2,511
Views
7
CrossRef citations to date
0
Altmetric
Disease management of COPD, AATD and NCFBE: What can we learn from real-life data?

Alpha-1-Antitrypsin Deficiency: Disease Management and Learning from Studies

ABSTRACT

Alpha-1-antitrypsin deficiency (AATD) is one of the most frequent genetic causes of liver and lung diseases. Despite its known association with chronic obstructive pulmonary disease (COPD), AATD is largely unrecognised and underdiagnosed. Cases of AATD exist within every COPD or spirometry population but must be actively investigated. AATD is a laboratory diagnosis that must be confirmed by a blood test. A number of clinical ‘clues’ can raise suspicion of AATD, potentially facilitating earlier diagnosis and initiation of appropriate treatment. Alpha-1-antitrypsin augmentation therapy has a clear role in patients with severe AATD and a FEV1 ≤65% predicted. Emerging evidence suggests that attenuating the decline in lung density may prolong the time to respiratory failure.

Identifying AATD cases

Alpha-1-antitrypsin deficiency (AATD) is an autosomal genetic disorder and one of the most frequent genetic causes of liver and lung diseases Citation(1). Inherited low concentrations of alpha-1-antitrypsin lead to unopposed proteolysis in the lungs, predisposing individuals to early-onset emphysema, which predominantly affects the lower zones of the lungs. Despite its known association with chronic obstructive pulmonary disease (COPD), and the substantial health and economic burden it carries, AATD is largely unrecognised and underdiagnosed Citation(2).

In the COPD population, certain patients may show clinical characteristics or ‘clues’ suggestive of AATD, as illustrated by the example in .

Table 1. Case study of AATD

The main diagnostic clues in this patient are absence of a significant smoking history, extent of lung damage for age, emphysema-predominant disease and the presence of antineutrophil cytoplasmic antibodies (ANCA)-positive vasculitis, which is known to be associated with AATD Citation(3). However, rather than having to rely on clinical clues (which may or may not be present), the American Thoracic Society/European Respiratory Society have issued clear recommendations to screen every COPD patient for AATD once in his/her lifetime Citation(4). AATD is a laboratory diagnosis that must be confirmed by a blood test. A proactive approach is therefore essential to identify cases and differentiate them from COPD. Case finding or targeted detection in at-risk groups is considered to be the most efficient method of identifying individuals most likely to benefit from management and treatment strategies for AATD.

AATD testing is recommended in the following populations Citation(4):

Symptomatic adults with persistent obstruction with emphysema, COPD and asthma;

Asymptomatic individuals with persistent obstruction with risk factors (smoking and exposure);

Individuals with unexplained liver disease;

Adults with necrotising panniculitis;

Family screening.

AATD prevalence

The true prevalence of AATD is difficult to determine due to the absence of population-based newborn screening studies in most world countries. The best approach to estimating prevalence identified to date has been to limit analysis to studies with adequate methodologies that examined control group cohorts (e.g. blood donors, workers, healthy unrelated persons). Using this approach, it must be accepted that true prevalence will be underestimated as individuals with disease are generally excluded from control group populations. Blanco and colleagues identified 68 cohorts comprising 75,390 individuals from 21 countries in Europe; about 125,000 individuals were estimated to carry the PiZZ genotype Citation(5). Investigating for AATD in COPD populations would be expected to yield higher estimates and, indeed, applying the estimated 0.63% prevalence of severe AAT deficiency in patients with fixed airflow obstruction (GOLD II–IV) reported by US investigators Citation(6) to the overall European population indicated a potential PiZZ genotype population of about 400,000 individuals. The actual prevalence of AATD probably lies somewhere between these estimates. Among COPD populations, there are almost certainly to be cases of AATD, but these cases must be actively investigated to reduce the diagnostic delay from the current 5–8 years Citation(2).

AATD knowledge relates to testing frequency

The failure to identify all patients with AATD relates in large part to a knowledge gap, which translates into a testing gap. German and Italian physicians from the specialties of pulmonology, internal medicine and general practice were surveyed about their knowledge of AATD and testing frequency Citation(7). Pulmonologists ranked highest for knowledge and testing frequency, whereas comparatively poor results for physicians from internal medicine and general practice () highlighted the need to increase knowledge and awareness of AATD among other specialties likely to encounter patients with respiratory diseases.

Figure 1. Self-reported level of knowledge about alpha-1-antitrypsin deficiency (AATD) and AATD testing frequency among pulmonologists (Pu), internal medicine specialists (In) and general practitioners (GP) from Germany (n = 180) and Italy (n = 181). Constructed from Citation(7).

Figure 1. Self-reported level of knowledge about alpha-1-antitrypsin deficiency (AATD) and AATD testing frequency among pulmonologists (Pu), internal medicine specialists (In) and general practitioners (GP) from Germany (n = 180) and Italy (n = 181). Constructed from Citation(7).

Underdiagnosis of AATD is a global issue. A study from Spain reported that, despite nearly 7,000 new cases of COPD diagnosed in the Catalan region each year from 2007 to 2012, less than 400 AAT tests per year in COPD patients were performed Citation(8,9). In Germany and Italy, only 18–25% of surveyed physicians (pulmonologists, internal medicine specialists and general practitioners) reported performing AATD testing in their COPD populations Citation(7). On the other hand, and in line with current recommendations, testing frequency was considerably greater in at-risk populations such as those diagnosed with COPD at <45 years of age (83 and 77%, respectively) and in family members of patients with AATD (69 and 72%, respectively) Citation(7).

Differentiating AATD and COPD

In addition to registry populations, several other sources can be used to estimate AATD prevalence in the general population. Our group in Marburg, Germany, interrogated a national insurance database for the International Classification of Diseases (ICD) code E88.0, which classifies cases of AATD (and bisalbuminaemia) Citation(10). Approximately 20,000 individuals with AATD were identified, inclusive of homozygous and heterozygous genotypes. To verify the internal validity of these estimates, we then investigated the prevalence of conditions known to be associated with AATD (vasculitis, chronic hepatitis, liver cirrhosis and panniculitis) among patients with AATD (ICD code E88.0) or COPD Citation(11). Chronic hepatitis and cirrhosis were significantly more prevalent in patients with AATD than in those with COPD. There was a trend towards more vasculitis in AATD patients, whereas panniculitis was associated solely with COPD ().

Figure 2. Prevalence of associated comorbidities in patients with alpha-1-antitrypsin deficiency (AATD) and chronic obstructive pulmonary disease (COPD) identified from a national insurance database in Germany. Reproduced from Citation(11) with permission.

Figure 2. Prevalence of associated comorbidities in patients with alpha-1-antitrypsin deficiency (AATD) and chronic obstructive pulmonary disease (COPD) identified from a national insurance database in Germany. Reproduced from Citation(11) with permission.

Compared with an age- and gender-matched reference population, AATD patients were found to have significantly more comorbidities, including arterial hypertension, congestive heart failure, ischaemic heart disease, chronic kidney disease, depression, diabetes, osteoporosis, gastroesophageal reflux disease and lung cancer (all < 0.001), although the analysis incorporated a degree of selection bias as AATD testing was performed in patients already with disease Citation(11). Patients with AATD typically have a more severe clinical course compared to those with other obstructive lung diseases, as evidenced by the significantly higher frequency of annual outpatient consultations and hospitalisations () observed in AATD patients compared to those with COPD, emphysema and asthma Citation(11).

Figure 3. Percentage of hospitalisations in Germany according to disease type: alpha-1-antitrypsin deficiency (AATD), chronic obstructive pulmonary disease (COPD), emphysema or asthma. Reproduced from Citation(11) with permission.

Figure 3. Percentage of hospitalisations in Germany according to disease type: alpha-1-antitrypsin deficiency (AATD), chronic obstructive pulmonary disease (COPD), emphysema or asthma. Reproduced from Citation(11) with permission.

Treatment of AATD

In terms of treating patients with AATD, smoking cessation (as required) is a very clear recommendation supported by good quality evidence. With regard to other treatments such as pulmonary rehabilitation, vaccination (influenza and pneumococcal), inhaled therapy and endoscopic lung volume reduction, scant data are available but AATD may be subsumed under the umbrella term COPD.

AAT augmentation therapy

The benefits of treating AATD patients with alpha-1-antitrypsin (AAT) augmentation therapy were first documented in analyses of patient registries. A comparison of the annual change in FEV1 between a German group who received weekly infusions of AAT 60 mg/kg and an untreated Danish group showed a significant treatment effect in the treated subgroup with a baseline FEV1 31–65% predicted Citation(12). Similar trends were observed in the National Heart, Lung, and Blood Institute Registry study Citation(13).

The role of computed tomography (CT) densitometry in assessing the effects of augmentation therapy in AATD patients was explored in the EXAcerbations and Computed Tomography scan as Lung Endpoints (EXACTLE) trial Citation(14). AATD patients (n = 77) were randomised to receive weekly infusions of AAT 60 mg/kg or placebo for up to 2.5 years. The mean decline in lung density was significantly attenuated in the group receiving AAT replacement therapy versus the placebo-treated group, although the differences did not relate to clinical outcomes. A subsequent analysis pooled EXACTLE data with those from a comparably designed Danish Dutch study (n = 54) Citation(15) to increase the statistical power Citation(16). Based on densitometric data from 119 patients, the mean decline in lung density from baseline to last CT scan was significantly less with AAT augmentation therapy than with placebo (−4.08 vs −6.38; p = 0.006).

To substantiate the benefits of AAT augmentation therapy as suggested by the pooled analysis, a large international study was conducted. In the double-blind, multicentre, phase III/IV RAPID Lung Density study, 180 adult patients with AATD-related emphysema and FEV1 35–70% predicted were randomised to receive AAT 60 mg/kg weekly or placebo for 24 months, followed by a 24-month extension phase in which all patients received open-label AAT treatment Citation(17). At the end of double-blind treatment, the annual rate of lung density loss (at total lung capacity) was significantly lower in the AAT group (−1.45 vs −2.19 g/L with placebo). During the extension phase of the study, the magnitude of lung density decline decreased in placebo-treated patients switched to open-label treatment, such that it aligned with that of patients treated with AAT from the beginning of the study and was in the range of that reported in the ECLIPSE study Citation(18). Nevertheless, analysis of secondary endpoints of the RAPID Lung Density study showed no discernible effects for AAT augmentation therapy on FEV1 or quality of life Citation(17).

Whether attenuating lung function decline is a clinically meaningful outcome was addressed by an observational study of the AATD registry in the United Kingdom Citation(19). For analysis purposes, augmentation-naïve PiZZ patients with two successive quantitative CT scans (n = 110) were divided into two groups: those with a decline in CT density and those with stable CT density. Even after adjusting for baseline lung density, a decline in lower zone lung density was associated with significantly decreased survival (p = 0.048), with the difference being driven mainly by patients with FEV1 >30% predicted ().

Figure 4. Survival curves in augmentation-naïve alpha-1-antitrypsin deficiency patients with a decline in CT density (▪) or stable CT density (•), based on two successive quantitative CT scans. Adapted from Citation(19) with permission.

Figure 4. Survival curves in augmentation-naïve alpha-1-antitrypsin deficiency patients with a decline in CT density (▪) or stable CT density (•), based on two successive quantitative CT scans. Adapted from Citation(19) with permission.

Collectively, the studies suggest that it is reasonable to treat AATD patients with augmentation therapy with the aim of stabilising lung density decline. However, patient selection is important. The role of augmentation therapy is clear in patients with severe AATD (AAT levels <11 μM) and a FEV1 ≤65% predicted. In patients with AATD-related lung disease and FEV1 >65% predicted, there is less evidence at present to support augmentation therapy Citation(20). Other factors such as biology, competing risk factors, CT appearance and disease activity must be considered. Individuals with a less significant smoking history, more severe emphysema, those who are rapid decliners (FEV1 and KCO) or have severe disease in relation to age are likely to benefit most from augmentation therapy. Individual patient discussions and shared decision making are essential.

Conclusions

Cases of AATD exist within every COPD or spirometry population but must be actively investigated. All patients with COPD, and particularly those who require more medical attention, should be examined closely to identify a specific AATD endotype. In appropriate patients, use of intravenous AAT augmentation therapy appears to prolong the time to respiratory failure.

Declaration of Interest

TG has received speaker's fees from AstraZeneca, Boehringer-Ingelheim, Chiesi, CSL-Behring, Grifols, GSK, Mundipharma and Novartis.

Editorial assistance was provided by Content Ed Net (Madrid, Spain) with funding from Grifols SA (Barcelona, Spain).

References

  • Silverman EK, Sandhaus RA. Clinical practice. Alpha1-antitrypsin deficiency. N Engl J Med 2009; 360(26):2749–2757.
  • Greulich T, Vogelmeier CF. Alpha-1-antitrypsin deficiency: increasing awareness and improving diagnosis. Ther Adv Respir Dis 2016; 10(1):72–84.
  • Lyons PA, Rayner TF, Trivedi S, Holle JU, Watts RA, Jayne DR, et al. Genetically distinct subsets within ANCA-associated vasculitis. N Engl J Med 2012; 367(3):214–223.
  • American Thoracic Society; European Respiratory Society. American Thoracic Society/European Respiratory Society statement: standards for the diagnosis and management of individuals with alpha-1 antitrypsin deficiency. Am J Respir Crit Care Med 2003; 168(7):818–900.
  • Blanco I, de Serres FJ, Fernandez-Bustillo E, Lara B, Miravitlles M. Estimated numbers and prevalence of PI*S and PI*Z alleles of alpha1-antitrypsin deficiency in European countries. Eur Respir J 2006; 27(1):77–84.
  • Rahaghi FF, Sandhaus RA, Brantly ML, Rouhani ML, Campos MA, Strange C, et al. The prevalence of alpha-1 antitrypsin deficiency among patients found to have airflow obstruction. COPD 2012; 9(4):352–358.
  • Greulich T, Ottaviani S, Bals R, Lepper PM, Vogelmeier C, Luisetti M, et al. Alpha1-antitrypsin deficiency – diagnostic testing and disease awareness in Germany and Italy. Respir Med 2013; 107(9):1400–1408.
  • Barrecheguren M, Monteagudo M, Simonet P, Llor C, Rodriguez E, Ferrer J, et al. Diagnosis of alpha-1 antitrypsin deficiency: a population-based study. Int J Chron Obstruct Pulmon Dis 2016;11:999–1004.
  • Barrecheguren M, Monteagudo M, Ferrer J, Borrell E, Llor C, Esquinas C, et al. Treatment patterns in COPD patients newly diagnosed in primary care. A population-based study. Respir Med 2016; 111:47–53.
  • Greulich T, Nell C, Herr C, Vogelmeier C, Kotke V, Weidmann S, et al. Results from a large targeted screening program for alpha-1-antitrypsin deficiency: 2003–2015. Orphanet J Rare Dis 2016; 11(1):75.
  • Greulich T, Nell C, Hohmann D, Grebe M, Janciauskiene S, Koczulla AR, Vogelmeier CF. The prevalence of diagnosed α1-antitrypsin deficiency and its comorbidities: results from a large population-based database. Eur Respir J 2017; 49(1). pii:1600154.
  • Seersholm N, Wencker M, Banik N, Viskum K, Dirksen A, Kok-Jensen A, et al. Does alpha1-antitrypsin augmentation therapy slow the annual decline in FEV1 in patients with severe hereditary alpha1-antitrypsin deficiency? Wissenschaftliche Arbeitsgemeinschaft zur Therapie von Lungenerkrankungen (WATL) alpha1-AT study group. Eur Respir J 1997; 10(10):2260–2263.
  • Survival and FEV1 decline in individuals with severe deficiency of alpha1-antitrypsin. The Alpha-1-Antitrypsin Deficiency Registry Study Group. Am J Respir Crit Care Med 1998; 158(1):49–59.
  • Dirksen A, Piitulainen E, Parr DG, Deng C, Wencker M, Shaker SB, et al. Exploring the role of CT densitometry: a randomised study of augmentation therapy in alpha1-antitrypsin deficiency. Eur Respir J 2009; 33(6):1345–1353.
  • Dirksen A, Dijkman JH, Madsen F, Stoel B, Hutchison DC, Ulrik CS, et al. A randomized clinical trial of alpha 1-antitrypsin augmentation therapy. Am J Respir Crit Care Med 1999; 160:1468–1472.
  • Stockley RA, Parr DG, Piitulainen E, Stolk J, Stoel BC, Dirksen A. Therapeutic efficacy of α-1 antitrypsin augmentation therapy on the loss of lung tissue: an integrated analysis of 2 randomised clinical trials using computed tomography densitometry. Respir Res 2010; 11:136.
  • Chapman KR, Burdon JG, Piitulainen E, Sandhaus RA, Seersholm N, Stocks JM, et al. Intravenous augmentation treatment and lung density in severe α1 antitrypsin deficiency (RAPID): a randomised, double-blind, placebo-controlled trial. Lancet 2015; 386(9991):360–368.
  • Coxson HO, Dirksen A, Edwards LD, Yates JC, Agusti A, Bakke P, et al. The presence and progression of emphysema in COPD as determined by CT scanning and biomarker expression: a prospective analysis from the ECLIPSE study. Lancet Respir Med 2013; 1(2):129–136.
  • Green CE, Parr DG, Edgar RG, Stockley RA, Turner AM. Lung density associates with survival in alpha 1 antitrypsin deficient patients. Respir Med 2016; 112:81–87.
  • Sandhaus RA, Turino G, Brantly ML, Campos M, Cross CE, Goodman K, et al. The diagnosis and management of alpha-1 antitrypsin deficiency in the adult. Chronic Obstr Pulm Dis (Miami) 2016; 3(3):668–682.