Next Article in Journal
Effectiveness of Equine-Assisted Activities and Therapies for Improving Adaptive Behavior and Motor Function in Autism Spectrum Disorder
Next Article in Special Issue
Special Issue: Rare Respiratory Diseases: A Personal and Public Health Issue
Previous Article in Journal
Clinical Features and Cutaneous Manifestations of Juvenile and Adult Patients of Dermatomyositis Associated with Myositis-Specific Autoantibodies
Previous Article in Special Issue
Alpha-1 Antitrypsin Screening in a Selected Cohort of Patients Affected by Chronic Pulmonary Diseases in Naples, Italy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Utility of Transient Elastography for the Screening of Liver Disease in Patients with Alpha1-Antitrypsin Deficiency

1
Liver Unit, Department of Internal Medicine, Hospital Universitari Vall d’Hebron, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
2
Pneumology Department, Hospital Universitari Vall d’Hebron/Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
3
Department of Medicine, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
4
Pneumology Department, University Hospital Complex of Vigo, Instituto de Investigación Biomédica Galicia Sur, 36213 Vigo, Spain
5
Pneumology Department, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Hospital Clínico de San Carlos, Departamento de Medicina, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
6
Department of Clinical Biochemistry, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
7
Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain
8
Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
*
Author to whom correspondence should be addressed.
Both authors have contributed equally and should be considered first authors.
J. Clin. Med. 2021, 10(8), 1724; https://doi.org/10.3390/jcm10081724
Submission received: 5 March 2021 / Revised: 12 April 2021 / Accepted: 13 April 2021 / Published: 16 April 2021
(This article belongs to the Special Issue Rare Respiratory Diseases: A Personal and a Public Health Problem)

Abstract

:
Screening of liver disease in alpha-1 antitrypsin deficiency (AATD) is usually carried out with liver enzymes, with low sensitivity. We conducted a multicenter cross-sectional study aiming to describe the utility of transient elastography for the identification of liver disease in patients with AATD. A total of 148 AATD patients were included. Among these, 54.7% were Pi*ZZ and 45.3% were heterozygous for the Z allele. Between 4.9% and 16.5% of patients had abnormal liver enzymes, without differences among genotypes. Liver stiffness measurement (LSM) was significantly higher in Pi*ZZ individuals than in heterozygous Z (5.6 vs. 4.6 kPa; p = 0.001). In total, in 8 (5%) individuals LSM was >7.5 kPa, considered significant liver fibrosis, and ≥10 kPa in 3 (1.9%) all being Pi*ZZ. Elevated liver enzymes were more frequently observed in patients with LSM > 7.5 kPa, but in 5 out of 8 of these patients all liver enzymes were within normal range. In patients with AATD, the presence of abnormal liver enzymes is frequent; however, most of these patients do not present significant liver fibrosis. Transient elastography can help to identify patients with liver fibrosis even with normal liver enzymes and should be performed in all Z-allele carriers to screen for liver disease.

1. Introduction

Alpha1-antitrypsin deficiency (AATD) is caused by a specific mutation of the SERPINA 1 gene which results in abnormal production and low circulating levels of alpha1-antitrypsin (AAT). It is one of the most common genetic diseases in adulthood and is associated with an increased risk of developing pulmonary emphysema and liver disease [1,2].
AAT is a protein synthesized and secreted mainly by hepatocytes, the main function of which is to protect lung tissue from damage caused by proteolytic enzymes such as neutrophil elastase [2]. AAT is a highly polymorphic protein with more than 120 variants, including about 60 deficient alleles. The normal allele, present in more than 95% of normal subjects, is called M [1,2]. The most frequent deficient alleles are S and Z, and they are found in 10% and 2% of the Spanish population, respectively [3,4,5,6].
The Z variant presents an alteration in its tertiary structure that facilitates misfolding of the protein and gives rise to the spontaneous formation of polymers, leading to the accumulation of the protein in the endoplasmic reticulum of the hepatocytes [7,8]. Liver damage is then caused by this protein accumulation, inducing apoptosis of the hepatocytes and a compensatory hepatocyte proliferation that eventually produces liver fibrosis that can evolve to cirrhosis or hepatocellular carcinoma [8,9]. Most patients with liver disease are homozygous for the deficient Z allele (Pi*ZZ), although different degrees of liver involvement have been described in heterozygotes (Pi*SZ and Pi*MZ), especially if associated with other co-factors such as alcohol consumption or metabolic syndrome [10,11].
Currently there is no non-invasive gold standard technique for the screening and early diagnosis of liver disease in patients with AATD [12]. In clinical practice, liver enzymes are routinely checked, while liver ultrasound is performed if necessary. However, it has been observed that transaminase levels have a low sensitivity to identify liver disease, and they correlate little with the degree of liver disease, especially in adulthood [13]. Serum biomarkers and image devices based on elastography technique have been developed to overcome this problem and to assess the presence of fibrosis in liver diseases of different etiologies [14,15].
Recently, there has been increasing interest in the use of elastographic methods, such as transient elastography, for screening liver disease in AATD patients [16,17,18]. However, the screening and management of asymptomatic liver disease in AATD can differ among centers due to a lack of consensus or guidelines. Therefore, the aim of our study was to describe the utility of transient elastography for the identification of liver disease in patients with AATD.

2. Materials and Methods

This was a multicenter cross-sectional study including patients older than 18 years with mild, moderate, and severe AATD (Pi*MS, SS, MZ, SZ, ZZ, and rare variants) consecutively recruited from the outpatient Pneumology Clinics of three AATD reference centers in Spain (Vall d’Hebron University Hospital, Barcelona, University Hospital Complex of Vigo, and Hospital Clínico San Carlos, Madrid) from 1 April 2017 to 1 January 2020. As part of the assessment of patients with AATD, all of them were offered blood analysis, full lung function tests, and transient elastography, and the only exclusion criterion was to refuse to sign informed consent. The study was approved by the Vall d’He- bron Hospital Ethics Committee (Barcelona, Spain), number PR(AG)335/2016, and all patients provided written informed consent.

2.1. Variables

During the first visit, a complete physical examination was performed in all patients with special interest in signs of chronic liver disease such as splenomegaly, jaundice, or palmar erythema. Sociodemographic and clinical characteristics were collected and other parameters such as body mass index (BMI), lung function tests (forced expiratory volume in the first second (FEV1), FEV1/forced ventilatory capacity (FVC), and carbon monoxide transfer coefficient (KCO)), comorbidities, treatments, and AAT augmentation therapy were reported. Diagnosis of chronic obstructive pulmonary disease (COPD) was established when the post-bronchodilator FEV1/FVC ratio was below 0.7.
Blood samples were obtained for determination of liver function tests: Aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP), international normalized ratio (INR), platelet count, and albumin. In addition, the Fibrosis-4 (FIB-4) score was calculated as age (years) × AST [IU/L]/(platelet count [109/L] × √ALT [IU/L]) and AST-to-platelet ratio index (APRI) as (AST [IU/L]/40 IU/L)/platelet count [109/L] × 100. Patients were classified according to the previously established FIB-4 cut-offs of <1.45 with a high negative predictive value for ruling out advanced fibrosis and >3.25 with a high specificity and a 65% positive predictive value for ruling in advanced fibrosis [19]. For APRI, we used the cut-off <0.5 for excluding cirrhosis (high negative predictive value) and >1.0 as a high specific cut-off for predicting cirrhosis [20].
The Enhanced Liver Fibrosis (ELF) test (Siemens Healthcare Diagnostics, Vienna, Austria) was available as a biomarker of liver fibrosis in one of the centers. The ELF test is a panel of markers that consists of 3 components: Type III procollagen peptide, hyaluronic acid, and tissue inhibitor of metalloproteinase-1. We explored the manufacturer- recommended 9.8 cut-off to rule in advanced fibrosis [21].

2.2. Liver Stiffness Measurement by Transient Elastography

Liver stiffness measurements (LSM) were performed in a fasting state using a Fibroscan 502 Touch (Echosens, Paris, France) using the M or XL probe as per device indication. Quality criteria used in all centers were at least 10 valid measurements and an interquartile-to-median ratio ≤ 30%. The LSM technique was carried out in accordance with the European Association for Study of the Liver (EASL) clinical guidelines [22].
Results were expressed in kilopascals (kPa). Normal liver stiffness values are around 5 kPa. Transient elastography has good re-producibility and has good diagnostic performance for estimating liver fibrosis. However, the accuracy is not as good for detecting significant fibrosis compared to advanced fibrosis or cirrhosis [22,23]. Since there are no specific LSM cut-offs for AATD liver disease, a LSM > 7.5 kPa was used as suggestive of significant fibrosis and ≥10 kPa was suggestive of advanced fibrosis according to previously established cut-offs in other liver diseases (mainly viral etiologies and alcoholic liver disease) [22,24].
The presence of steatosis was assessed by the controlled attenuation parameter (CAP) and results were expressed in decibel per meter (dB/m). The cut-off >268 dB/m was used as an indicator of moderate steatosis, and for severe steatosis the cut-off was >280 dB/m [25].

2.3. Statistical Analysis

Qualitative variables were described with absolute frequencies and percentages. The description of quantitative variables was performed using the mean, standard deviation (SD) or median, and interquartile range (IQR). The Kolmogorov–Smirnov test was used to assess the normality of distributions.
Patient characteristics were compared according to genotypes and other clinical conditions. In the case of quantitative variables, the Student’s t-test for normally distributed variables or the Mann–Whitney U-test if normality was not assumed was used, while ANOVA tests were performed in the case of variables with more than 2 categories. The Chi-squared test (Fisher test for frequencies < 5) was used for the comparison of categorical variables. A linear relationship between quantitative variables, in particular between surrogates of liver disease (LSM, CAP and FIB-4) and spirometric markers of airflow obstruction (FEV1(%) and FEV1/FVC), were analyzed using Spearman tests. For all the tests, p-values < 0.05 were considered statistically significant. The statistical package R Studio (V2.5.1) was used for the analyses.

3. Results

3.1. Demographic and Clinical Findings

A total of 148 AATD patients were included from January 2017 to December 2019. Among these, 81 (54.7%) were homozygous Pi*ZZ and 67 (45.3%) were heterozygous for the Z allele (29 Pi*SZ, 35 Pi*MZ, 1 Pi*FZ, 1 Pi*PlowellZ, 1 Pi*MmaltonZ).
The mean age was 52.5 and 57 years for heterozygous and Pi*ZZ, respectively, and 50% of the patients were male. Liver disease in infancy was reported as the cause of the diagnosis of AATD in 19.4% and 11.1% of heterozygous and homozygous patients, although there were no patients with an active diagnosis of liver disease at the time of the study. COPD was diagnosed in 22.7% of heterozygous subjects and up to 70% for Pi*ZZ patients. Consequently, the mean FEV1 (%) was significantly lower in Pi*ZZ compared with heterozygous (69% (SD: 30.5%) versus 92.9% (SD: 27.6%); p < 0.001). The baseline characteristics of the global population and the two genotype groups are shown in Table 1.

3.2. Clinical and Laboratory Signs of Liver Disease

Thirty-two patients (21.6%) had abnormal liver enzymes. The distribution of values showed significant differences only in AST values, which were significantly higher in Pi*ZZ patients (29.2 UI/L (SD: 15.4) vs. 25.0 UI/L (SD: 8.0; p = 0.029). The most frequent pattern was an elevation in GGT (14.9% of patients). Pi*ZZ patients had a higher FIB-4 score compared to heterozygous Z (1.6 (SD: 0.8) vs. 1.2 (SD:0.5); p < 0.001). Only 5 patients had FIB-4 > 3.25 and all were Pi*ZZ. The APRI score was higher in Pi*ZZ patients than in heterozygous Z (0.35 (SD: 0.18) vs. 0.27 (SD: 0.09); p = 0.007), but most of the patients had APRI values < 0.5, excluding advanced fibrosis or cirrhosis, and only one Pi*ZZ patient had an APRI score > 1.0. The ELF score was obtained in 52 patients (27 Pi*ZZ and 25 Pi*Z patients). Pi*ZZ had significantly higher values compared to Pi*Z phenotypes (8.6 (SD: 0.8) vs. 8 (SD: 0.6); p = 0.007). Only 1 Pi*ZZ patient showed values above the cut-off of 9.8 (Table 2).

3.3. Transient Elastography

The mean LSM was significantly higher in Pi*ZZ individuals than in heterozygous Z (5.6 (SD: 2.5) kPa vs. 4.6 (SD:1.2) kPa, respectively; p = 0.007). In total, LSM was >7.5 kPa in 8 (5%) individuals and ≥10 kPa in 3 (1.9%), all being Pi*ZZ (Figure 1). By lowering the cut-off of LSM to >7.1 kPa as suggested in other studies [11], we found 10 Pi*ZZ patients (12.3%) and 3 heterozygous patients (4.5%), two of whom were Pi*SZ patients with LSM 7.3 kPa, and one was a Pi*MZ patient with LSM 7.5 kPa.
Using the LSM > 8.45 kPa cut-off of the study by Clark et al. [13], we would have identified 4 Pi*ZZ patients (4.9%) suggestive of having F ≥ 3.
Almost one-third of the patients had severe steatosis according to CAP values > 280 dB/m, with no significant differences between homozygous and heterozygous patients. (Table 2).

3.4. Characteristics of Pi*ZZ Patients According to LSM Values

Pi*ZZ individuals with LSM > 7.5 kPa were older and had a higher BMI. Two-thirds consumed alcohol, and all had COPD (versus 67% in patients with LSM ≤ 7.5 kPa; p = 0.097).
Elevated liver enzymes were more frequently observed in patients with LSM > 7.5 kPa. Twenty-five percent of patients with LSM > 7.5 kPa had elevated AST values compared to 2.7% in patients with LSM ≤ 7.5 kPa (p = 0.048), and 37.5% of patients with LSM > 7.5 kPa had elevated GGT compared to 14.1% of patients with LSM ≤ 7.5 kPa (p = 0.120) (Table 3, Figure 2). Conversely, 11/61 patients (18%) had at least one elevated liver enzyme but with normal LSM values (LSM < 6 kPa). Correlations between LSM and liver enzymes were only significant, albeit weakly, between LSM and AST (0.311 (p < 0.001)), and LSM and GGT (0.389 (p < 0.001)).
Among the 8 patients with LSM > 7.5, 3 had GGT above the normal limit and 1 also had a FIB-4 score > 3.25 (Figure 3). The FIB-4 score (2.2 (SD: 0.7) versus 1.5 (SD: 0.8); p = 0.032), as well as CAP measurement (317.9 (SD: 48) dB/m vs. 249.6 (SD: 56.5) dB/m; p = 0.004), were also higher in Pi*ZZ patients with LSM > 7.5 kPa (Table 3). Severe steatosis, with CAP > 280 dB/m, was present in 6 patients (75%) with LSM > 7.5 kPa compared to 20 patients (27.4%) with LSM < 7.5 kPa (p = 0.041).
The APRI was higher in Pi*ZZ patients with LSM > 7.5 kPa than in those with LSM ≤ 7.5 kPa (0.56 vs. 0.33, p < 0.001). The APRI had a significant correlation with LSM (r = 0.353, p = 0.030).

3.5. Comparison between Pi*ZZ Patients with or without COPD

Fifty-seven Pi*ZZ patients (70.4%) had COPD. Pi*ZZ patients with COPD were older and more frequently had a history of smoking compared with non-COPD individuals. As expected, they had worse lung function with a lower FEV1 (1.6 (SD: 0.8) L vs. 3.5 (SD: 1) L; p < 0.001) and KCO (%) (43.7% (SD: 30.8%) vs. 68% (SD: 30.4%); p = 0.003).
Regarding the liver study, no differences were observed in transaminase levels, but the FIB-4 score was higher in COPD patients (1.7 (SD: 0.8) vs. 1.2 (SD: 0.8); p = 0.046). More individuals in the COPD group had a LSM > 7.5 kPa (14% vs. 0%; p = 0.097) and they also had higher CAP values (265.9 (SD: 58.3) dB/m vs. 233.5 (SD: 55.8) dB/m; p = 0.023) (Table 3). Significant, albeit weak, correlations were found between FIB-4 and FEV1 (mL) (r = −0.350, p = 0.002), and CAP and FEV1 (mL) and FEV1(%) (r = −0.391, p < 0.001 and r = −0.306, p = 0.006, respectively). No significant correlations were found between LSM or ELF and measures of airflow obstruction.

4. Discussion

In our study population, we found that 10% of Pi*ZZ individuals had transient elastography results suggestive of liver fibrosis, but none of the heterozygous individuals reached the suggested threshold. Although individuals with higher LSM had higher transaminase levels and FIB-4 scores, normal levels of these biomarkers did not reliably rule out liver disease, since some of the patients with normal values had high LSM values. All patients with high LSM also had COPD.
Transient elastography is a non-invasive tool that has proven to be useful in the diagnosis of liver fibrosis of different etiologies. More recently, its utility has also been explored in AATD-related liver disease with promising results [16,17,18,26]. Although different cut-offs have been proposed, there is no validated cut-off of LSM for AATD liver disease. In a study including 94 Pi*ZZ patients with paired LSM and liver biopsies, Clark et al. [26] observed that cut-offs of 5.54 and 8.45 kPa had the highest accuracy for detecting significant fibrosis (≥F2) and advanced fibrosis (≥F3), respectively. However, these cut-offs had a low specificity and a low positive predictive value. Hamesch et al. [17] increased the cut-off for significant fibrosis to >7.1 kPa in order to increase the positive predictive value, confirming the presence of ≥F2 in 22 out of 23 patients with liver biopsies [27], while Guillaud et al. [16] suggested an LSM > 7.2 kPa for significant fibrosis and LSM > 14 kPa for cirrhosis. In another study in 75 patients with AATD, the investigators offered a liver biopsy to all individuals with a LSM > 6 or altered liver enzymes in combination with an abnormal ultrasound. Among the 11 biopsies analyzed, they found that the LSM scores in patients with moderate or severe fibrosis were >8 kPa [18]. According to these results and the cut-offs previously established in other etiologies, we chose an arbitrary cut-off of LSM > 7.5 kPa as suggestive of significant fibrosis, and LSM ≥ 10 kPa as advanced fibrosis/cirrhosis. In our sample, there were two Pi*SZ patients with LSM = 7.3 kPa, one of whom was overweight and had diabetes mellitus and increased GGT values, and the other was a Pi*MZ patient with LSM = 7.5 kPa without other identified risk factors of liver disease. Since the etiology of liver disease has an impact on LSM and the data on AATD induced liver disease are limited [28], further studies are needed to validate the best LSM cut-off for screening of liver disease in AATD.
Ten percent of Pi*ZZ patients in our cohort had LSM > 7.5 kPa, similar to the prevalence of liver fibrosis reported in initial studies in AATD patients, which varied from 10–15% in clinical studies [29,30] to 37% in autopsy studies [31]. More recently, with the development of transient elastography, there has been growing interest in the early detection of liver disease in AATD. The study by Guillaud et al. [16] described 5 patients (18%) with LSM suggestive of significant fibrosis and 2 patients (7%) with LSM suggestive of advanced liver fibrosis/cirrhosis. Other studies have reported a higher prevalence; Hamesch et al. [17] described a prevalence of liver fibrosis of 23.6% among 403 Pi*ZZ individuals and observed that liver disease was 9 to 20 times more frequent in this population compared to non-AAT-deficient individuals. In a cohort of COPD Pi*ZZ patients referred for lung transplantation, Morer et al. [32] found that 13% of patients had significant fibrosis (F2) and 8% advanced fibrosis (≥F3). Similar to these numbers, 8 (14%) of our COPD Pi*ZZ patients had LSM > 7.5 kPa, while in 3 (5.7%) LSM was higher than 10 kPa, suggesting the presence of advanced fibrosis.
In our cohort, Pi*MZ individuals had lower values of LSM compared to Pi*ZZ individuals. The mean LSM was 4.7 kPa for the 34 Pi*MZ patients included. None of these patients had values above 7.5, and only one had LSM = 7.5 kPa. In this patient, other co-factors for liver disease such as obesity, alcohol consumption, or metabolic syndrome were not found. The incidence of liver disease could be higher in heterozygous Z than in the general population, although some authors have hypothesized that while the Pi*MZ genotype acts as a disease modifier, it is not sufficient per se to trigger clinically relevant liver impairment [33]. In a study that analyzed 1184 individuals with non-alcoholic fatty liver disease (NAFLD) and 2462 with chronic alcohol misuse, the Z variant increased the risk of patients with NAFLD to develop cirrhosis and was more frequently present in alcohol misusers with cirrhosis compared to those without significant liver injury [34]. In contrast, a recent analysis of data from the European alpha-1 liver cohort showed that 10% out 419 Pi*MZ had LSM values ≥ 7.1 kPa compared with 4% of non-Z carriers. After adjusting for potential confounders, Pi*MZ individuals still had significantly higher odds for LSM ≥ 7.1 kPa [12]. There is agreement that, in coexistence with other risk factors, and especially in the context of alcohol misuse or NAFLD, Z carriage is a strong risk factor for the development of cirrhosis [17,18] and may also lead to faster hepatic decompensations [35]. In our cohort, 60% of Pi*ZZ patients with LSM > 7.5 kPa had some alcohol consumption and had a higher BMI than those with LSM ≤ 7.5 kPa, and, therefore, these factors could have contributed to the progression of liver disease.
Liver enzymes have often been used to screen liver disease in AATD in clinical practice [36]. In our cohort, elevated liver enzymes and FIB-4 were more frequently observed in patients with LSM > 7.5 kPa, but normal levels were also frequently present in patients with high LSM. In fact, liver enzyme alterations ranged from only 25% of cases for AST and ALT to 37.5% for GGT in Pi*ZZ patients with LSM > 7.5 kPa. Patients with fibrosis or even cirrhosis may present normal serum liver enzymes [11], and this has also been observed in Pi*ZZ individuals [13,17]. On the other hand, up to 10% of AATD patients with normal liver function tests and ultrasound may have increased LSM values [16]. Furthermore, an increase in ALT has a low sensitivity for identifying liver disease in AATD individuals [13,15]. In the European alpha-1 liver cohort, heterozygous Pi*MZ carriers also had higher serum transaminases compared to non-carriers, although this percentage varied from 5.4% to 28.6% and was higher in individuals older than 50 years [12].
The relationship between lung and liver disease in individuals with AATD is controversial. The first series of patients with the deficiency suggested that lung and liver disease rarely coexisted in AATD, and liver disease was more frequently reported in AATD never smokers compared to smokers [37,38]. However, more recent studies using new diagnostic techniques have reported more frequent coexistence of the alterations in both organs [39]. In this line, all of our patients with elevated LSM also had COPD, although the correlation between lung function and LSM was not significant. Moreover, recruiting patients from respiratory departments may have influenced the high prevalence of COPD among patients with elevated LSM; although they were also older, with higher BMI and with a higher frequency of alcohol misuse compared with patients with normal LSM. Therefore, a clear relationship between elevated LSM and lung disease cannot be established from our results.
Our study had some limitations. First, the identification of liver fibrosis was only made by transient elastography as we did not perform liver biopsies. However, as there are no specific treatments for AATD liver disease to date, the performance of an invasive diagnostic technique in otherwise asymptomatic patients may not be justified. Second, this was a cross-sectional study, and data on the evolution of LSM over time were not available. Third, the design of our study did not allow us to investigate a causal relationship between AATD and liver alterations. Our sample size was not big enough for a multivariate analysis adjusted for known confounders of increased liver fibrosis. However, the study had some strengths: We recruited individuals from three reference centers, and, considering that AATD is a rare disease, we reported information from a large series of patients with homozygous and heterozygous AATD.
In conclusion, the results of this study support the assessment of liver disease in all AATD Pi*ZZ individuals and heterozygous Pi*Z individuals with additional liver risk factors. Transient elastography has been shown to be a valuable tool to screen for AATD liver disease, and collaboration between hepatologists and pneumologists is crucial for providing the best care to AATD patients. Due to the poor correlation between liver enzymes and other serum biomarkers and the underlying liver disease, all Z-allele carriers, even those with normal serum biomarker values, should be screened with transient elastography. Since AATD is a rare disease, international collaboration in large registries is needed to investigate the best screening strategy for lung and liver disease [12,17,40].

Author Contributions

Conceptualization, M.P., A.N., C.E., M.M., and M.B.; methodology, M.P., A.N., and C.E.; formal analysis, C.E.; investigation, M.P., A.N., C.E., M.T.-D., J.L.R.-H., M.C., R.T.-P., I.B., F.R.-F., J.G., M.M., and M.B.; resources, M.T.-D., J.L.R.-H., M.C., R.T.-P., J.G., and M.M.; data curation, M.P., A.N., C.E., M.T.-D., J.L.R.-H. and E.R.; writing—original draft preparation, M.P., A.N., M.M., and M.B.; writing—review and editing, C.E., M.T.-D., J.L.R.-H., M.C., R.T.-P., I.B., F.R.-F., and J.G.; supervision, J.G., M.M., and M.B.; project administration, M.M.; funding acquisition, M.T.-D., J.L.R.-H., M.C., and M.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Grifols through an unrestricted grant from the Catalan Center for Research in Alpha-1 antitrypsin deficiency of the Vall d’Hebron Research Institute (VHIR) in the Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain; from the Madrid Center for Research in Alpha-1 antitrypsin deficiency of the Hospital Clínico San Carlos, Madrid, Spain; from the Galicia Center for Research in Alpha-1 antitrypsin deficiency of the University Hospital Complex of Vigo, Spain; as well as a research grant from Fundació Catalana de Pneumologia (FUCAP).

Institutional Review Board Statement

The study was approved by the Vall d’Hebron Hospital Ethics Committee (Barcelona, Spain), number PR(AG)335/2016 (28 October 2016).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Data are available from the authors upon request.

Acknowledgments

Mònica Pons was the recipient of a Rio Hortega contract in the 2018 Strategic Action Health Call from the Instituto de Salud Carlos III for the years 2019–2020. Alexa Núñez was the recipient of a Rio Hortega contract in the 2019 Strategic Action Health Call from the Instituto de Salud Carlos III for the years 2020–2022. We thank Ignacio Martín Granizo and Montserrat Figueira Alvarez for their collaboration in the elastography in the University Hospital Complex of Vigo.

Conflicts of Interest

Myriam Calle received speaking or consulting fees from Boehringer Ingelheim, Grifols, Chiesi, CSL Behring, AstraZeneca, GlaxoSmithKline, Menarini, Gebro Pharma, Zambon, and Novartis. There is no real or perceived conflict of interest between these sources and the present paper. Juan Luis Rodríguez-Hermosa received speaking fees from Boehringer Ingelheim, GlaxoSmithKline, Grifols, CSL Behring, Zambon, and Gebro Pharma. There is no real or perceived conflict of interest between these sources and the present paper. Cristina Esquinas received speaker fees from CSL Behring. Marc Miravitlles received speaker or consulting fees from AstraZeneca, Bial, Boehringer Ingelheim, Chiesi, Cipla, CSL Behring, Laboratorios Esteve, Gebro Pharma, Kamada, GlaxoSmithKline, Grifols, Menarini, Mereo Biopharma, Novartis, pH Pharma, Rovi, TEVA, Spin Therapeutics, Verona Pharma, and Zambon, and research grants from Grifols. Miriam Barrecheguren received speaker fees from Grifols, Menarini, CSL Behring, and GSK, and consulting fees from GSK, Novartis, Boehringer Ingelheim, and Gebro Pharma. The remaining authors report no conflict of interest.

References

  1. Miravitlles, M.; Dirksen, A.; Ferrarotti, I.; Koblizek, V.; Lange, P.; Mahadeva, R.; MacElvaney, N.G.; Parr, D.; Piitulainen, E.; Roche, N.; et al. European Respiratory Society Statement: Diagnosis and treatment of pulmonary disease in alpha-1 antitrypsin deficiency. Eur. Respir. J. 2017, 50, 1700610. [Google Scholar] [CrossRef] [Green Version]
  2. Strnad, P.; McElvaney, N.G.; Lomas, D.A. Alpha1-Antitrypsin Deficiency. N. Engl. J. Med. 2020, 382, 1443–1455. [Google Scholar] [CrossRef]
  3. Blanco, I.; Bueno, P.; Diego, I.; Pérez-Holanda, S.; Casas-Maldonado, F.; Esquinas, C.; Miravitlles, M. Alpha-1 antitrypsin Pi*Z gene frequency and Pi*ZZ genotype numbers worldwide: An update. Int. J. Chron. Obstruct. Pulmon. Dis. 2017, 12, 561–569. [Google Scholar] [CrossRef] [Green Version]
  4. Blanco, I.; Bueno, P.; Diego, I.; Pérez-Holanda, S.; Lara, B.; Casas-Maldonado, F.; Esquinas, C.; Miravitlles, M. Alpha-1 antitrypsin Pi*SZ genotype: Estimated prevalence and number of SZ subjects worldwide. Int. J. Chron. Obstruct. Pulmon. Dis. 2017, 12, 1683–1694. [Google Scholar] [CrossRef] [Green Version]
  5. Janciauskiene, S.; DeLuca, D.S.; Barrecheguren, M.; Welte, T.; Miravitlles, M. Serum Levels of Alpha1-antitrypsin and Their Relationship with COPD in the General Spanish Population. Arch. Bronconeumol. 2020, 56, 76–83. [Google Scholar] [CrossRef]
  6. Ellis, P.; Turner, A. What Do Alpha-1 Antitrypsin Levels Tell Us About Chronic Inflammation in COPD? Arch. Bronconeumol. 2020, 56, 72–73. [Google Scholar] [CrossRef] [PubMed]
  7. Lomas, D.A. Twenty years of polymers: A personal perspective on alpha-1 antitrypsin deficiency. COPD 2013, 10, 17–25. [Google Scholar] [CrossRef] [PubMed]
  8. Mela, M.; Smeeton, W.; Davies, S.E.; Miranda, E.; Scarpini, C.; Coleman, N.; Alexander, G.J.M. The Alpha-1 Antitrypsin Polymer Load Correlates with Hepatocyte Senescence, Fibrosis Stage and Liver-Related Mortality. Chronic. Obstr. Pulm. Dis. 2020, 7, 151–162. [Google Scholar] [CrossRef] [PubMed]
  9. Bouchecareilh, M. Alpha-1 Antitrypsin Deficiency-Mediated Liver Toxicity: Why Do Some Patients Do Poorly? What Do We Know So Far? Chronic. Obstr. Pulm. Dis. 2020, 7, 172–181. [Google Scholar] [CrossRef]
  10. Teckman, J.H.; Jain, A. Advances in alpha-1-antitrypsin deficiency liver disease. Curr. Gastroenterol. Rep. 2014, 16, 367. [Google Scholar] [CrossRef]
  11. Hamesch, K.; Strnad, P. Non-Invasive Assessment and Management of Liver Involvement in Adults with Alpha-1 Antitrypsin Deficiency. Chronic. Obstr. Pulm. Dis. 2020, 7, 260–271. [Google Scholar]
  12. Schneider, C.V.; Hamesch, K.; Gross, A.; Mandorfer, M.; Moeller, L.S.; Pereira, V.; Pons, M.; Kuca, P.; Reichert, M.C.; Benini, F.; et al. European Alpha-1 Liver Study Group. Liver Phenotypes of European Adults Heterozygous or Homozygous for Pi∗Z Variant of AAT (Pi∗MZ vs Pi∗ZZ genotype) and Noncarriers. Gastroenterology 2020, 159, 534–548. [Google Scholar] [CrossRef]
  13. Clark, V.C.; Dhanasekaran, R.; Brantly, M.; Rouhani, F.; Schreck, P.; Nelson, D.R. Liver test results do not identify liver disease in adults with α(1)-antitrypsin deficiency. Clin. Gastroenterol. Hepatol. 2012, 10, 1278–1283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Del Poggio, P.; Colombo, S. Is transient elastography a useful tool for screening liver disease? World J. Gastroenterol. 2009, 15, 1409–1414. [Google Scholar] [CrossRef] [PubMed]
  15. Kim, R.G.; Nguyen, P.; Bettencourt, R.; Dulai, P.S.; Haufe, W.; Hooker, J.; Minocha, J.; Valasek, M.A.; Aryafar, H.; Brenner, D.A.; et al. Magnetic resonance elastography identifies fibrosis in adults with alpha-1 antitrypsin deficiency liver disease: A prospective study. Aliment. Pharmacol. Ther. 2016, 44, 287–299. [Google Scholar] [CrossRef]
  16. Guillaud, O.; Dumortier, J.; Traclet, J.; Restier, L.; Joly, P.; Chapuis-Cellier, C.; Lachaux, A.; Mornex, J.F. Assessment of Liver Fibrosis by Transient Elastography (Fibroscan®) in Patients with A1AT Deficiency. Clin. Res. Hepatol. Gastroenterol. 2019, 43, 77–81. [Google Scholar] [CrossRef] [PubMed]
  17. Hamesch, K.; Mandorfer, M.; Pereira, V.M.; Moeller, L.S.; Pons, M.; Dolman, G.E.; Reichert, M.C.; Heimes, C.V.; Woditsch, V.; Voss, J.; et al. European Alpha1-Liver Study Group. Liver Fibrosis and Metabolic Alterations in Adults with Alpha1 Antitrypsin Deficiency Caused by the Pi*ZZ Mutation. Gastroenterology 2019, 157, 705–719. [Google Scholar] [CrossRef]
  18. Abbas, S.H.; Pickett, E.; Lomas, D.A.; Thorburn, D.; Gooptu, B.; Hurst, J.R.; Marshall, A. Non-invasive testing for liver pathology in alpha-1 antitrypsin deficiency. BMJ Open. Respir. Res. 2020, 7, e000820. [Google Scholar] [CrossRef] [PubMed]
  19. Sterling, R.K.; Lissen, E.; Clumeck, N.; Sola, R.; Correa, M.C.; Montaner, J.; Sulkowski, M.S.; Torriani, F.J.; Dieterich, D.T.; Thomas, D.L.; et al. Development of a simple noninvasive index to predict significant fibrosis patients with HIV/HCV co-infection. Hepatology 2006, 43, 1317–1325. [Google Scholar] [CrossRef]
  20. Lin, Z.H.; Xin, Y.N.; Dong, Q.J.; Wang, Q.; Jiang, X.J.; Zhan, S.H.; Sun, Y.; Xuan, S.Y. Performance of the aspartate aminotransferase-to-platelet ratio index for the staging of hepatitis C-related fibrosis: An updated meta-analysis. Hepatology 2011, 53, 726–736. [Google Scholar] [CrossRef] [PubMed]
  21. Lichtinghagen, R.; Pietsch, D.; Bantel, H.; Manns, M.P.; Brand, K.; Bahr, M.J. The Enhanced Liver Fibrosis (ELF) score: Normal values, influence factors and proposed cut-off values. J. Hepatol. 2013, 59, 236–242. [Google Scholar] [CrossRef] [PubMed]
  22. European Association for Study of Liver. EASL-ALEH Clinical Practice Guidelines: Non-invasive Tests for Evaluation of Liver Disease Severity and Prognosis. J. Hepatol. 2015, 63, 237–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mulabecirovic, A.; Mjelle, A.B.; Gilja, O.H.; Vesterhus, M.; Havre, R.F. Liver elasticity in healthy individuals by two novel shear-wave elastography systems—Comparison by age, gender, BMI and number of measurements. PLoS ONE 2018, 13, e0203486. [Google Scholar] [CrossRef] [PubMed]
  24. Castera, L. Non-invasive tests for liver fibrosis in NAFLD: Creating pathways between primary healthcare and liver clinics. Liver Int. 2020, 40 (Suppl. 1), 77–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Karlas, T.; Petroff, D.; Sasso, M.; Fan, J.G.; Mi, Y.Q.; de Lédinghen, V.; Kumar, M.; Lupsor-Platon, M.; Han, K.H.; Cardoso, A.C.; et al. Individual patient data meta-analysis of controlled attenuation parameter (CAP) technology for assessing steatosis. J. Hepatol. 2017, 66, 1022–1030. [Google Scholar] [CrossRef] [PubMed]
  26. Clark, V.C.; Marek, G.; Liu, C.; Collinsworth, A.; Shuster, J.; Kurtz, T.; Nolte, J.; Brantly, M. Clinical and histologic features of adults with alpha-1 antitrypsin deficiency in a non-cirrhotic cohort. J. Hepatol. 2018, 69, 1357–1364. [Google Scholar] [CrossRef] [PubMed]
  27. Kümpers, J.; Fromme, M.; Schneider, C.V.; Trautwein, C.; Denk, H.; Hamesch, K.; Strnad, P. Assessment of liver phenotype in adults with severe alpha-1 antitrypsin deficiency (Pi*ZZ genotype). J. Hepatol. 2019, 71, 1272–1274. [Google Scholar] [CrossRef] [Green Version]
  28. Behairy, B.-S.; Sira, M.M.; Zalata, K.R.; Salama, E.-S.E.; Abd-Allah, M.A. Transient elastography compared to liver biopsy and morphometry for predicting fibrosis in pediatric chronic liver disease: Does etiology matter? World J. Gastroenterol. 2016, 22, 4238–4249. [Google Scholar] [CrossRef] [PubMed]
  29. Cox, D.W.; Smyth, S. Risk for liver disease in adults with alpha 1-antitrypsin deficiency. Am. J. Med. 1983, 74, 221–227. [Google Scholar] [CrossRef]
  30. Tanash, H.A.; Piitulainen, E. Liver disease in adults with severe alpha-1-antitrypsin deficiency. J. Gastroenterol. 2019, 54, 541–548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Fairbanks, K.D.; Tavill, A.S. Liver disease in alpha 1-antitrypsin deficiency: A review. Am. J. Gastroenterol. 2008, 103, 2136–2141. [Google Scholar] [CrossRef] [PubMed]
  32. Morer, L.; Choudat, L.; Dauriat, G.; Durand, F.; Cazals-Hatem, D.; Thabut, G.; Brugière, O.; Castier, Y.; Mal, H. Liver involvement in patients with PiZZ-emphysema, candidates for lung transplantation. Am. J. Transplant. 2017, 17, 1389–1395. [Google Scholar] [CrossRef] [Green Version]
  33. Fromme, M.; Oliverius, M.; Strnad, P. DEFI-ALFA: The French key to the alpha1 mystery? Liver Int. 2019, 39, 1019–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Strnad, P.; Buch, S.; Hamesch, K.; Fischer, J.; Rosendahl, J.; Schmelz, R.; Brueckner, S.; Brosch, M.; Heimes, C.V.; Woditsch, V. Heterozygous carriage of the alpha1-antitrypsin Pi*Z variant increases the risk to develop liver cirrhosis. Gut 2019, 68, 1099–1107. [Google Scholar] [CrossRef] [PubMed]
  35. Schaefer, B.; Mandorfer, M.; Viveiros, A.; Finkenstedt, A.; Ferenci, P.; Schneeberger, S.; Tilg, H.; Zoller, H. Heterozygosity for the alpha-1-antitrypsin Z allele in cirrhosis is associated with more advanced disease. Liver Transpl. 2018, 24, 744–751. [Google Scholar] [CrossRef]
  36. Hernández Pérez, J.M.; Blanco, I.; Sánchez Medina, J.A.; Díaz Hernández, L.; Pérez Pérez, J.A. Serum Levels of Glutamate-Pyruvate Transaminase, Glutamate-Oxaloacetate Transaminase and Gamma-Glutamyl Transferase in 1494 Patients with Various Genotypes for the Alpha-1 Antitrypsin Gene. J. Clin. Med. 2020, 9, 3923. [Google Scholar] [CrossRef] [PubMed]
  37. Stoller, J.K.; Tomashefski, J., Jr.; Crystal, R.G.; Arroliga, A.; Strange, C.; Killian, D.N.; Schluchter, M.D.; Wiedemann, H.P. Mortality in individuals with severe deficiency of alpha1-antitrypsin: Findings from the National Heart, Lung, and Blood Institute Registry. Chest 2005, 127, 1196–1204. [Google Scholar] [PubMed]
  38. Tanash, H.A.; Nilsson, P.M.; Nilsson, J.A.; Piitulainen, E. Clinical course and prognosis of never-smokers with severe alpha-1-antitrypsin deficiency (PiZZ). Thorax 2008, 63, 1091–1095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Dawwas, M.F.; Davies, S.E.; Griffiths, W.J.H.; Lomas, D.A.; Alexander, G.J. Prevalence and risk factors for liver involvement in individuals with PiZZ-related lung disease. Am. J. Respir. Crit. Care Med. 2013, 187, 502–508. [Google Scholar] [CrossRef] [PubMed]
  40. Barrecheguren, M.; Torres-Duran, M.; Casas-Maldonado, F.; Miravitlles, M. Spanish implementation of the new international alpha-1 anitrypsin deficiency international registry: The European Alpha-1 Research Collaboration (EARCO). Arch. Bronconeumol. 2021, 57, 81–82. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Comparison of mean LSM values by phenotype.
Figure 1. Comparison of mean LSM values by phenotype.
Jcm 10 01724 g001
Figure 2. All individuals from the cohort with liver enzymes above the highest level of normal based on LSM values. UPN: Upper limit of normal for GGT: >38 IU/L in females and >55 IU/L in males.
Figure 2. All individuals from the cohort with liver enzymes above the highest level of normal based on LSM values. UPN: Upper limit of normal for GGT: >38 IU/L in females and >55 IU/L in males.
Jcm 10 01724 g002
Figure 3. Relation between elevated GGT, FIB4, and LSM in Pi*ZZ patients. GGT: Gamma-glutamyl transferase; FIB-4: Fibrosis 4; LSM: Liver stiffness measurement; UPL: Upper limit of normal (according to sex-specific cut-offs: for GGT: >38 IU/L in females and >55 IU/L in males).
Figure 3. Relation between elevated GGT, FIB4, and LSM in Pi*ZZ patients. GGT: Gamma-glutamyl transferase; FIB-4: Fibrosis 4; LSM: Liver stiffness measurement; UPL: Upper limit of normal (according to sex-specific cut-offs: for GGT: >38 IU/L in females and >55 IU/L in males).
Jcm 10 01724 g003
Table 1. Baseline characteristics of the patients included by AAT genotype.
Table 1. Baseline characteristics of the patients included by AAT genotype.
ZZ (n = 81)Heterozygous Z
(n = 67)
p-Value
Age57.0 (14.4)52.5 (14.5)0.051 1
   Sex, men41 (50.6%)34 (50.7%)0.985 2
BMI25.1 (3.9)24.0 (7.0)0.398 1
Smoking exposure: 0.010 2
   Active43 (53.1%)22 (32.8%)
   Former smoker7 (8.6%)16 (23.9%)
   Never smoker31 (38.3%)29 (43.3%)
Alcohol consumption19 (23.5%)19 (28.4%)0.991 2
Diabetes mellitus0 (0%)2 (3.0%)0.203 2
Hypertension14 (17.5%)16 (23.9%)0.453 2
AAT levels, mg/dL33.3 (61.9)71.9 (20.8)<0.001 1
Reason for diagnosis: 0.002 2
   Liver disease9 (11.1%)13 (19.4%)
   Lung disease52 (64.2%)23 (34.3%)
   Family study17 (21.0%)28 (41.8%)
   Other3 (3.7%)3 (4.5%)
COPD57 (70.4%)15 (22.7%)<0.001 2
Asthma5 (7.8%)14 (21.2%)0.056 2
Neonatal jaundice6 (7.4%)3 (4.5%)0.513 2
FVC, L3.6 (1.5)3.9 (1.1)0.197 1
FVC, %90.0 (28.5)99.8 (19.8)0.033 1
FEV1, L2.1 (1.2)3.0 (1.2)<0.001 1
FEV1, %69.0 (30.5)92.9 (27.6)<0.001 1
FEV1/FVC0.6 (0.2)0.7 (0.2)0.001 1
KCO, %51.0 (32.5)58.9 (36.7)0.231 1
Footnote: BMI: Body mass index; COPD: Chronic obstructive pulmonary disease; FVC: Forced ventilatory capacity; FEV1: Forced expiratory volume in 1 s; KCO: Transfer coefficient of the lung for carbon monoxide; AAT: Alpha-1 antitrypsin. 1 Mann–Whitney U-test p-value, 2 Chi-squared p-value.
Table 2. Results of blood analysis and transient elastography in patients with different AAT genotypes.
Table 2. Results of blood analysis and transient elastography in patients with different AAT genotypes.
ZZ (n = 81)Heterozygous Z
(n = 67)
p-Value
Laboratory findings
Platelet count, ×109/L222 (59)239 (61)0.074 1
INR1.0 (0.2)1.0 (0.1)0.067 1
Bilirubin, mg/dL0.8 (0.5)0.7 (0.3)0.158
AST, IU/L29.2 (15.4)25.0 (8.0)0.029 1
AST > ULN4 (4.9%)4 (6%)0.869 2
ALT, IU/L26.6 (22.6)26.1 (13.4)0.967 1
ALT > ULN6 (7.4%)5 (7.5%)0.952 2
ALP, IU/L78.2 (29.6)81.8 (21)0.412 1
ALP > ULN6 (7.4%)2 (3%)0.294 2
GGT, IU/L36.2 (33.9)31.1 (29.4)0.336 1
GGT > ULN13 (16.5%)9 (13.6%)0.637 2
Albumin, g/dL4.3 (0.6)4.4 (0.3)0.044 1
Cholesterol, mg/dL207 (35)198 (36)0.161
FIB-41.6 (0.8)1.2 (0.5)<0.001
FIB-4 < 1.4538 (47.5%)51 (78.5%)<0.001 2
FIB-4 > 3.255 (6.2%)00.065 2
APRI0.35 (0.18)0.27 (0.09)<0.001 1
APRI < 0.567 (83)64 (91)0.023 2
APRI > 1.01 (1.2)00.956
ELF, n = 608.6 (0.8)8 (0.6)0.007 1
Transient elastography
LSM5.6 (2.4)4.6 (1.2)0.001 1
LSM > 7.5 kPa8 (9.9%)00.040 2
LSM ≥ 10 kPa3 (3.7%)0
CAP256 (59)253 (50)0.252 1
CAP 268–280 dB/m7 (8.6%)4 (6%)0.807 2
CAP > 280 dB/m26 (32.1%)21 (31.3%)
Footnote: INR: International normalized ratio; ULN: Upper limit of normal; AST: Aspartate aminotransferase; ALT: Alanine aminotransferase; ALP: Alkaline phosphatase; GGT: Gamma-glutamyl transferase; FIB-4: Fibrosis 4; APRI: AST to platelet ratio index; ELF: Enhanced liver fibrosis; LSM: Liver stiffness measurement; CAP: Controlled attenuation parameter. 1 Mann–Whitney U-test p-value, 2 Chi-squared p-value.
Table 3. Comparison between Pi*ZZ individuals based on liver stiffness (LSM) and diagnosis of chronic obstructive pulmonary disease (COPD).
Table 3. Comparison between Pi*ZZ individuals based on liver stiffness (LSM) and diagnosis of chronic obstructive pulmonary disease (COPD).
LSM ≤ 7.5
(n = 73)
LSM > 7.5
(n = 8)
p-ValueNo COPD
(n = 24)
COPD
(n = 57)
p-Value
Age56.2 (14.5)64.9 (11.4)0.07646.2 (14.5)61.6 (11.7)<0.001 1
   Sex, men37 (50.7%)4 (50%)1.0011 (45.8%)30 (52.6%)0.752 2
BMI24.6 (3.4)29.0 (5.3)0.05624.2 (3.7)25.4 (3.9)0.186 1
Smoking exposure: 0.527 <0.001 2
   Active37 (50.7%)6 (75%) 7 (29.2%)36 (63.2%)
   Former smoker7 (9.6%)0 (0%) 0 (0%)7 (12.3%)
   Never smoker29 (39.7%)2 (25%) 17 (70.8%)14 (24.6%)
Alcohol consumption16 (24.2%)3 (60%)0.1155 (25%)14 (27.2)1.000 2
Hypertension10 (13.9%)4 (50%)0.0281 (4.2%)13 (23.2%)0.054 2
COPD49 (67.1%)8 (100%)0.097057 (100%)0.001 2
Neonatal jaundice6 (8.2%)0 (0%)1.0004 (16.7%)2 (3.5%)0.060 2
FEV1, %70.4 (30.7)56.4 (27.3)0.20599.6 (13.1)56.2 (26.3)<0.001 1
Laboratory findings:
Platelet count, ×109/L224 (60)202 (49)0.267210 (48)226 (62)0.214 1
INR1.0 (0.2)1.1 (0.1)0.3781.0 (0.1)1.1 (0.2)0.040 1
Bilirubin, mg/dL0.8 (0.5)0.6 (0.2)0.2621.0 (0.9)0.7 (0.2)0.143 1
AST, UI/L27.2 (10.1)47.6 (34.7)0.14127.2 (10.6)30.0 (16.9)0.375 1
AST > ULN *2 (2.7%)2 (25%)0.0481 (4.2%)3 (5.3%)0.675 2
ALT, UI/L24.2 (14.2)48.8 (55.8)0.25425.5 (14.4)27.1 (25.3)0.719 1
ALT > ULN *4 (5.5%)2 (25.0%)0.1083 (12.5%)3 (5.3%)0.226 2
ALP, UI/L78.5 (30.9)75.9 (14.8)0.81670.3 (31)81.4 (28.7)0.130 1
ALP > ULN *6 (8.5%)0 (0%)1.0002 (8.3%)4 (7.0%)1.000 2
GGT, UI/L31.8 (19.3)75.6 (84.1)<0.00133.2 (22.2)37.2 (37.7)0.685 1
GGT > ULN *10 (14.1%)3 (37.5%)0.1205 (20.8%)8 (14%)0.589 2
Albumin, g/dL4.3 (0.6)4.4 (0.3)0.6154.5 (0.3)4.2 (0.6)0.004 1
Cholesterol, mg/dL206 (35)208 (39)0.901205 (39)207 (34)0.824 1
FIB-41.5 (0.8)2.2 (0.7)0.0321.3 (0.8)1.7 (0.8)0.046 1
FIB-4 < 1.45:37 (50.7%)1 (12.5%)0.05915 (62.5%)23 (40.4%)0.077 2
FIB-4 > 3.25:4 (5.5%)1 (12.5%)0.4181 (4.2%)4 (7%)1.000 2
APRI0.33 (0.1)0.56 (0.3)<0.0010.35 (0.17)0.35 (0.19)0.992 1
Transient elastography
LSM5.0 (1.1)10.8 (4.6)0.0095.3 (1.1)5.7 (2.8)0.361 1
CAP249 (56)318 (48)0.004233 (56)266 (58)0.023 1
LSM > 7.5 kPa:08 (100%)NA08 (14.0%)0.097 2
Footnote: BMI: Body mass index; COPD: Chronic obstructive pulmonary disease; FEV1: Forced expiratory volume in 1 s; AAT: Alpha-1 antitrypsin; INR: International normalized ratio; AST: Aspartate aminotransferase; ALT: Alanine aminotransferase; ALP: Alkaline phosphatase; GGT: Gamma-glutamyl transferase; ULN: Upper limit of normal; FIB-4: Fibrosis 4; APRI: AST to platelet ratio index; ELF: Enhanced liver fibrosis; LSM: Liver stiffness measurement; CAP: Controlled attenuation parameter. *: Upper limit of normal according to sex-specific cut-offs: For AST and ALT: >35 IU/L in female, >50 IU/L in male; for ALP: >120 IU/L for both genders; for GGT: >38 IU/L in females and >55 IU/L in males. 1 Mann–Whitney U-test p-value, 2 Chi-squared p-value.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pons, M.; Núñez, A.; Esquinas, C.; Torres-Durán, M.; Rodríguez-Hermosa, J.L.; Calle, M.; Tubio-Pérez, R.; Belmonte, I.; Rodríguez-Frías, F.; Rodríguez, E.; et al. Utility of Transient Elastography for the Screening of Liver Disease in Patients with Alpha1-Antitrypsin Deficiency. J. Clin. Med. 2021, 10, 1724. https://doi.org/10.3390/jcm10081724

AMA Style

Pons M, Núñez A, Esquinas C, Torres-Durán M, Rodríguez-Hermosa JL, Calle M, Tubio-Pérez R, Belmonte I, Rodríguez-Frías F, Rodríguez E, et al. Utility of Transient Elastography for the Screening of Liver Disease in Patients with Alpha1-Antitrypsin Deficiency. Journal of Clinical Medicine. 2021; 10(8):1724. https://doi.org/10.3390/jcm10081724

Chicago/Turabian Style

Pons, Mònica, Alexa Núñez, Cristina Esquinas, María Torres-Durán, Juan Luis Rodríguez-Hermosa, Myriam Calle, Ramón Tubio-Pérez, Irene Belmonte, Francisco Rodríguez-Frías, Esther Rodríguez, and et al. 2021. "Utility of Transient Elastography for the Screening of Liver Disease in Patients with Alpha1-Antitrypsin Deficiency" Journal of Clinical Medicine 10, no. 8: 1724. https://doi.org/10.3390/jcm10081724

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop