. https://doi.org/10.15252/emmm.201505047.CrossRefGoogle ScholarPubMed
Grote Beverborg, N, Spater, D, Knoll, R, Hidalgo, A, Yeh, ST, Elbeck, Z, Sillje, HHW, Eijgenraam, TR, Siga, H, Zurek, M, Palmer, M, Pehrsson, S, Albery, T, Bomer, N, Hoes, MF, Boogerd, CJ, Frisk, M, van Rooij, E, Damle, S, Louch, WE, Wang, QD, Fritsche-Danielson, R, Chien, KR, Hansson, KM, Mullick, AE, de Boer, RA and van der Meer, P (2021) Phospholamban antisense oligonucleotides improve cardiac function in murine cardiomyopathy. Nature Communications 12(1), 5180. https://doi.org/10.1038/s41467-021-25439-0.CrossRefGoogle ScholarPubMed
Guertl, B, Noehammer, C and Hoefler, G (2000) Metabolic cardiomyopathies. International Journal of Experimental Pathology 81(6), 349372. https://doi.org/10.1046/j.1365-2613.2000.00186.x.CrossRefGoogle ScholarPubMed
Halliday, BP (2022) State of the art: Multimodality imaging in dilated cardiomyopathy. Heart 108(23), 19101917. https://doi.org/10.1136/heartjnl-2022-321116.CrossRefGoogle ScholarPubMed
Halliday, BP and Prasad, SK (2019) The Interstitium in the hypertrophied heart. JACC: Cardiovascular Imaging 12(11 Pt 2), 23572368. https://doi.org/10.1016/j.jcmg.2019.05.033.Google ScholarPubMed
Heggermont, WA, Papageorgiou, AP, Heymans, S and van Bilsen, M (2016) Metabolic support for the heart: Complementary therapy for heart failure? European Journal of Heart Failure 18(12), 14201429. https://doi.org/10.1002/ejhf.678.CrossRefGoogle ScholarPubMed
Heidenreich, PA, Albert, NM, Allen, LA, Bluemke, DA, Butler, J, Fonarow, GC, Ikonomidis, JS, Khavjou, O, Konstam, MA, Maddox, TM, Nichol, G, Pham, M, Pina, IL, Trogdon, JG, American Heart Association Advocacy Coordinating Committee; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Radiology and Intervention; Council on Clinical Cardiology; Council on Epidemiology and Prevention and Stroke Council (2013) Forecasting the impact of heart failure in the United States: A policy statement from the American Heart Association. Circulation. Heart Failure 6(3), 606619. https://doi.org/10.1161/HHF.0b013e318291329a.CrossRefGoogle ScholarPubMed
Heidenreich, PA, Bozkurt, B, Aguilar, D, Allen, LA, Byun, JJ, Colvin, MM, Deswal, A, Drazner, MH, Dunlay, SM, Evers, LR, Fang, JC, Fedson, SE, Fonarow, GC, Hayek, SS, Hernandez, AF, Khazanie, P, Kittleson, MM, Lee, CS, Link, MS, Milano, CA, Nnacheta, LC, Sandhu, AT, Stevenson, LW, Vardeny, O, Vest, AR and Yancy, CW (2022) 2022 AHA/ACC/HFSA guideline for the management of heart failure: Executive summary: A report of the American College of Cardiology/American Heart Association joint committee on clinical practice guidelines. Circulation 145(18), e876e894. https://doi.org/10.1161/CIR.0000000000001062.Google Scholar
Herman, DS, Lam, L, Taylor, MR, Wang, L, Teekakirikul, P, Christodoulou, D, Conner, L, SR, DP, McDonough, B, Sparks, E, Teodorescu, DL, Cirino, AL, Banner, NR, Pennell, DJ, Graw, S, Merlo, M, Di Lenarda, A, Sinagra, G, Bos, JM, Ackerman, MJ, Mitchell, RN, Murry, CE, Lakdawala, NK, Ho, CY, Barton, PJ, Cook, SA, Mestroni, L, Seidman, JG and Seidman, CE (2012) Truncations of titin causing dilated cardiomyopathy. The New England Journal of Medicine 366(7), 619628. https://doi.org/10.1056/NEJMoa1110186.CrossRefGoogle ScholarPubMed
Hershberger, RE, Cowan, J, Morales, A and Siegfried, JD (2009) Progress with genetic cardiomyopathies: Screening, counseling, and testing in dilated, hypertrophic, and arrhythmogenic right ventricular dysplasia/cardiomyopathy. Circulation. Heart Failure 2(3), 253261. https://doi.org/10.1161/CIRCHEARTFAILURE.108.817346.CrossRefGoogle ScholarPubMed
Hershberger, RE, Hedges, DJ and Morales, A (2013) Dilated cardiomyopathy: The complexity of a diverse genetic architecture. Nature Reviews. Cardiology 10(9), 531547. https://doi.org/10.1038/nrcardio.2013.105.CrossRefGoogle ScholarPubMed
Huffman, MD, Berry, JD, Ning, H, Dyer, AR, Garside, DB, Cai, X, Daviglus, ML and Lloyd-Jones, DM (2013) Lifetime risk for heart failure among white and black Americans: Cardiovascular lifetime risk pooling project. Journal of the American College of Cardiology 61(14), 15101517. https://doi.org/10.1016/j.jacc.2013.01.022.CrossRefGoogle ScholarPubMed
Iuso, A, Wiersma, M, Schuller, HJ, Pode-Shakked, B, Marek-Yagel, D, Grigat, M, Schwarzmayr, T, Berutti, R, Alhaddad, B, Kanon, B, Grzeschik, NA, Okun, JG, Perles, Z, Salem, Y, Barel, O, Vardi, A, Rubinshtein, M, Tirosh, T, Dubnov-Raz, G, Messias, AC, Terrile, C, Barshack, I, Volkov, A, Avivi, C, Eyal, E, Mastantuono, E, Kumbar, M, Abudi, S, Braunisch, M, Strom, TM, Meitinger, T, Hoffmann, GF, Prokisch, H, Haack, TB, Brundel, B, Haas, D, Sibon, OCM and Anikster, Y (2018) Mutations in PPCS, encoding phosphopantothenoylcysteine synthetase, cause autosomal-recessive dilated cardiomyopathy. American Journal of Human Genetics 102(6), 10181030. https://doi.org/10.1016/j.ajhg.2018.03.022.CrossRefGoogle ScholarPubMed
Izawa, H, Murohara, T, Nagata, K, Isobe, S, Asano, H, Amano, T, Ichihara, S, Kato, T, Ohshima, S, Murase, Y, Iino, S, Obata, K, Noda, A, Okumura, K and Yokota, M (2005) Mineralocorticoid receptor antagonism ameliorates left ventricular diastolic dysfunction and myocardial fibrosis in mildly symptomatic patients with idiopathic dilated cardiomyopathy: A pilot study. Circulation 112(19), 29402945. https://doi.org/10.1161/CIRCULATIONAHA.105.571653.CrossRefGoogle ScholarPubMed
Japp, AG, Gulati, A, Cook, SA, Cowie, MR and Prasad, SK (2016) The diagnosis and evaluation of dilated cardiomyopathy. Journal of the American College of Cardiology 67(25), 29963010. https://doi.org/10.1016/j.jacc.2016.03.590.CrossRefGoogle ScholarPubMed
Jordan, E, Peterson, L, Ai, T, Asatryan, B, Bronicki, L, Brown, E, Celeghin, R, Edwards, M, Fan, J, Ingles, J, James, CA, Jarinova, O, Johnson, R, Judge, DP, Lahrouchi, N, Lekanne Deprez, RH, Lumbers, RT, Mazzarotto, F, Medeiros Domingo, A, Miller, RL, Morales, A, Murray, B, Peters, S, Pilichou, K, Protonotarios, A, Semsarian, C, Shah, P, Syrris, P, Thaxton, C, van Tintelen, JP, Walsh, R, Wang, J, Ware, J and Hershberger, RE (2021) Evidence-based assessment of genes in dilated cardiomyopathy. Circulation 144(1), 719. https://doi.org/10.1161/CIRCULATIONAHA.120.053033.CrossRefGoogle ScholarPubMed
Khalil, H, Kanisicak, O, Prasad, V, Correll, RN, Fu, X, Schips, T, Vagnozzi, RJ, Liu, R, Huynh, T, Lee, SJ, Karch, J and Molkentin, JD (2017) Fibroblast-specific TGF-beta-Smad2/3 signaling underlies cardiac fibrosis. The Journal of Clinical Investigation 127(10), 37703783. https://doi.org/10.1172/JCI94753.CrossRefGoogle ScholarPubMed
Koskenvuo, JW, Saarinen, I, Ahonen, S, Tommiska, J, Weckstrom, S, Seppala, EH, Tuupanen, S, Kangas-Kontio, T, Schleit, J, Helio, K, Hathaway, J, Gummesson, A, Dahlberg, P, Ojala, TH, Vepsalainen, V, Kytola, V, Muona, M, Sistonen, J, Salmenpera, P, Gentile, M, Paananen, J, Myllykangas, S, Alastalo, TP and Helio, T (2021) Biallelic loss-of-function in NRAP is a cause of recessive dilated cardiomyopathy. PLoS One 16(2), e0245681. https://doi.org/10.1371/journal.pone.0245681.CrossRefGoogle ScholarPubMed
Kuruvilla, S, Adenaw, N, Katwal, AB, Lipinski, MJ, Kramer, CM and Salerno, M (2014) Late gadolinium enhancement on cardiac magnetic resonance predicts adverse cardiovascular outcomes in nonischemic cardiomyopathy: A systematic review and meta-analysis. Circulation. Cardiovascular Imaging 7(2), 250258. https://doi.org/10.1161/CIRCIMAGING.113.001144.CrossRefGoogle ScholarPubMed
Laurini, E, Martinelli, V, Lanzicher, T, Puzzi, L, Borin, D, Chen, SN, Long, CS, Lee, P, Mestroni, L, Taylor, MRG, Sbaizero, O and Pricl, S (2018) Biomechanical defects and rescue of cardiomyocytes expressing pathologic nuclear lamins. Cardiovascular Research 114(6), 846857. https://doi.org/10.1093/cvr/cvy040.CrossRefGoogle ScholarPubMed
Lewis, GA, Dodd, S, Clayton, D, Bedson, E, Eccleson, H, Schelbert, EB, Naish, JH, Jimenez, BD, Williams, SG, Cunnington, C, Ahmed, FZ, Cooper, A, Rajavarma, V, Russell, S, McDonagh, T, Williamson, PR and Miller, CA (2021) Pirfenidone in heart failure with preserved ejection fraction: A randomized phase 2 trial. Nature Medicine 27(8), 14771482. https://doi.org/10.1038/s41591-021-01452-0.CrossRefGoogle ScholarPubMed
Lopez, B, Gonzalez, A and Diez, J (2010) Circulating biomarkers of collagen metabolism in cardiac diseases. Circulation 121(14), 16451654. https://doi.org/10.1161/CIRCULATIONAHA.109.912774.CrossRefGoogle ScholarPubMed
Lukas Laws, J, Lancaster, MC, Ben Shoemaker, M, Stevenson, WG, Hung, RR, Wells, Q, Marshall Brinkley, D, Hughes, S, Anderson, K, Roden, D and Stevenson, LW (2022) Arrhythmias as presentation of genetic cardiomyopathy. Circulation Research 130(11), 16981722. https://doi.org/10.1161/CIRCRESAHA.122.319835.CrossRefGoogle ScholarPubMed
Ma, N, Zhang, JZ, Itzhaki, I, Zhang, SL, Chen, H, Haddad, F, Kitani, T, Wilson, KD, Tian, L, Shrestha, R, Wu, H, Lam, CK, Sayed, N and Wu, JC (2018) Determining the pathogenicity of a genomic variant of uncertain significance using CRISPR/Cas9 and human-induced pluripotent stem cells. Circulation 138(23), 26662681. https://doi.org/10.1161/circulationaha.117.032273.CrossRefGoogle ScholarPubMed
Martos, R, Baugh, J, Ledwidge, M, O’Loughlin, C, Murphy, NF, Conlon, C, Patle, A, Donnelly, SC and McDonald, K (2009) Diagnosis of heart failure with preserved ejection fraction: Improved accuracy with the use of markers of collagen turnover. European Journal of Heart Failure 11(2), 191197. https://doi.org/10.1093/eurjhf/hfn036.CrossRefGoogle ScholarPubMed
McDonagh, TA, Metra, M, Adamo, M, Gardner, RS, Baumbach, A, Bohm, M, Burri, H, Butler, J, Celutkiene, J, Chioncel, O, Cleland, JGF, Coats, AJS, Crespo-Leiro, MG, Farmakis, D, Gilard, M, Heymans, S, Hoes, AW, Jaarsma, T, Jankowska, EA, Lainscak, M, Lam, CSP, Lyon, AR, McMurray, JJV, Mebazaa, A, Mindham, R, Muneretto, C, Francesco Piepoli, M, Price, S, Rosano, GMC, Ruschitzka, F, Kathrine Skibelund, A and Group, ESCSD (2021) 2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure. European Heart Journal 42(36), 35993726. https://doi.org/10.1093/eurheartj/ehab368.CrossRefGoogle ScholarPubMed
McNally, EM and Mestroni, L (2017) Dilated cardiomyopathy: Genetic determinants and mechanisms. Circulation Research 121(7), 731748. https://doi.org/10.1161/CIRCRESAHA.116.309396.CrossRefGoogle ScholarPubMed
Merlo, M, Gagno, G, Baritussio, A, Bauce, B, Biagini, E, Canepa, M, Cipriani, A, Castelletti, S, Dellegrottaglie, S, Guaricci, AI, Imazio, M, Limongelli, G, Musumeci, MB, Parisi, V, Pica, S, Pontone, G, Todiere, G, Torlasco, C, Basso, C, Sinagra, G, Filardi, PP, Indolfi, C, Autore, C and Barison, A (2023) Clinical application of CMR in cardiomyopathies: Evolving concepts and techniques: A position paper of myocardial and pericardial diseases and cardiac magnetic resonance working groups of Italian society of cardiology. Heart Failure Reviews 28(1), 7795. https://doi.org/10.1007/s10741-022-10235-9.CrossRefGoogle ScholarPubMed
Mortensen, SA, Rosenfeldt, F, Kumar, A, Dolliner, P, Filipiak, KJ, Pella, D, Alehagen, U, Steurer, G, Littarru, GP and Investigators, QSS (2014) The effect of coenzyme Q10 on morbidity and mortality in chronic heart failure: Results from Q-SYMBIO: A randomized double-blind trial. JACC Heart Failure 2(6), 641649. https://doi.org/10.1016/j.jchf.2014.06.008.CrossRefGoogle ScholarPubMed
Muchir, A, Wu, W, Choi, JC, Iwata, S, Morrow, J, Homma, S and Worman, HJ (2012) Abnormal p38alpha mitogen-activated protein kinase signaling in dilated cardiomyopathy caused by Lamin a/C gene mutation. Human Molecular Genetics 21(19), 43254333. https://doi.org/10.1093/hmg/dds265.CrossRefGoogle ScholarPubMed
Nagaraju, CK, Robinson, EL, Abdesselem, M, Trenson, S, Dries, E, Gilbert, G, Janssens, S, Van Cleemput, J, Rega, F, Meyns, B, Roderick, HL, Driesen, RB and Sipido, KR (2019) Myofibroblast phenotype and reversibility of fibrosis in patients with end-stage heart failure. Journal of the American College of Cardiology 73(18), 22672282. https://doi.org/10.1016/j.jacc.2019.02.049.CrossRefGoogle ScholarPubMed
Neubauer, S, Horn, M, Cramer, M, Harre, K, Newell, JB, Peters, W, Pabst, T, Ertl, G, Hahn, D, Ingwall, JS and Kochsiek, K (1997) Myocardial phosphocreatine-to-ATP ratio is a predictor of mortality in patients with dilated cardiomyopathy. Circulation 96(7), 21902196.CrossRefGoogle ScholarPubMed
Nielles-Vallespin, S, Khalique, Z, Ferreira, PF, de Silva, R, Scott, AD, Kilner, P, McGill, LA, Giannakidis, A, Gatehouse, PD, Ennis, D, Aliotta, E, Al-Khalil, M, Kellman, P, Mazilu, D, Balaban, RS, Firmin, DN, Arai, AE and Pennell, DJ (2017) Assessment of myocardial microstructural dynamics by in vivo diffusion tensor cardiac magnetic resonance. Journal of the American College of Cardiology 69(6), 661676. https://doi.org/10.1016/j.jacc.2016.11.051.CrossRefGoogle ScholarPubMed
Pelliccia, A, Solberg, EE, Papadakis, M, Adami, PE, Biffi, A, Caselli, S, La Gerche, A, Niebauer, J, Pressler, A, Schmied, CM, Serratosa, L, Halle, M, Van Buuren, F, Borjesson, M, Carre, F, Panhuyzen-Goedkoop, NM, Heidbuchel, H, Olivotto, I, Corrado, D, Sinagra, G and Sharma, S (2019) Recommendations for participation in competitive and leisure time sport in athletes with cardiomyopathies, myocarditis, and pericarditis: Position statement of the sport cardiology section of the European Association of Preventive Cardiology (EAPC). European Heart Journal 40(1), 1933. https://doi.org/10.1093/eurheartj/ehy730.CrossRefGoogle ScholarPubMed
Peters, S, Thompson, BA, Perrin, M, James, P, Zentner, D, Kalman, JM, Vandenberg, JI and Fatkin, D (2022) Arrhythmic phenotypes are a defining feature of dilated cardiomyopathy-associated SCN5A variants: A systematic review. Circulation: Genomic and Precision Medicine 15(1), e003432. https://doi.org/10.1161/CIRCGEN.121.003432.Google ScholarPubMed
Piek, A, de Boer, RA and Sillje, HH (2016) The fibrosis-cell death axis in heart failure. Heart Failure Reviews 21(2), 199211. https://doi.org/10.1007/s10741-016-9536-9.CrossRefGoogle ScholarPubMed
Pinto, YM, Elliott, PM, Arbustini, E, Adler, Y, Anastasakis, A, Bohm, M, Duboc, D, Gimeno, J, de Groote, P, Imazio, M, Heymans, S, Klingel, K, Komajda, M, Limongelli, G, Linhart, A, Mogensen, J, Moon, J, Pieper, PG, Seferovic, PM, Schueler, S, Zamorano, JL, Caforio, AL and Charron, P (2016) Proposal for a revised definition of dilated cardiomyopathy, hypokinetic non-dilated cardiomyopathy, and its implications for clinical practice: A position statement of the ESC working group on myocardial and pericardial diseases. European Heart Journal 37(23), 18501858. https://doi.org/10.1093/eurheartj/ehv727.CrossRefGoogle Scholar
Pirruccello, JP, Bick, A, Wang, M, Chaffin, M, Friedman, S, Yao, J, Guo, X, Venkatesh, BA, Taylor, KD, Post, WS, Rich, S, Lima, JAC, Rotter, JI, Philippakis, A, Lubitz, SA, Ellinor, PT, Khera, AV, Kathiresan, S and Aragam, KG (2020) Analysis of cardiac magnetic resonance imaging in 36,000 individuals yields genetic insights into dilated cardiomyopathy. Nature Communications 11(1), 2254. https://doi.org/10.1038/s41467-020-15823-7.CrossRefGoogle Scholar
Prondzynski, M, Kramer, E, Laufer, SD, Shibamiya, A, Pless, O, Flenner, F, Muller, OJ, Munch, J, Redwood, C, Hansen, A, Patten, M, Eschenhagen, T, Mearini, G and Carrier, L (2017) Evaluation of MYBPC3 trans-splicing and gene replacement as therapeutic options in human iPSC-derived Cardiomyocytes. Molecular Therapy - Nucleic Acids 7, 475486. https://doi.org/10.1016/j.omtn.2017.05.008.CrossRefGoogle ScholarPubMed
Raafs, AG, Verdonschot, JAJ, Henkens, M, Adriaans, BP, Wang, P, Derks, K, Abdul Hamid, MA, Knackstedt, C, van Empel, VPM, Diez, J, Brunner-La Rocca, HP, Brunner, HG, Gonzalez, A, Bekkers, S, Heymans, SRB and Hazebroek, MR (2021) The combination of carboxy-terminal propeptide of procollagen type I blood levels and late gadolinium enhancement at cardiac magnetic resonance provides additional prognostic information in idiopathic dilated cardiomyopathy - A multilevel assessment of myocardial fibrosis in dilated cardiomyopathy. European Journal of Heart Failure 23(6), 933944. https://doi.org/10.1002/ejhf.2201.CrossRefGoogle ScholarPubMed
Reichart, D, Lindberg, EL, Maatz, H, Miranda, AMA, Viveiros, A, Shvetsov, N, Gartner, A, Nadelmann, ER, Lee, M, Kanemaru, K, Ruiz-Orera, J, Strohmenger, V, DeLaughter, DM, Patone, G, Zhang, H, Woehler, A, Lippert, C, Kim, Y, Adami, E, Gorham, JM, Barnett, SN, Brown, K, Buchan, RJ, Chowdhury, RA, Constantinou, C, Cranley, J, Felkin, LE, Fox, H, Ghauri, A, Gummert, J, Kanda, M, Li, R, Mach, L, McDonough, B, Samari, S, Shahriaran, F, Yapp, C, Stanasiuk, C, Theotokis, PI, Theis, FJ, van den Bogaerdt, A, Wakimoto, H, Ware, JS, Worth, CL, Barton, PJR, Lee, YA, Teichmann, SA, Milting, H, Noseda, M, Oudit, GY, Heinig, M, Seidman, JG, Hubner, N and Seidman, CE (2022) Pathogenic variants damage cell composition and single cell transcription in cardiomyopathies. Science 377(6606), eabo1984. https://doi.org/10.1126/science.abo1984.CrossRefGoogle ScholarPubMed
Reichart, D, Magnussen, C, Zeller, T and Blankenberg, S (2019) Dilated cardiomyopathy: From epidemiologic to genetic phenotypes: A translational review of current literature. Journal of Internal Medicine 286(4), 362372. https://doi.org/10.1111/joim.12944.CrossRefGoogle ScholarPubMed
Romano, R, Ghahremani, S, Zimmerman, T, Legere, N, Thakar, K, Ladha, FA, Pettinato, AM and Hinson, JT (2022) Reading frame repair of TTN truncation variants restores Titin quantity and functions. Circulation 145(3), 194205. https://doi.org/10.1161/CIRCULATIONAHA.120.049997.CrossRefGoogle ScholarPubMed
Roura, S and Bayes-Genis, A (2009) Vascular dysfunction in idiopathic dilated cardiomyopathy. Nature Reviews. Cardiology 6(9), 590598. https://doi.org/10.1038/nrcardio.2009.130.CrossRefGoogle ScholarPubMed
Rubis, P, Dziewiecka, E, Wisniowska-Smialek, S, Banys, P, Urbanczyk-Zawadzka, M, Krupinski, M, Mielnik, M, Karabinowska, A and Garlitski, A (2022) Natural history of myocardial fibrosis in dilated cardiomyopathy. European Heart Journal 43(Supplement_2), ehac544.1749. https://doi.org/10.1093/eurheartj/ehac544.1749.CrossRefGoogle Scholar
Sacchetto, C, Sequeira, V, Bertero, E, Dudek, J, Maack, C and Calore, M (2019) Metabolic alterations in inherited cardiomyopathies. Journal of Clinical Medicine 8(12), 2195. https://doi.org/10.3390/jcm8122195.CrossRefGoogle ScholarPubMed
Schafer, S, de Marvao, A, Adami, E, Fiedler, LR, Ng, B, Khin, E, Rackham, OJ, van Heesch, S, Pua, CJ, Kui, M, Walsh, R, Tayal, U, Prasad, SK, Dawes, TJ, Ko, NS, Sim, D, Chan, LL, Chin, CW, Mazzarotto, F, Barton, PJ, Kreuchwig, F, de Kleijn, DP, Totman, T, Biffi, C, Tee, N, Rueckert, D, Schneider, V, Faber, A, Regitz-Zagrosek, V, Seidman, JG, Seidman, CE, Linke, WA, Kovalik, JP, O’Regan, D, Ware, JS, Hubner, N and Cook, SA (2016) Titin-truncating variants affect heart function in disease cohorts and the general population. Nature Genetics 49, 4653. https://doi.org/10.1038/ng.3719.CrossRefGoogle ScholarPubMed
Schultheiss, H-P, Fairweather, D, Caforio, ALP, Escher, F, Hershberger, RE, Lipshultz, SE, Liu, PP, Matsumori, A, Mazzanti, A, McMurray, J and Priori, SG (2019) Dilated cardiomyopathy. Nature Reviews Disease Primers 5(1), 32. https://doi.org/10.1038/s41572-019-0084-1.CrossRefGoogle ScholarPubMed
Shah, SJ, Katz, DH, Selvaraj, S, Burke, MA, Yancy, CW, Gheorghiade, M, Bonow, RO, Huang, CC and Deo, RC (2015) Phenomapping for novel classification of heart failure with preserved ejection fraction. Circulation 131(3), 269279. https://doi.org/10.1161/CIRCULATIONAHA.114.010637.CrossRefGoogle ScholarPubMed
Sharma, UC, Pokharel, S, van Brakel, TJ, van Berlo, JH, Cleutjens, JP, Schroen, B, Andre, S, Crijns, HJ, Gabius, HJ, Maessen, J and Pinto, YM (2004) Galectin-3 marks activated macrophages in failure-prone hypertrophied hearts and contributes to cardiac dysfunction. Circulation 110(19), 31213128. https://doi.org/10.1161/01.CIR.0000147181.65298.4D.CrossRefGoogle ScholarPubMed
Stoll, VM, Clarke, WT, Levelt, E, Liu, A, Myerson, SG, Robson, MD, Neubauer, S and Rodgers, CT (2016) Dilated cardiomyopathy: Phosphorus 31 MR spectroscopy at 7 T. Radiology 281(2), 409417. https://doi.org/10.1148/radiol.2016152629.CrossRefGoogle Scholar
Suresh, A, Martens, P and Tang, WHW (2022) Biomarkers for myocarditis and inflammatory cardiomyopathy. Current Heart Failure Reports 19(5), 346355. https://doi.org/10.1007/s11897-022-00569-8.CrossRefGoogle ScholarPubMed
Sweeney, M, Corden, B and Cook, SA (2020) Targeting cardiac fibrosis in heart failure with preserved ejection fraction: Mirage or miracle? EMBO Molecular Medicine 12(10), e10865. https://doi.org/10.15252/emmm.201910865.CrossRefGoogle ScholarPubMed
Tadros, R, Francis, C, Xu, X, Vermeer, AMC, Harper, AR, Huurman, R, Kelu Bisabu, K, Walsh, R, Hoorntje, ET, Te Rijdt, WP, Buchan, RJ, van Velzen, HG, van Slegtenhorst, MA, Vermeulen, JM, Offerhaus, JA, Bai, W, de Marvao, A, Lahrouchi, N, Beekman, L, Karper, JC, Veldink, JH, Kayvanpour, E, Pantazis, A, Baksi, AJ, Whiffin, N, Mazzarotto, F, Sloane, G, Suzuki, H, Schneider-Luftman, D, Elliott, P, Richard, P, Ader, F, Villard, E, Lichtner, P, Meitinger, T, Tanck, MWT, van Tintelen, JP, Thain, A, McCarty, D, Hegele, RA, Roberts, JD, Amyot, J, Dube, MP, Cadrin-Tourigny, J, Giraldeau, G, L’Allier, PL, Garceau, P, Tardif, JC, Boekholdt, SM, Lumbers, RT, Asselbergs, FW, Barton, PJR, Cook, SA, Prasad, SK, O’Regan, DP, van der Velden, J, Verweij, KJH, Talajic, M, Lettre, G, Pinto, YM, Meder, B, Charron, P, de Boer, RA, Christiaans, I, Michels, M, Wilde, AAM, Watkins, H, Matthews, PM, Ware, JS and Bezzina, CR (2021) Shared genetic pathways contribute to risk of hypertrophic and dilated cardiomyopathies with opposite directions of effect. Nature Genetics 53(2), 128134. https://doi.org/10.1038/s41588-020-00762-2.CrossRefGoogle ScholarPubMed
Tayal, U, Gregson, J, Buchan, R, Whiffin, N, Halliday, BP, Lota, A, Roberts, AM, Baksi, AJ, Voges, I, Jarman, JWE, Baruah, R, Frenneaux, M, Cleland, JGF, Barton, P, Pennell, DJ, Ware, JS, Cook, SA and Prasad, SK (2022) Moderate excess alcohol consumption and adverse cardiac remodelling in dilated cardiomyopathy. Heart 108(8), 619625. https://doi.org/10.1136/heartjnl-2021-319418.CrossRefGoogle ScholarPubMed
Tayal, U and Prasad, SK (2018) Titin cardiomyopathy: Why we need to go big to understand the giant. European Heart Journal 39(10), 874875. https://doi.org/10.1093/eurheartj/ehy109.CrossRefGoogle ScholarPubMed
Tayal, U, Verdonschot, JAJ, Hazebroek, MR, Howard, J, Gregson, J, Newsome, S, Gulati, A, Pua, CJ, Halliday, BP, Lota, AS, Buchan, RJ, Whiffin, N, Kanapeckaite, L, Baruah, R, Jarman, JWE, O’Regan, DP, Barton, PJR, Ware, JS, Pennell, DJ, Adriaans, BP, Bekkers, S, Donovan, J, Frenneaux, M, Cooper, LT, Januzzi, JL, Cleland, JGF, Cook, SA, Deo, RC, Heymans, SRB and Prasad, SK (2022) Precision phenotyping of dilated cardiomyopathy using multidimensional data. Journal of the American College of Cardiology 79(22), 22192232. https://doi.org/10.1016/j.jacc.2022.03.375.CrossRefGoogle ScholarPubMed
Tayal, U, Ware, JS, Lakdawala, NK, Heymans, S and Prasad, SK (2021) Understanding the genetics of adult-onset dilated cardiomyopathy: What a clinician needs to know. European Heart Journal 42(24), 23842396. https://doi.org/10.1093/eurheartj/ehab286.CrossRefGoogle ScholarPubMed
Teerlink, JR, Diaz, R, Felker, GM, McMurray, JJV, Metra, M, Solomon, SD, Adams, KF, Anand, I, Arias-Mendoza, A, Biering-Sorensen, T, Bohm, M, Bonderman, D, Cleland, JGF, Corbalan, R, Crespo-Leiro, MG, Dahlstrom, U, Echeverria, LE, Fang, JC, Filippatos, G, Fonseca, C, Goncalvesova, E, Goudev, AR, Howlett, JG, Lanfear, DE, Li, J, Lund, M, Macdonald, P, Mareev, V, Momomura, SI, O’Meara, E, Parkhomenko, A, Ponikowski, P, Ramires, FJA, Serpytis, P, Sliwa, K, Spinar, J, Suter, TM, Tomcsanyi, J, Vandekerckhove, H, Vinereanu, D, Voors, AA, Yilmaz, MB, Zannad, F, Sharpsten, L, Legg, JC, Varin, C, Honarpour, N, Abbasi, SA, Malik, FI, Kurtz, CE and GALACTIC-HF Investigators (2021) Cardiac myosin activation with Omecamtiv Mecarbil in systolic heart failure. The New England Journal of Medicine 384(2), 105116. https://doi.org/10.1056/NEJMoa2025797.CrossRefGoogle ScholarPubMed
Tuunanen, H, Engblom, E, Naum, A, Nagren, K, Scheinin, M, Hesse, B, Juhani Airaksinen, KE, Nuutila, P, Iozzo, P, Ukkonen, H, Opie, LH and Knuuti, J (2008) Trimetazidine, a metabolic modulator, has cardiac and extracardiac benefits in idiopathic dilated cardiomyopathy. Circulation 118(12), 12501258. https://doi.org/10.1161/CIRCULATIONAHA.108.778019.CrossRefGoogle ScholarPubMed
Vaduganathan, M, Claggett, BL, Jhund, PS, Cunningham, JW, Pedro Ferreira, J, Zannad, F, Packer, M, Fonarow, GC, McMurray, JJV and Solomon, SD (2020) Estimating lifetime benefits of comprehensive disease-modifying pharmacological therapies in patients with heart failure with reduced ejection fraction: A comparative analysis of three randomised controlled trials. Lancet 396(10244), 121128. https://doi.org/10.1016/S0140-6736(20)30748-0.CrossRefGoogle ScholarPubMed
Verdonschot, JAJ, Hazebroek, MR, Derks, KWJ, Barandiaran Aizpurua, A, Merken, JJ, Wang, P, Bierau, J, van den Wijngaard, A, Schalla, SM, Abdul Hamid, MA, van Bilsen, M, van Empel, VPM, Knackstedt, C, Brunner-La Rocca, HP, Brunner, HG, Krapels, IPC and Heymans, SRB (2018) Titin cardiomyopathy leads to altered mitochondrial energetics, increased fibrosis and long-term life-threatening arrhythmias. European Heart Journal 39(10), 864873. https://doi.org/10.1093/eurheartj/ehx808.CrossRefGoogle ScholarPubMed
Verdonschot, JAJ, Hazebroek, MR, Ware, JS, Prasad, SK and Heymans, SRB (2019) Role of targeted therapy in dilated cardiomyopathy: The challenging road toward a personalized approach. Journal of the American Heart Association 8(11), e012514. https://doi.org/10.1161/JAHA.119.012514.CrossRefGoogle ScholarPubMed
Verdonschot, JAJ, Merlo, M, Dominguez, F, Wang, P, Henkens, M, Adriaens, ME, Hazebroek, MR, Mase, M, Escobar, LE, Cobas-Paz, R, Derks, KWJ, van den Wijngaard, A, Krapels, IPC, Brunner, HG, Sinagra, G, Garcia-Pavia, P and Heymans, SRB (2020) Phenotypic clustering of dilated cardiomyopathy patients highlights important pathophysiological differences. European Heart Journal 42, 162. https://doi.org/10.1093/eurheartj/ehaa841.CrossRefGoogle Scholar
Voors, AA, Tamby, JF, Cleland, JG, Koren, M, Forgosh, LB, Gupta, D, Lund, LH, Camacho, A, Karra, R, Swart, HP, Pellicori, P, Wagner, F, Hershberger, RE, Prasad, N, Anderson, R, Anto, A, Bell, K, Edelberg, JM, Fang, L, Henze, M, Kelly, C, Kurio, G, Li, W, Wells, K, Yang, C, Teichman, SL, Del Rio, CL and Solomon, SD (2020) Effects of danicamtiv, a novel cardiac myosin activator, in heart failure with reduced ejection fraction: Experimental data and clinical results from a phase 2a trial. European Journal of Heart Failure 22(9), 16491658. https://doi.org/10.1002/ejhf.1933.CrossRefGoogle ScholarPubMed
Ware, JS, Amor-Salamanca, A, Tayal, U, Govind, R, Serrano, I, Salazar-Mendiguchia, J, Garcia-Pinilla, JM, Pascual-Figal, DA, Nunez, J, Guzzo-Merello, G, Gonzalez-Vioque, E, Bardaji, A, Manito, N, Lopez-Garrido, MA, Padron-Barthe, L, Edwards, E, Whiffin, N, Walsh, R, Buchan, RJ, Midwinter, W, Wilk, A, Prasad, S, Pantazis, A, Baski, J, O’Regan, DP, Alonso-Pulpon, L, Cook, SA, Lara-Pezzi, E, Barton, PJ and Garcia-Pavia, P (2018) Genetic etiology for alcohol-induced cardiac toxicity. Journal of the American College of Cardiology 71(20), 22932302. https://doi.org/10.1016/j.jacc.2018.03.462.CrossRefGoogle ScholarPubMed
Ware, JS and Cook, SA (2018) Role of titin in cardiomyopathy: From DNA variants to patient stratification. Nature Reviews. Cardiology 15(4), 241252. https://doi.org/10.1038/nrcardio.2017.190.CrossRefGoogle ScholarPubMed
Ware, JS, Li, J, Mazaika, E, Yasso, CM, DeSouza, T, Cappola, TP, Tsai, EJ, Hilfiker-Kleiner, D, Kamiya, CA, Mazzarotto, F, Cook, SA, Halder, I, Prasad, SK, Pisarcik, J, Hanley-Yanez, K, Alharethi, R, Damp, J, Hsich, E, Elkayam, U, Sheppard, R, Kealey, A, Alexis, J, Ramani, G, Safirstein, J, Boehmer, J, Pauly, DF, Wittstein, IS, Thohan, V, Zucker, MJ, Liu, P, Gorcsan, J III, McNamara, DM, Seidman, CE, Seidman, JG, Arany, Z and Imac and Investigators, I (2016) Shared genetic predisposition in peripartum and dilated cardiomyopathies. The New England Journal of Medicine 374(3), 233241. https://doi.org/10.1056/NEJMoa1505517.CrossRefGoogle ScholarPubMed
Watson, WD, Green, PG, Lewis, AJM, Arvidsson, P, De Maria, GL, Arheden, H, Heiberg, E, Clarke, WT, Rodgers, CT, Valkovic, L, Neubauer, S, Herring, N and Rider, OJ (2023) Retained metabolic flexibility of the failing human heart. Circulation 148, 109. https://doi.org/10.1161/CIRCULATIONAHA.122.062166.CrossRefGoogle ScholarPubMed
Wu, W, Muchir, A, Shan, J, Bonne, G and Worman, HJ (2011) Mitogen-activated protein kinase inhibitors improve heart function and prevent fibrosis in cardiomyopathy caused by mutation in Lamin a/C gene. Circulation 123(1), 5361. https://doi.org/10.1161/CIRCULATIONAHA.110.970673.CrossRefGoogle ScholarPubMed
Yancy, CW, Jessup, M, Bozkurt, B, Butler, J, Casey, DE, Drazner, MH, Fonarow, GC, Geraci, SA, Horwich, T, Januzzi, JL, Johnson, MR, Kasper, EK, Levy, WC, Masoudi, FA, McBride, PE, McMurray, JJ, Mitchell, JE, Peterson, PN, Riegel, B, Sam, F, Stevenson, LW, Tang, WH, Tsai, EJ, Wilkoff, BL, American College of Cardiology Foundation and American Heart Association Task Force on Practice Guidelines (2013) 2013 ACCF/AHA guideline for the management of heart failure: A report of the American College of Cardiology Foundation/American Heart Association task force on practice guidelines. Journal of the American College of Cardiology 62(16), e147e239. https://doi.org/10.1016/j.jacc.2013.05.019.CrossRefGoogle Scholar
Zeppenfeld, K, Tfelt-Hansen, J, de Riva, M, Winkel, BG, Behr, ER, Blom, NA, Charron, P, Corrado, D, Dagres, N, de Chillou, C, Eckardt, L, Friede, T, Haugaa, KH, Hocini, M, Lambiase, PD, Marijon, E, Merino, JL, Peichl, P, Priori, SG, Reichlin, T, Schulz-Menger, J, Sticherling, C, Tzeis, S, Verstrael, A, Volterrani, M and Group, ESCSD (2022) 2022 ESC guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death. European Heart Journal 43(40), 39974126. https://doi.org/10.1093/eurheartj/ehac262.CrossRefGoogle ScholarPubMed
Zhang, L, Lu, Y, Jiang, H, Zhang, L, Sun, A, Zou, Y and Ge, J (2012) Additional use of trimetazidine in patients with chronic heart failure: A meta-analysis. Journal of the American College of Cardiology 59(10), 913922. https://doi.org/10.1016/j.jacc.2011.11.027.CrossRefGoogle ScholarPubMed
Zhou, J, Ng, B, Ko, NSJ, Fiedler, LR, Khin, E, Lim, A, Sahib, NE, Wu, Y, Chothani, SP, Schafer, S, Bay, BH, Sinha, RA, Cook, SA and Yen, PM (2019) Titin truncations lead to impaired cardiomyocyte autophagy and mitochondrial function in vivo. Human Molecular Genetics 28(12), 19711981. https://doi.org/10.1093/hmg/ddz033.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Dilated cardiomyopathy (DCM) – a family of diseases. Selected syndromic causes of dilated cardiomyopathy include Barth syndrome, haemochromatosis, Kearns–Sayre syndrome, and Carvajal syndrome (adapted with permission from Halliday, 2022).

Figure 1

Figure 2. Precision therapies for genotype-positive, phenotype-negative (G+ P−) individuals would likely involve genotype-specific therapies, and lifestyle interventions. Treatments that could be introduced at an early disease stage include anti-fibrotic agents, and therapies to target cardiac metabolism (such as SGLT2 inhibitors) whereas advanced disease therapies include antiarrhythmics for those at the highest risk, ICD therapy and guideline-directed heart failure therapy (GDMT) (HF, heart failure; LVSD, left ventricular systolic dysfunction).

Figure 2

Table 1. Genes with definite/strong association with DCM and their functional and phenotypic implications (14, 17)

Figure 3

Figure 3. Cellular locations of some of the proteins with their respective genes associated with dilated cardiomyopathy.

Figure 4

Figure 4. Precision medicine for dilated cardiomyopathy.

Author comment: Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? — R0/PR1

Comments

Dear Professor Dominiczak

Many thanks for asking to contirbute a review on precision therapy for dilated cardiomyopathy. We believe this is an exciting, dynamic topic that holds great potential to improve the outcomes of our patients. We have focused our review on the areas that hold the greatest potential for immediate translation including the integration of data from advanced imaging, genomics and biomarkers to provide insight into specific disease drivers that may be targeted by genotype-specific therapies or mechanism-based therapies for those with gene elusive disease.

We believe this will be of interest to your readership.

Many thanks and best wishes

Brian Halliday

Review: Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? — R0/PR2

Conflict of interest statement

Reviewer declares none.

Comments

Javed and Halliday presented a very insightful, informative and well balanced review on the current knowledge on background and potential methods of precision therapy in dilated cardiomyopathy (DCM). Indeed, a very interesting, well composed approach.

Whether and what more to take into consideration?

In the paragraph 2, Authors omitted autosomal recessive transmission that may occur, especially in younger more severe affected subjects, both with homozygotes (like NRAP) and compound heterozygotes. Rare metabolic causes, e.g. type IV glycogenosis, especially in pediatric/adolescent population could also be taken into consideration. Furthermore, syndromic forms, with other striated muscle disease, related mainly to dystrophinopathies could be mentioned, and the role of CK assessment, helpful in the diagnostic proces, could be stressed.

Apart from additional role of common genetic variation in determining the risk of developing DCM, it seems relevant to unveil the role of susceptibility variants, often marked as VUS-es in known genes associated with DCM, e.g. RBM20. As it is known in TAAD patients ( PMID: 29961567), similarly VUSes in the genes associated with DCM can be low penetrant „risk variants”, in particular in the setting of acute onset heart failure.

In the paragraph 3 I wonder whether endothelial dysfunction should not be addressed.

In the paragraph 5 refering to biomarkers, the role of hs troponins in the early stage of cardiomyopathies could be shown (PMID: 32408651).

On page 14 maybe it is not so clear, with regard to Verdonschot publication (2020). Authors identified 4 phenogroups (mild systolic dysfunction, auto immune, cardiac arrhythmia, severe systolic dysfunction) and 3 transcriptomic profiles (pro-inflammatory, pro-fibrotic, metabolic).

Recommendation: Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? — R0/PR3

Comments

Congratulations to the authors. This paper has approached the multi paradigms that are present in the current context of dilated cardiomyopathy, and for the next few years. This theme is wide, but the authors have carried out a dense revision on this limited space to the manuscript. Please consider the following corrections:

In figure 1. There is a blue circle under the “idiopathic circle” which has no title. What does this blue circle represent, in fact? You should describe it in the figure. Further, this figure can be better with an explanation on what “syndromic causes” (cited in the picture) means. You can write about it at the figure footer.

Figure 2: What means G+P- should be described in the figure footer, as well as LVSD, GDMT and HF, although these terms could be identified in the text. You should also cite examples of what precision therapies can be applied in each phase of the disease progression.

Page 7, line 21: If a causative genetic variant is identified, cascade screening will be able to identify the 50% of relatives who are carriers and who have an elevated risk of developing disease.

I think that you are considering 50% the risk of heritable transmission associated with autosomal dominant disease; however, in some pedigrees, all the offspring (100%) or nobody (0%) have inherited the causal variant. This comment seems to be also wrong in X-linked, recessive or mitochondrial etiologies where 50% is an error. Please, remove the 50% in the sentence.

Page 7, line 45: Nevertheless, subtle markers of reduced cardiac function have been found in carriers in the general population(Schafer et al. 2016), suggesting they may be more susceptible to extrinsic insults.

Please, cite what are the extrinsic insults.

Page 8, line 15. You should explain LVEF, because it was the first time that you cite this abbreviation in the text.

Decision: Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? — R0/PR4

Comments

No accompanying comment.

Author comment: Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? — R1/PR5

Comments

Dear Professor Dominiczak,

Thank you for inviting us to submit the attached review on “Precision therapy in dilated cardiomyopathy: pipedream or paradigm shift?”. We have addressed the comments raised by reviewers and the editor. We hope that this article will be of interest to a broad audience and provides a balanced overview of this broad subject.

Yours faithfully,

Dr Brian Halliday

Recommendation: Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? — R1/PR6

Comments

Dear,

We are very grateful for the modifications made by you authors. However, there is one review that is not correct:

>>> Page 7, line 21: If a causative genetic variant is identified, cascade screening will be able to identify the 50% of relatives who are carriers and who have an elevated risk of developing disease.

>>> I think that you are considering 50% the risk of heritable transmission associated with autosomal dominant disease; however, in some pedigrees, all the offspring (100%) or nobody (0%) have inherited the causal variant. This comment seems to be also wrong in X-linked, recessive or mitochondrial etiologies where 50% is an error. Please remove the 50% in the sentence.

>>> Page 6, line 10. The text has been modified to read: “If a causative rare variant in an autosomal gene is identified, cascade testing will be able to identify 50% of the relatives who are carriers and who have an elevated risk of developing diseases.

As we had mentioned previously. Saying that it is able to identify 50% of relatives who are carriers is a misinterpretation of a dominant autosomal risk. Please include the sentence as follows:

“If a causative genetic variant is identified, cascade screening will be able to identify the 50% of relatives who are carriers and who have an elevated risk of developing disease.”

Best regards

Decision: Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? — R1/PR7

Comments

No accompanying comment.

Author comment: Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? — R2/PR8

Comments

17th October 2023

Prof. Anna Dominiczak

Editor-in-Chief, Cambridge Prisms: Precision Medicine

Dear Prof Dominiczak,

Thank you for inviting us to contribute to your journal. Kindly find attached our revised manuscript titled @Precision therapy in dilated cardiomyopathy: pipedream or paradigm shift?" We hope that this provides a balanced overview of this broad subject which will be of interest to your audience. We have addressed comments raised by editors and reviewers, and look forward to your response.

Yours faithfully,

Dr Brian Halliday

Recommendation: Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? — R2/PR9

Comments

Congratulations on your review manuscript. We hope that it can help physicians and other precision medicine professionals to do the best care to DCM patients.

Best regards

Decision: Precision therapy in dilated cardiomyopathy: Pipedream or paradigm shift? — R2/PR10

Comments

No accompanying comment.