IMR Press / RCM / Volume 25 / Issue 1 / DOI: 10.31083/j.rcm2501005
Open Access Review
Recent Progresses in the Multimodality Imaging Assessment of Myocardial Fibrosis
Show Less
1 Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200127 Shanghai, China
2 Department of Nephrology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200127 Shanghai, China
3 Department of Nephrology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, 310011 Hangzhou, Zhejiang, China
4 State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200127 Shanghai, China
*Correspondence: jiangmeng0919@163.com (Meng Jiang); pujun310@hotmail.com (Jun Pu)
These authors contributed equally.
Rev. Cardiovasc. Med. 2024, 25(1), 5; https://doi.org/10.31083/j.rcm2501005
Submitted: 21 August 2023 | Revised: 20 September 2023 | Accepted: 25 September 2023 | Published: 8 January 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Myocardial fibrosis, a common pathophysiological consequence of various cardiovascular diseases, is characterized by fibroblast activation and excessive deposition of extracellular matrix (ECM) collagen. Accumulating evidence indicates that myocardial fibrosis contributes to ventricular stiffness, systolic and diastolic dysfunction, and ultimately leads to the development of heart failure (HF). Early detection and targeted treatment of myocardial fibrosis is critical to reverse ventricular remodeling and improve clinical outcomes in patients with cardiovascular diseases. However, despite considerable progresses made in understanding molecular mechanisms of myocardial fibrosis, non-invasive imaging to assess myocardial fibrosis and guide clinical treatment is still not widely available, limiting the development of innovative treatment strategies. This review summarizes recent progresses of imaging modalities for detecting myocardial fibrosis, with a focus on nuclear medicine, echocardiography and cardiac magnetic resonance (CMR).

Keywords
myocardial fibrosis
multimodality imaging assessment
nuclear medicine
echocardiography
cardiac magnetic resonance
1. Introduction

Myocardial fibrosis, defined as an excessive accumulation of extracellular matrix (ECM) proteins, results in pathological ventricular remodeling and, eventually leads to heart failure (HF) [1]. Myocardial fibrosis can be divided into several subtypes including: replacement fibrosis, reactive interstitial fibrosis, endomyocardial fibrosis [2] and infiltrative interstitial fibrosis. Reactive interstitial fibrosis is an adaptive, non-specific response distinguished by a scattered microscopic distribution in the myocardium, occasionally accompanied by local peripheral distribution of blood vessels [3], with sustained activation of pro-fibrotic growth factors including transforming growth factor-β (TGF-β) (Fig. 1), fibroblast growth factor-2 and connective tissue growth factor [4]. Such interstitial form of fibrosis is typically secondary to long-term pressure and volume overload, resulting in hyperactive renin-angiotensin-aldosterone system and adrenergic system, as presented in valvular heart disease [5], chronic hypertension [6], and cardiomyopathies such as hypertrophic cardiomyopathy [7], dilated cardiomyopathy [8] and diabetic cardiomyopathy [7], but also in distal non-infarcted myocardium following myocardial infarction [9]. Early mild replacement interstitial fibrosis is reversible with specific treatment [10]. Infiltrative myocardial fibrosis is caused by excessive storage of misfolded, insoluble, aggregated proteins (amyloidosis) [11] or globotriaosylceramide (Fabry disease) [12] in the extracellular matrix. Replacement myocardial fibrosis often occurs after acute myocardial infarction (AMI), where necrotic myocardial cells are replaced by collagen fibers, forming fibrous cardiac scar, which ensures the integrity of the heart from rupture in the early stages of myocardial infarction [13]. On the other hand, if left untreated and overburdened post-AMI, the fibrotic tissue can spread to the non-infarcted myocardium, resulting in decreased tissue compliance and cardiac dysfunction [14]. In addition, the excessive deposition of ECM damages the mechanical-electrical coupling of myocytes, impairing myocardial contractility and raising the incidence of malignant arrhythmias and sudden death [15]. In addition, epidemiological studies and clinical trials have shown that myocardial fibrosis is an independent risk factor of adverse cardiac events such as AMI, HF, arrhythmia and cardiovascular death [7].

Fig. 1.

Cellular process of myocardial fibrosis after myocardial infarction. Repairing macrophages are recruited to engulf apoptotic neutrophils, and release inhibitory transmitters such as transforming growth factor-β and other potent inflammatory inhibitors. These factors can induce the activation of resting fibroblasts with higher expression of fibroblast activation protein (FAP), and trigger the profibrotic process. While cardiac scar tissue maintains the structural integrity and pressure-generating capacity, persistent myocardial fibrosis leads to adverse changes in the structure and compliance of the ventricles, resulting in the progression of heart failure (HF).

Myocardial fibrosis can be detected by a variety of methods in clinical practice. Traditionally, endomyocardial biopsy is the gold standard for determining myocardial fibrosis, despite its invasive and inconvenient properties. In addition, the diagnosis of myocardial fibrosis by endomyocardial biopsy can be challenging due to its low diagnostic yield, especially for diffuse myocardial fibrosis [16, 17]. In the past, imaging examination such as electrocardiogram and echocardiography were applied to observe cardiac electrical conduction, cardiac structure, and function. In recent years, novel imaging techniques have provided more evidence for determining the characteristics of myocardial tissue, such as single-photon emission computed tomography (SPECT) and cardiac magnetic resonance (CMR). Different imaging tests have their unique features. Since multimodality imaging plays an important role in the initial assessment and diagnosis of myocardial fibrosis, here we discuss current available noninvasive imaging techniques and their values in guiding clinical treatment and improving patient outcomes.

2. Echocardiography

Echocardiography, based on the principle of ultrasonic ranging, is a preferred non-invasive technique to examine the anatomical structure and function of the heart and great vessels [18]. Echocardiography has outstanding advantages such as convenience, rapidity, and non-invasively bedside use. Fibrosis can be hinted when structural and functional changes such as abnormal myocardial thickening, and systolic or diastolic dysfunction are observed [19]. The strategy of integrated backscatter analysis (IB) in standard 2-dimensional (2D) ultrasound images is the first attempt for noninvasive evaluation of myocardial fibrosis after infarction using echocardiography [20]. It measures two parameters of ultrasonic tissue characterization: the amplitude of the cardiac cycle-dependent variation of the backscatter integral signal (cdv-IB) and the mean value of IB [21]. IB signal calibrated by the backscatter power from the pericardium. Moreover, in patients with dilated or hypertrophic cardiomyopathy, m-IB during a cardiac cycle was reported to correlate with the severity of myocardial fibrosis [22]. The intensity of septal IB signal increases in patients with hypertrophic cardiomyopathy (HCM). As a marker of interstitial fibrosis, it is associated with a progressive increase in Doppler parameters related to ventricular stiffness such as pulmonary venous backward velocities and mitral peak velocity at atrial contraction [23]. Losi and colleagues [23] further showed that in HCM patients, the occurrence of ventricular tachyarrhythmias was significantly associated with higher IB signal rather than septal thickness. In addition, echocardiographic measurements based on backscatter techniques include signal intensity coefficient (SIC), which utilizes the greyscale signal intensity values generated at the myocardium-pericardium interface resulting from interactions between the ultrasound signal and myocardial tissue [24]. SIC produces measurable differences between diseased and healthy myocardium. In populations carrying genetic variants associated with HCM, SIC values significantly correlate with left ventricular (LV) hypertrophy [24]. However, this observation is not applicable in patients with coronary artery disease. Higher calibrated integrated backscatter (cIB) was not confirmed as a marker of increased myocardial fibrosis, but was associated with higher soluble vascular endothelial growth factor receptor-1 (sVEGFR-1) and soluble receptor for advanced glycation end products (RAGE) plasma levels. Meanwhile, correlation between cIB and myocardial fibrosis has not been proven by histological examination and CMR evaluation [25, 26].

2.1 Pulse-Cancellation Echocardiography

In 2016, Dr. Gaibazzi and colleagues [19, 27] attempted to identify myocardial scar or fibrotic areas using “echocardiographic scar” (eScar). This technique combined 2D ultrasound imaging with multipulse modulation and inversion to achieve a higher spatial and temporal resolution than 3D imaging. Compared with standard harmonic imaging, eScar is designed to distinguish scars from normal myocardium. Using the CMR-late gadolinium enhancement (LGE) technique as reference, eScar has been proven to be able to identify the presence and location of cardiac scars in patients with ST-elevation myocardial infarction (STEMI) [19]. While, sensitivity in apical myocardial segments, quality of image, and gain dependence are still noteworthy problems for eScar echocardiography [19]. Nevertheless, eScar has been applied in the prediction of appropriate implantable cardioverter-defibrillator (ICD) shocks in patients after myocardial infarction [28]. Intriguingly, eScar also shows the ability to assess subclinical myocardial involvement and predict disease activity in patients with systemic lupus erythematosus (SLE), an autoimmune disease involving multiple systems throughout the body [29]. In this pilot study, eScar identified myocardial scars at the inferoseptal myocardial segments in 19% of SLE patients while in none of the controls. Therefore, as a rapid and inexpensive technique, eScar can be routinely applied in routine clinical practice for cardiac monitoring in patients with multi-organic diseases such as SLE [29].

2.2 Two-Dimensional and Three-Dimensional Speckle Tracking Echocardiography

Myocardial strain including global longitudinal strain (GLS), global circumferential strain (GCS), global radial strain (GRS) and tangential strain (TS) from the scatter-tracking technique have been applied to assess fibrotic myocardium [30]. In general, GLS are recommended as the most sensitive myocardial deformation parameter, which reflects impaired subendocardial fibres [31]. Using CMR as a reference, echographic GLS is significantly related to the estimated degree of fibrosis in patients with HCM [31], and Anderson-Fabry disease [32, 33], and heart transplant recipients [34]. However, similar correlations were not observed between GCS, GRS and myocardial fibrosis.

Patients with advanced heart failure prominent present with right ventricular (RV) enlargement, increased myocardial fibrosis and systolic dysfunction. Myocardial deformation of the RV free wall is one of the most accurate functional indicators and is associated with RV myocardial fibrosis and functional capacity [35]. Longitudinal strain from speckle tracking echocardiography has been proven useful in assessing the severity of right ventricular fibrosis [36].

Novel parameters including mechanical dispersion and myocardial work are able to offer additional possibilities for the evaluation of myocardial fibrosis. Both mechanical dispersion (the standard deviation of the time to peak negative strain in LV segments) and myocardial work (reflects the stroke work of the pressure-strain circuit by combining LV deformation and afterload information) have been reported in pilot studies as stronger predictors of LV myocardial fibrosis compared to GLS [37, 38].

3. Cardiac Magnetic Resonance (CMR)

CMR has become the preferred imaging modality for evaluating myocardial fibrosis due to its ability in soft tissue characterization. T1-weighted images for scar and T2-weighted images for edema visualization are essential sequences to characterize soft tissue [39]. CMR imaging-derived parameters, particularly by LGE and T1 mapping sequences, are widely used to identify fibrotic myocardium. LGE can depict local replacement myocardial fibrosis as seen in large focal post-infarct scars, while T1 mapping has the potential in detecting and quantifying diffuse myocardial fibrosis, since it evaluates the T1 relaxation time of myocardial tissue [40].

3.1 Late Gadolinium Enhancement

LGE is a clinically useful non-invasive CMR sequence for the detection of focal cardiac fibrosis. The reduced density of capillaries in the fibrotic myocardial tissue leads to a higher concentration of the contrast agent retained in the fibrotic region [41]. Graphically, fibrotic tissue was significantly enhanced on LGE images compared to normal myocardial tissue [42]. In patients with myocardial infarction, a delayed contrast enhancement by magnetic resonance imaging (MRI) was recommended to distinguish viable from non-viable myocardium throughout the infarct healing process [43]. Furthermore, a significant correlation was found between LGE and collagen deposition in the myocardial tissue, which is an indirect indication of fibrosis severity as measured by extracellular matrix volume [44, 45].

The use of LGE is rapidly expanding to assess myocardial fibrosis in cardiomyopathies [46]. Several patterns of LGE that are distinct from ischemic cardiomyopathy have been identified. However, these patterns are not specific enough to be used as diagnostic criteria [47]. Around one third of patients with dilated cardiomyopathy presented non-ischemic LGE pattern (mid-lateral or subepicardial), which is also a predictor of adverse cardiovascular events, including heart failure, ventricular arrhythmias, sudden cardiac death (SCD) and all-cause mortality [48]. Patchy fibrosis in the mid-ventricular layer is the typical pattern characterized by LGE in patients with HCM [49]. Epidemiological study has shown that HCM-related myocardial fibrosis is closely related to arrhythmia, and is remarkably associated with subsequent SCD after adjusting for other risk factors [49]. Moreover, a recent meta-analysis demonstrated LGE as the single best imaging marker to predict adverse outcomes in HCM patients [50].

In addition to risk prediction, the severity of myocardial fibrosis assessed by LGE CMR can be used to guide clinical treatment, such as optimization of the timing of ICD implantation [51]. In addition, LGE CMR-based assessment of myocardial fibrosis plays an important prognostic role in aortic stenosis, Eisenmenger’s syndrome, hypertension and diabetes mellitus [52, 53].

Despite increasing applications of LGE CMR, the setting of intensity threshold for cardiac fibrosis by LGE imaging is still not clear in clinical practice [44]. Scarred myocardium is defined as higher signal intensity than normal myocardium in LGE, and official guidelines advocate a threshold of 2-standard deviation (SD) [54]. However, other techniques also can be applied, including the 3, 4, 5, or 6 SD method, manual quantification (mapping the region of interest around the scar), and the full width at half maximum (FWHM) technique that uses half of the maximal signal within the scar as a threshold. LGE volume varied substantially depending on the quantification method used. The 2-SD technique produced a 2-fold higher LGE volume than the FWHM, 6-SD and manual techniques, while the FWHM technique displayed the best reproducibility [55].

Additionally, since the LGE interpretation is based on the difference of contrast agent distribution among tissues, the application in diffuse myocardial fibrosis detection was not feasible. Also, the increased extracellular matrix due to inflammation and edema may lead to interpretation errors in the assessment of fibrotic myocardium [44].

3.2 T1 Mapping

T1 mapping technique has the advantage in detecting diffuse myocardial fibrosis resulting from valvular disease or various cardiomyopathies. In contrast to LGE, T1 mapping does not depend on the contrast between normal and scarred myocardium. It provides a quantitative assessment of the tissue characterization based on a fully quantitative pixel analysis. In combination with hematocrit, these data allowed the quantification of extracellular volume (ECV) to evaluate myocardial fibrosis. ECV fits well with the histological extracellular space. Both T1 mapping and ECV has shown high reproducibility in detecting and quantifying histological collagen volume fractions [56].

Alternative fibrosis often occurs after myocardial infarction, and T1 mapping sequence can dichotomously identify infarct areas as a potential tool for measuring infarct size [57], which showed good agreement between native T1 mapping and LGE imaging modality [58]. In patients with severe aortic valve disease, diffuse myocardial fibrosis assessed by anterior septal-basal ECV correlates with histological myocardial fibrosis. Prolonged T1 value and elevated ECV can also be detected in dilated cardiomyopathy suggesting the presence of myocardial fibrosis occurrence [59]. T1 and ECV in detecting fibrosis have also been studied in hypertrophic cardiomyopathy. Even in the absence local LGE and hemodynamic obstruction, prolonged myocardial T1 and increased ECV suggest diffuse myocardial fibrosis in patients with HCM, which is also associated with left ventricular hypertrophy [60]. Native T1 and ECV quantification show high diagnostic performance for cardiac amyloidosis and can be used as non-invasive markers to assess disease severity and prognosis [61, 62]. The location and pattern of fibrosis favor the separation between healthy and fibrotic myocardium [63] and can distinguish hypertrophic cardiomyopathy from other hypertrophic heart diseases such as hypertensive heart disease [64].

Although CMR is currently the recommended imaging modality for clinical detection of myocardial fibrosis, patients with metal implants or pacemakers are prohibited to undergo CMR examination. Claustrophobic patients who have difficulty in overcoming psychological barriers to accept long time onboard examinations, and patients with congestive heart failure are usually not able to tolerate prolonged lying down. Moreover, normal range of T1 threshold is sensitive to the physical properties of contrast agent, acquisition time, and renal function and hematocrit of patients [44].

4. Computed Tomography (CT)

Recently, animal and clinical studies have demonstrated the feasibility of contrast enhanced CT in detecting fibrosis by CT delayed enhancement (CT-DE). The principle of CT-DE is similar to that of CMR LGE [65]. CT-DE allows quantitative assessment of ECV to evaluate fibrosis. CT-based ECV quantification is effective in assessing myocardial fibrosis, showing a strong correlation with CMR findings. CT-ECV also displayed high diagnostic accuracy in distinguishing LGE-positive from LGE-negative segments [65]. Furthermore, previous study indicated that CT was able to assess myocardial fibrosis in cases where CMR is not available, which still requires verification by further large-scale studies [66]. However, despite excellent specificity, the clinical use of CT-DE is limited by its low sensitivity. The study by Bettencourt et al. [65] showed a sensitivity of 53% and a specificity of 98% in 105 patients with suspected coronary artery disease.

Although higher volume of iodinated contrast agents and lower energies improve spatial resolution, the contrast difference between normal and infarcted myocardium detected by CT-DE is suboptimal compared to CMR [67]. To circumvent this limitation, dual-energy CT improves the characterization of tissue composition and image quality by using an X-ray source that emits 2 different spectra or by employing a 2-layer detector to achieve continuous acquisition of CT in different photon spectra [68].

5. Nuclear Medicine

Nuclear medicine a well-established advanced imaging modality for the diagnosis, and evaluation of cardiovascular disease. The combination of radionuclide imaging with biologically targeted molecules provides unique insight into disease mechanisms at the molecular level, which allows an early detection of damaged myocardium before pathological changes occur.

Myocardial fibrosis is recognized as excessive deposition of collagen. The collagen-targeted contrast agent is the first targeted probe for the detection of myocardial fibrosis after a heart attack. 99mTc-streptavidin-coupled-collagelin and 99mTc-CBP1495 are two collagen-targeting peptide tracers that have relatively high affinity for collagen. Significantly increased uptake of these tracers was observed in fibrotic tissues of rat models [69]. However, collagen-targeted peptides can only show the late products of myocardial fibrosis, and thus they are not sensitive for fibrosis detection at earlier stages of the disease. It is also not possible to determine whether myocardial fibrosis is ongoing. Velikyan et al. [70] recently reported a 68Ga-labelled collagelin analogue, which showed promises for the detection of early active fibrosis by binding to monomeric collagen before the collagen fibres mature. However, there is still plenty of uncertainty for clinical application.

Molecular targets of activated fibroblasts at early disease stages are predictive of the extent and severity of cardiac fibrosis. In a rat model of myocardial fibrosis, angiotensin II (Ang II) was highly expressed in activated macrophages and myofibroblasts. Through acting on Ang II type 1 receptors (At1R), Ang II induced the expression of TGF-β, which is the growth factor most closely associated with the development of tissue fibrosis [71]. Positron emission tomography (PET) experiments using 11C-KR31173 in a porcine myocardial infarction model suggested that the radioactive probe detection of At1R is feasible. The application in human were safe, and showed detectable retention of specific myocardial markers, but at lower levels than that in pigs [72]. Cy5.5-Arg-Gly-Asp (RGD) imaging peptide, a targeting marker for myofibroblasts, can also display interstitial changes in myocardial remodeling and assess fibrosis in response to anti-angiotensin therapy [73]. In the early post-myocardial infarction period, the range of tracer uptake measured by 99Tc-RGD imaging after 3 weeks is comparable to that of CMR imaging. The scar size shown by 99Tc-RGD imaging predicts the eventual scar formation after myocardial infarction.

Fibroblast activation protein (FAP) is expressed at high levels in activated fibroblasts and shows low expression in most normal organs [74]. Radioactively labeled fibroblast activation protein inhibitor (FAPI) is developed to detect activated fibroblasts and initially shows great promise in the diagnosis and treatment of cancer patients. The application of FAPI in cardiovascular disease began with the incidental observation of FAPI in cancer patients by PET. A correlation between tracer uptake and reduced ejection fraction has been observed by FAPI-PET imaging in patients with metastatic cancer [75]. FAPI binds to FAP and accumulates strongly in tissue with high fibroblast activation, showing a high bright signal compared to normal myocardium, with a low background signal. By exploiting the molecular characteristics of myocardial fibrosis, where fibroblasts are highly activated to produce collagen fibers, FAPI can be used as a specific target for the management and treatment of cardiovascular disease. More recently, it has been utilized in murine models and in humans for the assessment of myocardial fibrosis following myocardial infarction (MI) [76, 77, 78]. Serial imaging with 68Ga-FAPI in a MI model established by coronary artery ligation showed intense radiotracer uptake around the infarcted area, and the uptake peaked at day 6 [76]. In the study by Zhang et al. [79] aseline uptake volume (UV) was a powerful predictor of LV remodeling at 1 year after STEMI in 26 patients with ST-segment elevation myocardial infarction (STEMI) who underwent Ga-DOTA-FAPI-04 PET (OR = 1.048, p = 0.011). Lyu et al. [80] found that FAPI imaging was able to detect myocardial fibrosis in diabetic, obese and elderly patients, providing additional evidence for early intervention and clinical decision-making in the management of patients at elevated risk of CVD.

In addition, 68Ga-FAPI PET has been used in a rat model of HF to visualize myocardial fibrosis and monitor HF progression [79]. A study by Guokun Wang et al. [81] showed that fibroblast activation in the heart and liver after pressure overload could be monitored using 68Ga-FAPI-04 PET/CT and that this non-invasive technique was a better predictor of subsequent worsening of heart failure. FAP activity is heterogeneously increased in the myocardium of patients with hypertrophic cardiomyopathy, and their PET-measured FAPI uptake is a potential predictor for 5-year risk of sudden death from cardiovascular causes [82].

However, the cost of test, the worries about radiation, and the poor understanding of nuclear medicine have limited its use clinically. In the future, if these obstacles can be overcome, it will open a new era of targeted treatment and management of patients with myocardial fibrosis.

6. Conclusion and Future Perspective

Early detection and targeted treatment of myocardial fibrosis is essential to improve clinical outcomes in patients with cardiovascular diseases. Multimodality non-invasive imaging approaches can directly or indirectly evaluate the presence and severity of cardiac fibrosis, with advantages and disadvantages of each technique summarized in Fig. 2. In summary, CMR is the gold standard for noninvasive detection and quantification of myocardial fibrosis in clinical practice, whereas other techniques show promises as valuable alternatives. Molecular imaging is developing rapidly and has been a promising technique not only for studying pathological mechanisms, but also for investigating the efficacy of individualized therapeutic regimens to meet the growing need for precision medicine. All the progresses made in the development of novel radiopharmaceuticals targeting specific cardiovascular molecules indicated that the revolution in personalized medicine has only just begun.

Fig. 2.

Multimodality imaging assessment of myocardial fibrosis. The multimodality imaging approaches that are able to assess myocardial fibrosis in clinical practice. LGE, CMR-late gadolinium enhancement; PET, positron emission tomography; MRI, magnetic resonance imaging; ECV, extracellular volume; CT, computed tomography; SPECT, single-photon emission computed tomography.

Author Contributions

HZ, KWX, YYQ, ZGZ, MJ and JP contributed to the conception and design of this review. HZ and KWX prepared the initial draft. ZGZ, YYQ, MJ and JP were involved in manuscript proofreading and critical revisions. MJ and JP provided thorough and comprehensive guidance. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We would like to thank all cardiologists from Renji Hospital, who have devoted their efforts to the diagnosis and treatment of cardiovascular diseases.

Funding

MJ received funding support from the National Natural Science Foundation of China (U21A20341, 81971570); Shanghai Academic/Technology Leader Program (21XD1432100) and Shanghai Science and Technology Commission Program (20Y11910500, 22DZ2292400). JP received funding support from the National Natural Science Foundation of China (81930007); National Key Research and Development Program of China (2022YFC3602400); Shanghai Municipal Health Commission (2023ZZ02021 and GWVI-11.1-26). KWX is supported by the National Natural Science Foundation of China (Grant No. 82000634) and Shanghai Sailing Program (Grant No. 20YF1425000). YYQ was supported by Zhejiang Provincial Natural Science Foundation of China (Grant No. LQ21H050002). ZGZ is sponsored by Shanghai Pujiang Program (21PJD039) and the National Natural Science Foundation of China (Grant No. 82300366).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Bacmeister L, Schwarzl M, Warnke S, Stoffers B, Blankenberg S, Westermann D, et al. Inflammation and fibrosis in murine models of heart failure. Basic Research in Cardiology. 2019; 114: 19.
[2]
Duraes AR, de Souza Lima Bitar Y, Roever L, Neto MG. Endomyocardial fibrosis: past, present, and future. Heart Failure Reviews. 2020; 25: 725–730.
[3]
Weber KT, Janicki JS, Shroff SG, Pick R, Chen RM, Bashey RI. Collagen remodeling of the pressure-overloaded, hypertrophied nonhuman primate myocardium. Circulation Research. 1988; 62: 757–765.
[4]
Frangogiannis NG. Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Molecular Aspects of Medicine. 2019; 65: 70–99.
[5]
Schwarz F, Flameng W, Schaper J, Langebartels F, Sesto M, Hehrlein F, et al. Myocardial structure and function in patients with aortic valve disease and their relation to postoperative results. The American Journal of Cardiology. 1978; 41: 661–669.
[6]
Tanaka M, Fujiwara H, Onodera T, Wu DJ, Hamashima Y, Kawai C. Quantitative analysis of myocardial fibrosis in normals, hypertensive hearts, and hypertrophic cardiomyopathy. British Heart Journal. 1986; 55: 575–581.
[7]
Habib M, Adler A, Fardfini K, Hoss S, Hanneman K, Rowin EJ, et al. Progression of Myocardial Fibrosis in Hypertrophic Cardiomyopathy: A Cardiac Magnetic Resonance Study. JACC. Cardiovascular Imaging. 2021; 14: 947–958.
[8]
Brooks A, Schinde V, Bateman AC, Gallagher PJ. Interstitial fibrosis in the dilated non-ischaemic myocardium. Heart. 2003; 89: 1255–1256.
[9]
Marijianowski MM, Teeling P, Becker AE. Remodeling after myocardial infarction in humans is not associated with interstitial fibrosis of noninfarcted myocardium. Journal of the American College of Cardiology. 1997; 30: 76–82.
[10]
López B, Querejeta R, González A, Sánchez E, Larman M, Díez J. Effects of loop diuretics on myocardial fibrosis and collagen type I turnover in chronic heart failure. Journal of the American College of Cardiology. 2004; 43: 2028–2035.
[11]
Desai HV, Aronow WS, Peterson SJ, Frishman WH. Cardiac amyloidosis: approaches to diagnosis and management. Cardiology in Review. 2010; 18: 1–11.
[12]
Zarate YA, Hopkin RJ. Fabry’s disease. Lancet. 2008; 372: 1427–1435.
[13]
Talman V, Ruskoaho H. Cardiac fibrosis in myocardial infarction-from repair and remodeling to regeneration. Cell and Tissue Research. 2016; 365: 563–581.
[14]
Travers JG, Kamal FA, Robbins J, Yutzey KE, Blaxall BC. Cardiac Fibrosis: The Fibroblast Awakens. Circulation Research. 2016; 118: 1021–1040.
[15]
de Bakker JM, van Capelle FJ, Janse MJ, Tasseron S, Vermeulen JT, de Jonge N, et al. Fractionated electrograms in dilated cardiomyopathy: origin and relation to abnormal conduction. Journal of the American College of Cardiology. 1996; 27: 1071–1078.
[16]
Raafs AG, Verdonschot JAJ, Henkens MTHM, Adriaans BP, Wang P, Derks K, et al. 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. 2021; 23: 933–944.
[17]
López B, Ravassa S, Moreno MU, José GS, Beaumont J, González A, et al. Diffuse myocardial fibrosis: mechanisms, diagnosis and therapeutic approaches. Nature Reviews. Cardiology. 2021; 18: 479–498.
[18]
Pizzuto F, Voci P, Romeo F. Value of echocardiography in predicting future cardiac events after acute myocardial infarction. Current Opinion in Cardiology. 2003; 18: 378–384.
[19]
Gaibazzi N, Bianconcini M, Marziliano N, Parrini I, Conte MR, Siniscalchi C, et al. Scar Detection by Pulse-Cancellation Echocardiography: Validation by CMR in Patients With Recent STEMI. JACC. Cardiovascular Imaging. 2016; 9: 1239–1251.
[20]
Hoyt RH, Collins SM, Skorton DJ, Ericksen EE, Conyers D. Assessment of fibrosis in infarcted human hearts by analysis of ultrasonic backscatter. Circulation. 1985; 71: 740–744.
[21]
Mizuno R, Fujimoto S, Yamaji K, Yutani C, Hashimoto T, Nakamura S. Myocardial ultrasonic tissue characterization for estimating histological abnormalities in hypertrophic cardiomyopathy: comparison with endomyocardial biopsy findings. Cardiology. 2001; 96: 16–23.
[22]
Mizuno R, Fujimoto S, Saito Y, Nakamura S. Non-invasive quantitation of myocardial fibrosis using combined tissue harmonic imaging and integrated backscatter analysis in dilated cardiomyopathy. Cardiology. 2007; 108: 11–17.
[23]
Losi MA, Betocchi S, Chinali M, Barbati G, D’Alessandro G, Cacace A, et al. Myocardial texture in hypertrophic cardiomyopathy. Journal of the American Society of Echocardiography. 2007; 20: 1253–1259.
[24]
Hiremath P, Lawler PR, Ho JE, Correia AW, Abbasi SA, Kwong RY, et al. Ultrasonic Assessment of Myocardial Microstructure in Hypertrophic Cardiomyopathy Sarcomere Mutation Carriers With and Without Left Ventricular Hypertrophy. Circulation. Heart Failure. 2016; 9: e003026.
[25]
Prior DL, Somaratne JB, Jenkins AJ, Yii M, Newcomb AE, Schalkwijk CG, et al. Calibrated integrated backscatter and myocardial fibrosis in patients undergoing cardiac surgery. Open Heart. 2015; 2: e000278.
[26]
Jellis C, Wright J, Kennedy D, Sacre J, Jenkins C, Haluska B, et al. Association of imaging markers of myocardial fibrosis with metabolic and functional disturbances in early diabetic cardiomyopathy. Circulation. Cardiovascular Imaging. 2011; 4: 693–702.
[27]
Gaibazzi N, Tuttolomondo D, Guaricci AI, Di Giannuario G. Pulse-Cancellation Echocardiography for Clinical Evaluation of Myocardial Scar Burden. Current Cardiology Reports. 2021; 23: 100.
[28]
Gaibazzi N, Suma S, Lorenzoni V, Sartorio D, Pressman G, Siniscalchi C, et al. Myocardial Scar by Pulse-Cancellation Echocardiography Is Independently Associated with Appropriate Defibrillator Intervention for Primary Prevention after Myocardial Infarction. Journal of the American Society of Echocardiography. 2020; 33: 1123–1131.
[29]
Giollo A, Vinco G, Cioffi G, Frizzera F, Quinternetto A, Bergamini C, et al. Subclinical Myocardial Fibrosis in Systemic Lupus Erythematosus as Assessed by Pulse-Cancellation Echocardiography: A Pilot Study. Journal of Clinical Medicine. 2022; 11: 4788.
[30]
Vollema EM, Sugimoto T, Shen M, Tastet L, Ng ACT, Abou R, et al. Association of Left Ventricular Global Longitudinal Strain With Asymptomatic Severe Aortic Stenosis: Natural Course and Prognostic Value. JAMA Cardiology. 2018; 3: 839–847.
[31]
Geyer H, Caracciolo G, Abe H, Wilansky S, Carerj S, Gentile F, et al. Assessment of myocardial mechanics using speckle tracking echocardiography: fundamentals and clinical applications. Journal of the American Society of Echocardiography. 2010; 23: 351–369; quiz 453–455.
[32]
Krämer J, Niemann M, Liu D, Hu K, Machann W, Beer M, et al. Two-dimensional speckle tracking as a non-invasive tool for identification of myocardial fibrosis in Fabry disease. European Heart Journal. 2013; 34: 1587–1596.
[33]
Popović ZB, Kwon DH, Mishra M, Buakhamsri A, Greenberg NL, Thamilarasan M, et al. Association between regional ventricular function and myocardial fibrosis in hypertrophic cardiomyopathy assessed by speckle tracking echocardiography and delayed hyperenhancement magnetic resonance imaging. Journal of the American Society of Echocardiography. 2008; 21: 1299–1305.
[34]
Sun W, Shen X, Wang J, Zhu S, Zhang Y, Wu C, et al. Association Between 2D- and 3D-Speckle-Tracking Longitudinal Strain and Cardiovascular Magnetic Resonance Evidence of Diffuse Myocardial Fibrosis in Heart Transplant Recipients. Frontiers in Cardiovascular Medicine. 2021; 8: 727745.
[35]
Lisi M, Cameli M, Righini FM, Malandrino A, Tacchini D, Focardi M, et al. RV Longitudinal Deformation Correlates With Myocardial Fibrosis in Patients With End-Stage Heart Failure. JACC. Cardiovascular Imaging. 2015; 8: 514–522.
[36]
Lisi M, Cameli M, Mandoli GE, Pastore MC, Righini FM, D’Ascenzi F, et al. Detection of myocardial fibrosis by speckle-tracking echocardiography: from prediction to clinical applications. Heart Failure Reviews. 2022; 27: 1857–1867.
[37]
Cui C, Li Y, Liu Y, Huang D, Hu Y, Wang Y, et al. Association Between Echocardiographic Non-invasive Myocardial Work Indices and Myocardial Fibrosis in Patients With Dilated Cardiomyopathy. Frontiers in Cardiovascular Medicine. 2021; 8: 704251.
[38]
Haland TF, Almaas VM, Hasselberg NE, Saberniak J, Leren IS, Hopp E, et al. Strain echocardiography is related to fibrosis and ventricular arrhythmias in hypertrophic cardiomyopathy. European Heart Journal. Cardiovascular Imaging. 2016; 17: 613–621.
[39]
Karamitsos TD, Arvanitaki A, Karvounis H, Neubauer S, Ferreira VM. Myocardial Tissue Characterization and Fibrosis by Imaging. JACC. Cardiovascular Imaging. 2020; 13: 1221–1234.
[40]
Díez J, González A, Kovacic JC. Myocardial Interstitial Fibrosis in Nonischemic Heart Disease, Part 3/4: JACC Focus Seminar. Journal of the American College of Cardiology. 2020; 75: 2204–2218.
[41]
Kim RJ, Chen EL, Lima JA, Judd RM. Myocardial Gd-DTPA kinetics determine MRI contrast enhancement and reflect the extent and severity of myocardial injury after acute reperfused infarction. Circulation. 1996; 94: 3318–3326.
[42]
Bucciarelli-Ducci C, Wu E, Lee DC, Holly TA, Klocke FJ, Bonow RO. Contrast-enhanced cardiac magnetic resonance in the evaluation of myocardial infarction and myocardial viability in patients with ischemic heart disease. Current Problems in Cardiology. 2006; 31: 128–168.
[43]
Fieno DS, Kim RJ, Chen EL, Lomasney JW, Klocke FJ, Judd RM. Contrast-enhanced magnetic resonance imaging of myocardium at risk: distinction between reversible and irreversible injury throughout infarct healing. Journal of the American College of Cardiology. 2000; 36: 1985–1991.
[44]
Mewton N, Liu CY, Croisille P, Bluemke D, Lima JAC. Assessment of myocardial fibrosis with cardiovascular magnetic resonance. Journal of the American College of Cardiology. 2011; 57: 891–903.
[45]
Kim RJ, Fieno DS, Parrish TB, Harris K, Chen EL, Simonetti O, et al. Relationship of MRI delayed contrast enhancement to irreversible injury, infarct age, and contractile function. Circulation. 1999; 100: 1992–2002.
[46]
Arbelo E, Protonotarios A, Gimeno JR, Arbustini E, Barriales-Villa R, Basso C, et al. 2023 ESC Guidelines for the management of cardiomyopathies. European Heart Journal. 2023; ehad194.
[47]
Zhu L, Wang Y, Zhao S, Lu M. Detection of myocardial fibrosis: Where we stand. Frontiers in Cardiovascular Medicine. 2022; 9: 926378.
[48]
Salerno M, Robinson AA. Risk Stratification in Nonischemic Dilated Cardiomyopathy in the Era of Personalized Medicine: Can Cardiac Magnetic Resonance With Late Gadolinium Imaging “Enhance” Our Strategy? JACC. Cardiovascular Imaging. 2018; 11: 1285–1287.
[49]
Rubinshtein R, Glockner JF, Ommen SR, Araoz PA, Ackerman MJ, Sorajja P, et al. Characteristics and clinical significance of late gadolinium enhancement by contrast-enhanced magnetic resonance imaging in patients with hypertrophic cardiomyopathy. Circulation. Heart Failure. 2010; 3: 51–58.
[50]
Georgiopoulos G, Figliozzi S, Pateras K, Nicoli F, Bampatsias D, Beltrami M, et al. Comparison of Demographic, Clinical, Biochemical, and Imaging Findings in Hypertrophic Cardiomyopathy Prognosis: A Network Meta-Analysis. JACC. Heart Failure. 2023; 11: 30–41.
[51]
Becker MAJ, Cornel JH, van de Ven PM, van Rossum AC, Allaart CP, Germans T. The Prognostic Value of Late Gadolinium-Enhanced Cardiac Magnetic Resonance Imaging in Nonischemic Dilated Cardiomyopathy: A Review and Meta-Analysis. JACC. Cardiovascular Imaging. 2018; 11: 1274–1284.
[52]
Gong C, Guo J, Wan K, Wang L, Chen X, Guo J, et al. Detection and evaluation of myocardial fibrosis in Eisenmenger syndrome using cardiovascular magnetic resonance late gadolinium enhancement and T1 mapping. Journal of Cardiovascular Magnetic Resonance. 2022; 24: 60.
[53]
Papanastasiou CA, Kokkinidis DG, Kampaktsis PN, Bikakis I, Cunha DK, Oikonomou EK, et al. The Prognostic Role of Late Gadolinium Enhancement in Aortic Stenosis: A Systematic Review and Meta-Analysis. JACC. Cardiovascular Imaging. 2020; 13: 385–392.
[54]
Hundley WG, Bluemke D, Bogaert JG, Friedrich MG, Higgins CB, Lawson MA, et al. Society for Cardiovascular Magnetic Resonance guidelines for reporting cardiovascular magnetic resonance examinations. Journal of Cardiovascular Magnetic Resonance. 2009; 11: 5.
[55]
Flett AS, Hasleton J, Cook C, Hausenloy D, Quarta G, Ariti C, et al. Evaluation of techniques for the quantification of myocardial scar of differing etiology using cardiac magnetic resonance. JACC. Cardiovascular Imaging. 2011; 4: 150–156.
[56]
Gao Y, Wang HP, Liu MX, Gu H, Yuan XS, Biekan J, et al. Early detection of myocardial fibrosis in cardiomyopathy in the absence of late enhancement: role of T1 mapping and extracellular volume analysis. European Radiology. 2023; 33: 1982–1991.
[57]
Messroghli DR, Walters K, Plein S, Sparrow P, Friedrich MG, Ridgway JP, et al. Myocardial T1 mapping: application to patients with acute and chronic myocardial infarction. Magnetic Resonance in Medicine. 2007; 58: 34–40.
[58]
Liu X, Hou JL, Yang ZG, Xia CC, Xie LJ, Ye PF, et al. Native T_1 mapping for characterization of acute and chronic myocardial infarction in swine: Comparison with contrast-enhanced MRI. Journal of Magnetic Resonance Imaging. 2018; 47: 1406–1414.
[59]
Iles L, Pfluger H, Phrommintikul A, Cherayath J, Aksit P, Gupta SN, et al. Evaluation of diffuse myocardial fibrosis in heart failure with cardiac magnetic resonance contrast-enhanced T1 mapping. Journal of the American College of Cardiology. 2008; 52: 1574–1580.
[60]
Xu J, Zhuang B, Sirajuddin A, Li S, Huang J, Yin G, et al. MRI T1 Mapping in Hypertrophic Cardiomyopathy: Evaluation in Patients Without Late Gadolinium Enhancement and Hemodynamic Obstruction. Radiology. 2020; 294: 275–286.
[61]
Lin L, Li X, Feng J, Shen KN, Tian Z, Sun J, et al. The prognostic value of T1 mapping and late gadolinium enhancement cardiovascular magnetic resonance imaging in patients with light chain amyloidosis. Journal of Cardiovascular Magnetic Resonance. 2018; 20: 2.
[62]
Martinez-Naharro A, Kotecha T, Norrington K, Boldrini M, Rezk T, Quarta C, et al. Native T1 and Extracellular Volume in Transthyretin Amyloidosis. JACC. Cardiovascular Imaging. 2019; 12: 810–819.
[63]
Kramer CM, Chandrashekhar Y, Narula J. T1 mapping by CMR in cardiomyopathy: a noninvasive myocardial biopsy? JACC. Cardiovascular Imaging. 2013; 6: 532–534.
[64]
Hinojar R, Varma N, Child N, Goodman B, Jabbour A, Yu CY, et al. T1 Mapping in Discrimination of Hypertrophic Phenotypes: Hypertensive Heart Disease and Hypertrophic Cardiomyopathy: Findings From the International T1 Multicenter Cardiovascular Magnetic Resonance Study. Circulation. Cardiovascular Imaging. 2015; 8: e003285.
[65]
Bettencourt N, Ferreira ND, Leite D, Carvalho M, Ferreira WDS, Schuster A, et al. CAD detection in patients with intermediate-high pre-test probability: low-dose CT delayed enhancement detects ischemic myocardial scar with moderate accuracy but does not improve performance of a stress-rest CT perfusion protocol. JACC. Cardiovascular Imaging. 2013; 6: 1062–1071.
[66]
Berliner JI, Kino A, Carr JC, Bonow RO, Choudhury L. Cardiac computed tomographic imaging to evaluate myocardial scarring/fibrosis in patients with hypertrophic cardiomyopathy: a comparison with cardiac magnetic resonance imaging. The International Journal of Cardiovascular Imaging. 2013; 29: 191–197.
[67]
Esposito A, Palmisano A, Antunes S, Maccabelli G, Colantoni C, Rancoita PMV, et al. Cardiac CT With Delayed Enhancement in the Characterization of Ventricular Tachycardia Structural Substrate: Relationship Between CT-Segmented Scar and Electro-Anatomic Mapping. JACC. Cardiovascular Imaging. 2016; 9: 822–832.
[68]
Patino M, Prochowski A, Agrawal MD, Simeone FJ, Gupta R, Hahn PF, et al. Material Separation Using Dual-Energy CT: Current and Emerging Applications. Radiographics. 2016; 36: 1087–1105.
[69]
Zheng L, Ding X, Liu K, Feng S, Tang B, Li Q, et al. Molecular imaging of fibrosis using a novel collagen-binding peptide labelled with 99mTc on SPECT/CT. Amino Acids. 2017; 49: 89–101.
[70]
Velikyan I, Rosenström U, Rosestedt M, Eriksson O, Antoni G. Improved Radiolytic Stability of a 68Ga-labelled Collagelin Analogue for the Imaging of Fibrosis. Pharmaceuticals. 2021; 14: 990.
[71]
Verjans JWH, Lovhaug D, Narula N, Petrov AD, Indrevoll B, Bjurgert E, et al. Noninvasive imaging of angiotensin receptors after myocardial infarction. JACC. Cardiovascular Imaging. 2008; 1: 354–362.
[72]
Fukushima K, Bravo PE, Higuchi T, Schuleri KH, Lin X, Abraham MR, et al. Molecular hybrid positron emission tomography/computed tomography imaging of cardiac angiotensin II type 1 receptors. Journal of the American College of Cardiology. 2012; 60: 2527–2534.
[73]
van den Borne SWM, Isobe S, Verjans JW, Petrov A, Lovhaug D, Li P, et al. Molecular imaging of interstitial alterations in remodeling myocardium after myocardial infarction. Journal of the American College of Cardiology. 2008; 52: 2017–2028.
[74]
Tillmanns J, Hoffmann D, Habbaba Y, Schmitto JD, Sedding D, Fraccarollo D, et al. Fibroblast activation protein alpha expression identifies activated fibroblasts after myocardial infarction. Journal of Molecular and Cellular Cardiology. 2015; 87: 194–203.
[75]
Heckmann MB, Reinhardt F, Finke D, Katus HA, Haberkorn U, Leuschner F, et al. Relationship Between Cardiac Fibroblast Activation Protein Activity by Positron Emission Tomography and Cardiovascular Disease. Circulation. Cardiovascular Imaging. 2020; 13: e010628.
[76]
Varasteh Z, Mohanta S, Robu S, Braeuer M, Li Y, Omidvari N, et al. Molecular Imaging of Fibroblast Activity After Myocardial Infarction Using a 68Ga-Labeled Fibroblast Activation Protein Inhibitor, FAPI-04. Journal of Nuclear Medicine. 2019; 60: 1743–1749.
[77]
Notohamiprodjo S, Nekolla SG, Robu S, Villagran Asiares A, Kupatt C, Ibrahim T, et al. Imaging of cardiac fibroblast activation in a patient after acute myocardial infarction using 68Ga-FAPI-04. Journal of Nuclear Cardiology. 2022; 29: 2254–2261.
[78]
Kessler L, Kupusovic J, Ferdinandus J, Hirmas N, Umutlu L, Zarrad F, et al. Visualization of Fibroblast Activation After Myocardial Infarction Using 68Ga-FAPI PET. Clinical Nuclear Medicine. 2021; 46: 807–813.
[79]
Zhang M, Quan W, Zhu T, Feng S, Huang X, Meng H, et al. [68Ga]Ga-DOTA-FAPI-04 PET/MR in patients with acute myocardial infarction: potential role of predicting left ventricular remodeling. European Journal of Nuclear Medicine and Molecular Imaging. 2023; 50: 839–848.
[80]
Lyu Z, Han W, Zhao H, Jiao Y, Xu P, Wang Y, et al. A clinical study on relationship between visualization of cardiac fibroblast activation protein activity by Al18F-NOTA-FAPI-04 positron emission tomography and cardiovascular disease. Frontiers in Cardiovascular Medicine. 2022; 9: 921724.
[81]
Wang G, Yang Q, Wu S, Xu X, Li X, Liang S, et al. Molecular imaging of fibroblast activity in pressure overload heart failure using [68 Ga]Ga-FAPI-04 PET/CT. European Journal of Nuclear Medicine and Molecular Imaging. 2023; 50: 465–474.
[82]
Wang L, Wang Y, Wang J, Xiao M, Xi XY, Chen BX, et al. Myocardial Activity at 18F-FAPI PET/CT and Risk for Sudden Cardiac Death in Hypertrophic Cardiomyopathy. Radiology. 2023; 306: e221052.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top