Chronic Obstructive Pulmonary Diseases:Journal of the COPD Foundation

Running Head: Alpha-1 Antitrypsin Deficiency Clinical Practice Guidelines

Funding support: The writing of this document was supported by the Alpha-1 Foundation, Coral Gables, Florida

Date of acceptance: February 4, 2016

Abbreviations: alpha-1 antitrypsin deficiency, AATD; American Thoracic Society, ATS; European Respiratory Society, ERS; chronic obstructive pulmonary disease, COPD; alpha-1 antitrypsin, AAT; proteinase inhibitor genotype with 2 ZZ alleles, Pi*ZZ; COPD related to AATD, AATD-COPD; COPD in the absence of AATD, AAT-replete COPD; granulomatosis with polyangiitis, GPA; polymerase chain reaction, PCR; proteinase inhibitor, Pi; forced expiratory volume in 1 second, FEV1; computed tomography, CT; aspartate aminotransferase, AST; alanine aminotransferase, ALT; gamma-glutamyltransferase, GGT; international normalized ratio, INR; U.S. Food and Drug Administration; FDA; total lung capacity, TLC

Citation: Sandhaus RA, Turino G, Brantly ML, et al. The diagnosis and management of alpha-1 antitrypsin deficiency in the adult. Chronic Obstr Pulm Dis. 2016; 3(3): 668-682. doi: http://doi.org/10.15326/jcopdf.3.3.2015.0182

This article is a statement of the Medical and Scientific Advisory Committee of the Alpha-1 Foundation and has an online data supplement. This is a clinician-centric article.

Summary of Recommendations

Testing for Alpha-1 Antitrypsin Deficiency (AATD):

  • All individuals with COPD regardless of age or ethnicity should be tested for AATD.
  • All individuals with unexplained chronic liver disease should be tested for AATD.
  • All individuals with necrotizing panniculitis, granulomatosis with polyangiitis, or unexplained bronchiectasis should be tested for AATD.
  • Parents, siblings, and children, as well as extended family of individuals identified with an abnormal gene for AAT, should be provided genetic counseling and offered testing for AATD (see guideline document for special considerations about testing minors).
  • For family testing after a proband is identified, AAT level testing alone is not recommended because it does not fully characterize disease risk from AATD.
  • For diagnostic testing of symptomatic individuals, we recommend genotyping for at least the S and Z alleles. Advanced or confirmatory testing should include Pi-typing, AAT level testing, and/or expanded genotyping.

Pulmonary function testing in those with AATD:

  • Initial evaluation with complete lung function testing is recommended.
  • Annual follow-up of adults with at least a spirometry test is recommended.

Computed Tomography (CT) scan of the chest in the evaluation in those with AATD:

  • In newly diagnosed patients who are symptomatic and/or have abnormal pulmonary function testing, a baseline CT scan of the chest is recommended.
  • Serial chest CT scanning to monitor progression of disease is not recommended.

Monitoring for liver disease in those with AATD:

  • Monitoring for liver disease at annual intervals (or more frequently as indicated by clinical circumstances), with physical examination including focused exam for signs of liver disease, liver ultrasound, and laboratory monitoring of AST, ALT, GGT, albumin, bilirubin, INR, and platelets is recommended.

Management of lung disease in those with AATD:

  • Every effort should be made to prevent exposure to tobacco smoke and facilitate cessation in those who are smoking.
  • Lung volume reduction surgery is not recommended for individuals with COPD related to AATD.

Intravenous augmentation therapy in those with AATD is recommended for:

  • Individuals with an FEV1 less than or equal to 65% predicted.
    • For those with lung disease related to AATD and an FEV1 greater than 65%, we recommend discussion with each individual regarding the potential benefits of reducing lung function decline with consideration of the cost of therapy and lack of evidence for such benefit.
  • Individuals with necrotizing panniculitis.

Intravenous augmentation therapy is not recommended for:

  • Individuals with the MZ genotype of AATD.
  • Individuals with lung disease due to AATD who continue to smoke.
  • Individuals with AATD and emphysema or bronchiectasis who do not have airflow obstruction.
  • The treatment of liver disease due to AATD.
  • Individuals who have undergone liver transplantation.

Additional recommendations regarding dosing of intravenous augmentation therapy:

  • Weekly doses higher than the current FDA-approved dose are not recommended.
  • Monitoring of trough AAT blood levels to evaluate the adequacy of AAT augmentation dosing is not recommended.
 

Introduction

Alpha-1 antitrypsin deficiency (AATD), also known as alpha-1 proteinase inhibitor deficiency, is a genetic condition that leads to increased risk of lung and liver disease and several other conditions. The spectrum of AATD-related disease and the age at clinical onset is quite broad. Individuals with AATD may lead healthy lives without any of these medical conditions, but factors such as smoking, occupational exposure to dust and fumes, and some liver insults can increase the likelihood of disease.

The molecular mechanisms leading to lung disease and liver disease are different. AATD lung disease is due to the relative deficiency in the blood and lungs of the alpha-1 antitrypsin (AAT) protein, a major circulating serine proteinase inhibitor. Although recent evidence suggests a more complicated cascade of proteolytic and inflammatory factors as the cause of emphysema in AATD, unopposed neutrophil elastase activity within the pulmonary interstitium with resultant connective tissue destruction remains an important contributor to the pathogenesis of emphysema. In addition, neutrophil elastase has been implicated in the mucus hypersecretion associated with chronic bronchitis. There is a high prevalence of anatomic bronchiectasis in individuals with AATD, with some individuals manifesting signs and symptoms of clinical bronchiectasis. Overall, the pulmonary manifestations of AATD include the entire spectrum of disorders associated with chronic obstructive pulmonary disease (COPD).

The deficiency of circulating AAT in the most common form of clinically relevant AATD (PI*ZZ type) is caused by the accumulation of AAT protein within hepatocytes, the major site of synthesis of this protein. This accumulation is due to misfolding and polymerization of AAT within the hepatocyte rough endoplasmic reticulum and this accumulation is associated with an increased risk of liver disease. AAT polymers formed or trapped in the lung are pro-inflammatory and may contribute to the pulmonary disease of AATD. While the lung disease of AATD is generally an adult-onset condition, the liver disease of AATD can present from birth through old age. The fulminant liver failure occurring in infants with AATD is thought to have a different mechanism than the cirrhosis seen in some adults with AATD.

Several publications have clarified the likely prevalence of AATD in the United States and Europe as between 1:2500 and 1:5000 PI*ZZ in the entire population and emphasize that only a small minority of affected individuals have been diagnosed.3,4 In addition, there is often a significant delay in the diagnosis of AATD from the time that symptoms first appear. Finally, there appears to be an increased prevalence of abnormal AAT genotypes among those with COPD or chronic liver disease.

In 2003, the “ATS/ERS: Standards for the Diagnosis and Management of Individuals with Alpha-1 Antitrypsin Deficiency”2 was published. This 83-page document provided a comprehensive literature review and graded recommendations for identifying and treating individuals with AATD. Unfortunately, despite an accompanying executive summary, the length and density of the document has undermined its widespread clinical application and impact. Both to update the 2003 document with more recently published findings and to encourage review and use of these recommendations more widely, the current committee has sought to provide a much more concise and usable document, backed by online documentation. It is the committee’s hope that the clinical impact will be enhanced with this new format.

There are many unmet needs in the clinician community. We recognize that there is need for a clinician-centric, easily accessible, literature-based summary of the current clinical practice among clinician scientists with experience in the evaluation and treatment of AATD. In concert with Voltaire’s quote that “perfection must not be the enemy of the good,” where gaps in the literature exist, expert opinion may be substituted until a more evidence-based approach can be applied. Educated guidance in this setting is better than no guidance.

Methods

A health sciences librarian (S.K.) used medical subject headings and text words to search for articles referencing AATD or any related synonyms (See online data supplement Table 1). The Ovid platform was used to search MEDLINE, EMBASE, the Cochrane Central Register of Controlled Trials , the Database of Abstracts of Reviews of Effects, and the Cochrane Database of Systematic Reviews. The peer-reviewed search was limited to human studies and articles in English or in any language with English abstracts. Literature indexed since the publication of the previous standards document was searched for the period January 2002 through December 2014. The panel reviewed an additional set of older and newer literature identified as important by members of the writing committee. Additionally, members were asked to cite important papers from 2015.

The chairs reviewed citations for relevance based on titles and abstracts. Writing groups were established from the principal categories of clinical questions. Chairs and committee members sorted the remaining citations into non-discrete sets for each clinical question. Full text was assessed for risk of bias. If more than one writing committee used the same citation, screening was compared and reconciled.

Through 2014, no definitive randomized controlled trials had been published. A meta-analysis of 2 pilot studies was published and reviewed but was felt to have major methodologic shortcomings. However, just prior to finalizing this document, a large, randomized, placebo-controlled, blinded study of intravenous augmentation therapy was published5 and this study has been included in our deliberations.

Each recommendation is rated by the strength of the opinion (strong, medium, weak) and the quality of evidence (high, moderate, low). The strength of the recommendation was determined by voting among the writing committee and a strong recommendation, even in the face of weak evidence, reflects a broad consensus among the expert scientists and clinicians within the writing committee. In situations where there was a lack of consensus, a minority opinion was included following the recommendation.

In organizing this document, we have adopted several nomenclature conventions. Individuals with COPD due to AATD are referred to as having AATD-COPD and those with COPD without AATD are said to have AAT-replete COPD. The clinical questions are divided into 3 groups:

  1. Detection, which refers to strategies to identify individuals with AATD among various at-risk populations;
  2. Diagnostic Testing, which refers to the actual tests used (e.g., genotyping) and to the sequence of tests deployed to secure a diagnosis of AATD;
  3. Clinical Evaluation and Treatment. Within the Treatment section, the order of clinical questions progresses from those regarding general therapy of COPD and liver disease in individuals with AATD to issues regarding the specific management of AATD (e.g., augmentation therapy).

Finally, in the context that the document discusses AATD that is ascertained both in those with symptoms (who may already be under clinical care) and in those without symptoms and not yet recognized, we refer to those with AATD as individuals rather than as patients. We refer to the infusion of purified pooled human plasma alpha-1 antitrypsin as augmentation therapy. Lastly, the discussion of each clinical question is followed by several key references when appropriate. As this document updates the prior 2003 ATS/ERS standards document, references cited in the body of this document are those published since 2003 whenever possible.

For the complete literature evaluation results and methodology, please refer to the online data supplement for these guidelines.

Questions

1. Should individuals be tested for AATD?

In general, the reason for making a specific diagnosis is that the management of the patient or of family members is impacted by the test result. Examples of relevant impacts include changing how patients are monitored; how patients are counseled, e.g., with regard to lifestyle or genetic risks; or how patients are treated. Testing methods may vary by the specific clinical context in which the testing is done. For example, testing strategies for population-based screening, if appropriate, will differ from those used for targeted detection or clinical case-finding. These general considerations inform many of the specific clinician-centric questions that arise regarding AATD. In the sections that follow, specific testing-related clinical questions are posed, with responses, the related evidence, and the grading of the recommendation.

Should specific populations be tested for AATD?

Background: The goal of testing is to identify individuals and at-risk families for whom interventions might confer benefit. Populations enriched for AATD include families of affected individuals, people with COPD, and people with liver disease. As AATD is a genetic disorder, testing of first-degree relatives is certainly logical. In general, deficient family members are healthy or less severely affected or symptomatic compared to the proband, also called the index case. It is less clear how deep into an affected individual’s pedigree such testing should extend, although identifying additional family members with AATD would prompt a new round of testing of their first-degree relatives. Population-based screenings performed thus far have been of limited size and scope. Prevalence estimates may vary by ancestry and ethnicity of a population.

Genetic counseling provides individuals with the opportunity to make an informed decision about being tested and the potential consequences of results. Genetic counseling can be provided by a well-informed primary provider.

Recommendation 1a: All individuals with COPD, regardless of age or ethnicity, should be tested for AATD (strong recommendation, moderate quality of evidence).

Rationale: High value is placed on understanding the etiology of COPD in a given individual and the potential for providing specific therapy. Low value is placed on the high cost of testing. Testing of adults should not be limited on the basis of age, gender, or race. There are no specific demographic or clinical characteristics that rule out the diagnosis of AATD. For example, while there is a lower prevalence of AATD in African American and Asian populations, such individuals with consistent clinical features should be tested for AATD.

Key references:6-8

Recommendation 1b: All individuals with unexplained chronic liver disease should be tested for AATD (strong recommendation, low quality of evidence).

Rationale: High value is placed on understanding disease etiology. Low value is placed on the lack of AATD specific therapy for these conditions.

Key reference:9

Recommendation 1c: All individuals with necrotizing panniculitis, granulomatosis with polyangiitis (GPA, formerly Wegener’s granulomatosis), or unexplained bronchiectasis should be tested for AATD (strong recommendation, low quality of evidence).

Rationale: High value is placed on understanding disease etiology. Low value is placed on lack of AATD specific therapy for some of these conditions.

Key references: 10-13

Recommendation 1d: Adult siblings of individuals identified with an abnormal gene for AAT, whether heterozygote or homozygote, should be provided with genetic counseling and offered testing for AATD (strong recommendation, moderate quality evidence).

Recommendation 1e: Parents, children, minor siblings and the extended family of individuals identified with an abnormal gene for AAT should be provided genetic counseling and offered testing for AATD (weak recommendation, low quality of evidence).

Rationale for 1d and 1e: High value is placed on knowledge gained for family members to allow for risk modification (smoking avoidance) and proper monitoring. Low value is placed on the potential negative aspects of genetic testing (anxiety, insurability, feelings of guilt).

Remarks:The role of testing in the pediatric age group, especially in children without overt liver disease, is not well studied and not supported by current literature or experience. Parental testing is a potential alternative to testing of younger children to predict risk of deficient genotype. The range of serum AAT levels among individuals with specific genotypes is quite broad and therefore overlap between different genotypes can exist. Thus, family testing should be done with a genotype. The services of an AATD-aware genetic counselor may be appropriate.

The Genetic Information Non-discrimination Act was passed in 2008. This act is intended to protect individuals’ insurability (with respect to health insurance, but not life insurance) and employment from discrimination based on genetic information. Counseling family members about the benefits versus risks of genetic testing should occur. Testing of family members should be offered at each clinic visit until all who want testing have received it. Options for confidential testing of family members are available and should be discussed as appropriate. Specific information on such programs is available on the Alpha-1 Foundation website (www.alpha1.org).

Key references:7,14,15

Should AAT levels be used for the primary diagnosis of AATD?

Background: More than 95% of all severely AAT deficient individuals have either the ZZ or SZ genotype. Recent evidence indicates that MZ individuals who smoke are at increased risk for airflow obstruction. In this context the Z allele is the single most frequent genetic risk factor for airflow obstruction. The most sensitive and specific method for identification of individuals at risk for inherited airflow obstruction is direct identification of the Z allele by genotyping (e.g., by using polymerase chain reaction [PCR] probes for the Z allele). Genotyping panels that also include the S allele identify SZ individuals. Genotyping for the S and Z alleles is greater than 99% specific and sensitive for these alleles.

AAT levels are insufficient to identify at risk individuals because the AAT level changes with inflammation, pregnancy, and in children. General recommendations of the American College of Medical Genetics16 suggest that confirmatory testing should be considered using a second method. In AATD testing, the most widely used confirmatory methods are proteinase inhibitor (Pi) typing and/or AAT genotyping. More advanced testing, such as expanded genotyping for rare AAT alleles and gene sequencing, may also be considered confirmatory testing. The type of primary AAT testing must reflect the clinical circumstance under which the testing is done (targeted detection versus screening).

Recommendation 1f: For family testing after a proband is identified, AAT level testing alone is not recommended because it does not fully characterize disease risk from AATD.

Recommendation 1g: For diagnostic testing of symptomatic individuals, we recommend genotyping for at least the S and Z alleles. Advanced or confirmatory testing should include Pi-typing, AAT level testing, and/or expanded genotyping.

Rationale: High value is placed on accurate testing and minimization of false negative results. Low value is placed on the cost of testing. Positive results should be confirmed using an independent testing method. Test kits that use a combination of 2 or more methods (e.g., level and genotyping) have advantages over a single testing methodology. The range of serum AAT levels among individuals with specific genotypes is sufficiently broad that there is overlap between different genotypes. Thus, serum AAT levels cannot discriminate between different genotypes and additional AAT testing is needed. Finally, the rare Null genotypes (genotypes that lead to the production of no or truncated AAT protein) should always be considered when there is discordance between level, and genotype or Pi-type, especially since unintended paternity/maternity issues can arise in this setting.

Key references:17,18

2. Should the clinical evaluation of individuals with AATD be different from those without AATD?

Differences in clinical evaluation between patient groups are justifiable only to the extent that the results will impact the specific management of the patient. AATD is associated with an increased risk of a variety of conditions such as emphysema, bronchiectasis, liver disease, GPA, and panniculitis. Thus, the clinical evaluation of an individual with AATD should pay special attention to the early detection and follow-up of associated conditions. Emphasis on these known conditions might lead to different clinical approaches such as a focus on diffusing capacity or chest CT of the patient with COPD associated with AATD compared to the workup of the patient with AAT-replete COPD.

Should complete pulmonary function testing be part of the evaluation of individuals with AATD?

Background: While more readily available, spirometry alone (forced expiratory measurements of volume and flow) may not accurately reflect the degree of parenchymal destruction associated with AATD-related pulmonary emphysema. There are well-documented cases of individuals with significant emphysema and little or no impairment in spirometry. Measurements of static lung volumes and gas transfer (diffusing capacity) can enhance the detection and assessment of extent of, usually panlobular, emphysema.

Recommendation 2a: Initial evaluation with complete lung function testing is recommended for all individuals with AATD (weak recommendation, low quality evidence).

Minority opinion: Spirometry may be sufficient in asymptomatic individuals with AATD.

Rationale: High value is placed on establishing an initial assessment of all aspects of pulmonary function. Low value is placed on the cost of the testing. Although they reflect different aspects of the same pathological process (emphysema), expiratory flow (forced expiratory volume in 1 second, [FEV1]), hyperinflation, and diffusing capacity are not always well correlated with one another, and these should be determined when assessing the overall severity of pulmonary impairment in individuals with pulmonary symptoms and AATD.

Should individuals with AATD have follow-up pulmonary function testing to monitor for progression of disease?

Background: Some individuals with AATD have normal pulmonary function at the time of diagnosis. The natural history of these individuals is not completely understood, though some data suggest that PI*ZZ never smokers who are asymptomatic have a normal expected survival.19

Recommendation 2b: Follow-up of adults with AATD with normal baseline spirometry should include annual spirometry (strong recommendation, low quality evidence).

Minority opinion: No study has assessed the value of annual spirometry (e.g., compared with other testing strategies) as the optimal method or interval for monitoring.

Rationale: High value is placed on detecting the accelerated rate of lung function loss early in the disease process. Low value is placed on the cost of lung function testing. As AATD lung disease is characterized by accelerated lung function loss, the early detection of the development of obstructive lung disease allows proper risk avoidance, implementation of COPD management, and consideration of specific therapy for AATD. Since the lung disease associated with AATD often starts as purely parenchymal destruction, more complete pulmonary function testing (including measures of diffusing capacity) may be considered.

Key references:20

Should a baseline computed tomography (CT) scan of the chest be included in the evaluation of those with AATD? Should individuals with AATD get chest CT scans at regular intervals to assess disease status?

Background: Studies have demonstrated that decreased lung density on chest CT directly reflects the lung tissue loss in pulmonary emphysema and correlates with mortality. In addition, clinical studies have shown a slowing of the rate of this density loss with intravenous augmentation therapy administration compared with placebo infusions. However, CT scanning exposes individuals to radiation. Furthermore, how the clinician should respond to the results of the CT scan in regard to clinical management remains unclear.

Current lung cancer screening guidelines recommend regular low-dose chest CTs starting at age 55 and continuing for 20-25 years (depending on the recommendation source) for individuals with a significant smoking history and for individuals who have stopped smoking within a specified interval since abstinence was achieved. Our recommendations are not intended to conflict with these guidelines.

Recommendation 2c: In newly diagnosed patients who are symptomatic and/or have abnormal pulmonary function testing, we recommend a baseline CT scan of the chest (weak recommendation, low quality evidence).

Minority opinion: While CT scans can show associated conditions in AATD (e.g., anatomic bronchiectasis, atypical mycobacterial infection, emphysema), whether detection of these findings impacts therapy in patients without suggestive clinical features is unclear. Furthermore, whether the benefits of clinical vigilance for these findings offset the radiation risk and cost of CT scans also remains unclear.

Rationale: A baseline CT may identify pulmonary emphysema even in the presence of a normal, complete pulmonary function test. The availability of a baseline study for comparison with future studies, especially when early panlobular disease can be difficult to detect without a comparator, can be helpful in later evaluations.

Recommendation 2d: We do not recommend serial chest CT scanning to monitor progression of disease (strong recommendation, low quality evidence).

Rationale: High value is placed on evidence-based data and cost. Chest CTs have shown a high frequency of radiographic (95%) and clinical (27%) bronchiectasis in a large study of individuals with AATD. However, no study to date has demonstrated improved clinical outcomes based on the knowledge provided by chest CTs. Occasionally, individuals with AATD and normal spirometry have symptoms referable to emphysema or bronchiectasis for which CT can establish a diagnosis.

Key references:11,21,22

Should individuals with Alpha-1 be monitored for the liver complications of AATD?

Background: Adults with AATD have an increased risk of cirrhosis and of hepatocellular carcinoma. Liver complications of AATD are relatively common, and may be found histologically in many older adults. Liver disease is generally restricted to ZZ and SZ individuals, though other rare genotypes are at risk (e.g., Mmalton and Siiyama). The role of concomitant hepatosteatosis, liver toxins including ethanol, and infectious agents, such as the hepatitis virus in the promotion of liver disease in AATD has been suggested but there are meager supporting data. There are few data in AATD to define the age at which to begin adult screening or define the optimal testing frequency.

Recommendation 2e: We recommend that individuals with AATD be monitored for liver disease at annual intervals (or more frequently as indicated by clinical circumstances), with physical examination including a focused exam for signs of liver disease, liver ultrasound, and laboratory monitoring of aspartate aminotransferase (AST), alanine aminotransferase (ALT), gamma-glutamyltransferase (GGT), albumin, bilirubin, international normalized ratio (INR), and platelets (strong recommendation, low quality of evidence).

Minority opinion: Clinical liver disease in AATD has a bimodal distribution in children and adults. Clinically significant liver disease is rare in adults less than 40 years of age.

Rationale: High value is placed on evidence-based data. Low value is placed on cost. There are very few data regarding evaluation of AATD separately from other liver diseases. Individuals with AATD have frequent, often transient elevations of transaminases. However, measurements of synthetic function (albumin) and correlates of splenomegaly (e.g., platelet count, liver ultrasound) are more specific for cirrhosis. Abnormal tests should prompt an abdominal ultrasound. The American Association for the Study of Liver Disease recommends an hepatic ultrasound every 6 months to monitor for hepatocellular carcinoma in individuals with a greater than 2% per year risk of hepatocellular carcinoma.23 This is likely to include any patient with cirrhosis, but may also include non-cirrhotic patients with significant injury, significant inflammation, or comorbidities. There are no prospective studies of this recommendation in AATD. Occasionally, individuals with normal laboratory values have symptoms referable to cirrhosis. Liver ultrasound, elastography, and biopsy can be helpful in these individuals.

Key references:24,25

3. Should the diagnosis of AATD alter medical management?

Differences in treatment between conditions are justified only when pathobiologic differences between the conditions produce different natural histories, prognoses, or differential responses to treatment. In the context of AATD, differences between managing patients with AAT-replete COPD and those with AATD relate to the specific pathogenetic role of the deficiency of AAT in predisposing to COPD. The specific questions and recommendations that follow represent those frequently asked by clinicians regarding whether to treat AATD patients differently from those with AAT-replete COPD.

Should usual medical management of COPD be altered in those with COPD due to AATD?

Background: The rate of lung function decline in smokers with AATD-COPD is considerably higher than that seen in AAT-replete COPD. This is likely due to the combined effects of cigarette smoke in reducing the anti-proteinase activity of the AAT molecule and promotion of pulmonary inflammatory infiltration.

There are no reliable data that suggest a differential treatment response to bronchodilators, inhaled corticosteroids, pulmonary rehabilitation, supplemental oxygen therapy, or immunizations in individuals with AATD-COPD and AAT-replete COPD. One study involving a small number of individuals with AATD-COPD found that treatment with an inhaled long-acting beta-agonist/glucocorticoid combination significantly improved FEV1 as would be expected in AAT-replete COPD. Also, one study26 found that in contrast to AAT-replete COPD patients undergoing transplant, AATD-COPD individuals receiving double lung transplantation experienced a more rapid FEV1 decline than single lung transplant recipients. Multiple studies have shown that survivorship following lung transplantation is similar in AAT-replete and AATD- COPD (Banga et al26 and International Society for Heart and Lung Transplantation data27).

Similarly, at the time of this writing, there is no specific therapy for AATD-associated liver disease. Management of liver disease is not altered by the diagnosis of AATD.

There are data to suggest that any benefit from lung volume reduction surgery is shorter-lived in individuals with AATD-COPD than those with AAT-replete COPD. In addition, there is often a basal predominance of the emphysema in AATD lung disease or generalized emphysema with a low diffusing capacity, making lung volume reduction surgery more technically difficult and with a higher mortality (National Emphysema Treatment Trial study28).

Recommendation 3a: We recommend every effort be made to prevent exposure to tobacco smoke and facilitate cessation in those who are smoking (strong recommendation, high quality evidence).

Rationale: High value is placed on observational data from well-designed registries and biochemical understanding of AATD. Data suggest that identification of AATD at birth is associated with a lower rate of smoking initiation and that the diagnosis of AATD in adults is associated with a greater willingness to attempt quitting and greater success in quitting.

Key references:29,30

Recommendation 3b: We do not recommend lung volume reduction surgery for individuals with AATD-COPD (weak recommendation, low quality evidence).

Rationale: High value is placed on the multicenter treatment experience. Low value is placed on case reports of significant long-lasting improvement in some individuals. AATD-COPD characteristically has lower lobe-predominant, panacinar/panlobular emphysema that has been shown to respond less well to surgical lung volume reduction than in individuals with upper lobe-predominant heterogeneous COPD. While some individuals with AATD-COPD have upper lobe-predominant heterogeneous disease, no current data suggest that these individuals respond comparably to those with AAT-replete COPD. Newer therapies of bronchoscopic lung volume reduction are being evaluated in AATD.

Key references:31

Should AATD individuals with COPD be treated with intravenous augmentation therapy?

Background: Intravenous administration of purified preparations of pooled donor-derived human AAT has been shown to augment levels of AAT and the AAT-related anti-elastase capacity of serum and lung epithelial lining fluid. The current U.S. Food and Drug Administration (FDA)-approved intravenous augmentation therapy dose for chronic administration is 60 mg/kg body weight, administered weekly.

Recommendation 3c: Intravenous augmentation therapy is recommended for individuals with AATD and an FEV1 in the range of 30%-65% predicted (strong recommendation, high quality evidence).

Rationale: This recommendation places a high value on treating individuals with moderate obstruction, a group with well-documented benefit. It also places high value on the finding that intravenous augmentation therapy is associated with lower levels of elastin degradation products in individuals with AATD and on lower rates of loss of CT lung density in individuals with AATD-COPD receiving augmentation therapy compared with those receiving placebo. It weighs the cost of therapy against these expected benefits.

Key references:5,32

Recommendation 3d: For individuals with FEV1 less than 30% predicted, intravenous augmentation therapy is recommended (weak recommendation, low quality of evidence).

Rationale: High value is placed on the potential to prolong survival in this group, the finding that intravenous augmentation therapy is associated with lower levels of elastin degradation products in individuals with AATD, and lower rates of loss of CT lung density in individuals with AATD-COPD receiving augmentation therapy. Low value is placed on the cost of this therapy.

Recommendation 3e: For those with FEV1 greater than 65%, we recommend discussion with each individual regarding the potential benefits of reducing lung function decline with consideration of the cost of therapy and lack of evidence for such benefit (strong recommendation, low quality of evidence).

Rationale: This recommendation to discuss the pros and cons of augmentation therapy places a high value on treating high-risk individuals early in the course of their disease to avoid an accelerated rate of lung function decline and its consequences. It also places high value on the finding that intravenous augmentation therapy is associated with lower levels of elastin degradation products in individuals with AATD and on lower rates of loss of CT lung density in individuals with AAT-replete COPD receiving augmentation therapy. Factors such as age, a rapid decline in FEV1, decreasing diffusing capacity, or progression of emphysema on imaging studies can be important to inform a decision regarding treatment.

Recommendation 3f: Weekly doses of intravenous augmentation therapy higher than the current FDA-approved dose are not recommended (weak recommendation, low quality of evidence).

Rationale: High value is placed on the few studies evaluating clinical and biochemical outcomes with different intravenous augmentation therapy regimens. Dosing schedules other than what is currently approved (e.g., 120 mg/kg every 2 weeks) appear to provide less complete biochemical protection against lung destruction in some individuals. Higher doses (i.e., 120 mg/kg/week) appear to be safe and increase AAT levels towards more physiologic values but clinical efficacy has not been proven. Some value is placed on the cost of intravenous augmentation therapy.

Note: Short-term lifestyle considerations may prompt alternate dosing regimens, such as allowing an individual to enjoy a two-week vacation at a distant location by administering a double dose just before departure. There have been no dose-ranging studies completed to establish the appropriate dose based on clinical endpoints.

Key references:33

Recommendation 3g: Intravenous augmentation therapy is not recommended for affected individuals with lung disease who continue to smoke (weak recommendation, low quality evidence).

Minority opinion: There is no current evidence that augmentation does not benefit individuals with AATD-COPD who continue to smoke.

Rationale: High value is placed on the proven benefits of tobacco cessation in the natural history of COPD (decreased FEV1 decline and decreased mortality), the high cost of therapy, and relative lower benefits of intravenous augmentation therapy in active smokers, as cigarette smoke causes oxidative inactivation of the elastase inhibiting capacity of AAT in vitro and in vivo.

Recommendation 3h: Monitoring of trough AAT levels to document adequate AAT augmentation dosing is not recommended (strong recommendation, moderate quality evidence).

Rationale: High value is placed on the lack of studies addressing doses of intravenous augmentation therapy other than 60 mg/kg weekly for the treatment of AATD-COPD. Low value is placed on the low cost of AAT level measurements.

Remarks: Potential costs and benefits should be discussed with the individual patient; actions will be driven by individual and physician judgment as well as by cost considerations. Intravenous augmentation therapy is an expensive intervention that is associated with significant inconvenience from regular IV infusions. Augmentation slows the progression of anatomic emphysema as assessed by CT densitometry in clinical trials. There is currently no high-quality evidence that intravenous augmentation therapy lessens the incidence or severity of exacerbations in AATD-related lung disease. One large observational cohort study in which the majority of individuals had FEV1 <30% predicted found that intravenous augmentation therapy improved survival in individuals with AATD with airflow obstruction.

Key references:34,35

Should AATD individuals without emphysema be treated with intravenous augmentation therapy?

Recommendation 3i: We do not recommend intravenous augmentation therapy in individuals with bronchiectasis without airflow obstruction (weak recommendation, low quality evidence).

Rationale: High value is placed on the cost and lack of specific evidence of benefit in this setting. Although intravenous augmentation therapy has been shown to decrease inflammatory markers in sputum and lung epithelial lining fluid and to enhance anti-elastase capacity, low value is placed on the concept that decreasing the neutrophil elastase burden that characterizes the secretions of bronchiectatic airways will decrease symptoms and disease progression.

Recommendation 3j: Intravenous augmentation therapy is not recommended for the treatment of liver disease due to AATD (strong recommendation, low quality of evidence).

Rationale: High value is placed on our current understanding of mechanisms underlying liver disease related to AATD. While the AATD-related lung disease is caused by a deficiency of circulating AAT, the liver disease associated with AATD is thought to be due to an over-abundance of polymerized AAT protein trapped within hepatocytes. Therefore, augmenting the circulating levels of AAT would seem unlikely to be of benefit. Clinical experience treating individuals with both lung and liver disease with intravenous augmentation therapy suggests that liver disease neither improves nor worsens with this therapy.

Recommendation 3k: We recommend intravenous augmentation therapy for the treatment of necrotizing panniculitis in individuals with AATD (strong recommendation, low quality of evidence).

Rationale: High value is placed on improving this often serious dermatologic condition. Several case reports have documented the relative effectiveness of intravenous augmentation therapy in preventing or controlling the skin lesions associated with this condition. Higher doses of intravenous augmentation therapy than those approved for the treatment of lung disease due to AATD may be needed. Low value is placed on the high cost of therapy.

Key references:9,36

Recommendation 3l: Intravenous augmentation therapy is not recommended for individuals who have undergone liver transplantation (strong recommendation, high level of evidence).

Rationale: High value is placed on the fact that a successful liver transplantation for severe AATD liver disease should lead to normal circulating levels of a normal AAT protein. The successfully transplanted normal donor liver will synthesize and release normal quantities of AAT to the blood.

Recommendation 3m: Intravenous augmentation therapy is not recommended for individuals with the MZ genotype who have COPD (strong recommendation, low quality evidence).

Rationale: High value is placed on the cost of therapy and the lack of evidence to support such therapy in this group. There are no data that intravenous augmentation therapy improves outcomes in those with the MZ or other heterozygote genotypes that include a normal M gene. There is neither a biologic rationale nor evidence to support treating these individuals. The primary treatment of individuals with the MZ genotype and COPD is smoking cessation and other evidence-based therapies for COPD.

Key references:37,38

Conclusions and Future Directions

The formal evidence that supports the recommendations in these guidelines includes observational studies; case-control studies; small, randomized controlled trials; and a single, well-powered randomized controlled trial, which was published during the final preparation of these guidelines. This last trial demonstrated that augmentation therapy provided a statistically significant reduction in emphysema progression evaluated by CT densitometry performed at total lung capacity (TLC) but not at TLC plus functional residual capacity. Even so, there remain gaps in available studies to address important management issues. Therefore, the clinical observations and opinions of experts in the diagnosis and management of AATD played a prominent role in crafting this document and the recommendations that are offered. Acknowledging these limitations, the writing committee and the Medical and Scientific Advisory Committee of the Alpha-1 Foundation believe that this document will provide meaningful guidance for clinicians caring for individuals with this widely under-diagnosed condition.

Until universal neonatal screening for AATD is adopted, the true prevalence of this condition and its natural history will not be fully understood. As additional well-powered studies of therapies for AATD are published and perhaps newer, more convenient therapies are developed and tested using accepted clinical endpoints, many of the expert-based recommendations in this document may be replaced by fully evidence-based documentation. The authors look forward to that time.

Alpha-1 antitrypsin deficiency is the major, known genetic risk factor for chronic obstructive lung disease and a prominent genetic risk factor for liver disease. It is the committee’s hope that providing clear guidance will improve detection of individuals with AATD and promote evidence-based treatment of those who are diagnosed.

Limitations

In this attempt to offer clinician-centric recommendations regarding the care of individuals with AATD, several important limitations are noteworthy. First, because AATD is an uncommon disease, formidable challenges exist in assembling large cohorts for adequately powered randomized clinical trials upon which to base the most rigorous clinical conclusions. As such, some uncertainty persists regarding key clinical questions such as the cost-effectiveness of intravenous augmentation therapy and the optimal strategies for targeted detection of affected individuals. Clinical recommendations require synthesizing the literature and studies from disparate settings and sources. Second, because progress in uncommon diseases like AATD requires close collaborations between clinical leaders, academic leaders, the patient community, government, and pharma, engagement of the authors with patient organizations like the Alpha-1 Foundation would disqualify them from participating in writing clinical practice guidelines under current medical society criteria. We have addressed this perceived conflict through disclosure and we highlight the paradox that the very engagement that permits the expertise needed to write this document and to receive and understand the questions that clinicians pose is a curious disqualifier.

Drugs for intravenous augmentation therapy are marketed in the United States and Europe by a number of different manufacturers. The cost of this therapy is very high. Intravenous augmentation therapy was approved in the United States based on its biochemical efficacy --its ability to increase serum and lung levels of AAT above putative protective threshold values. Numerous observational studies have provided evidence that individuals receiving this therapy for their AATD-related lung disease show a decreased rate of lung function decline and one large study showed an improvement in survival compared to AATD-affected individuals who never received intravenous augmentation therapy. Several small, randomized pilot trials provided suggestive trends toward effectiveness and the community of clinicians caring for lung-affected individuals with AATD, as well as affected individuals themselves, rely on this therapy to supplement the usual treatment of COPD. In fact, the degree of acceptance of intravenous augmentation therapy by these communities within the United States and other countries has made it virtually impossible to perform placebo-controlled trials to formally evaluate effectiveness. Even so, relying heavily on enrollment in countries where there is limited or no availability of augmentation therapies, a single, adequately powered, randomized, placebo-controlled, blinded efficacy study has now been published.5

Because AATD is an uncommon genetic condition, it becomes difficult to provide high-level literature support for the various treatments that are commonly prescribed for individuals with AATD. Even the usual medications used to treat COPD and liver disease in the general population are being used “off label” in the AATD community, since virtually all studies leading to the approval of these therapies excluded individuals with AATD. Most bronchoscopic lung volume reduction therapies being investigated in the United States (and available in Europe) have excluded individuals with AATD as well. A final qualification is that the cost-effectiveness of intravenous augmentation therapy has been difficult to address since the overall benefit is still under study.39

Declaration of Interest

The work of this writing committee was supported by the Alpha-1 Foundation through payment of travel and meeting expenses and payment of library searching and printing fees. No other assistance was provided. Specifically, there was not honoraria paid and there was not writing assistance of any sort provided. Ms. Knight and Drs. Turino, Brantly, and Goodman have nothing to disclose. Dr. Sandhaus reports grants from CSL Behring. He is employed part-time by the Alpha-1 Foundation and AlphaNet, 2 not-for-profit entities that support research and health management in alpha-1 antitrypsin deficiency and receive part of their funding from pharmaceutical industry sources. Dr. Sandhaus serves on the Medical and Scientific Advisory Committee of the COPD Foundation. Dr. Campos reports grants from Grifols and CSL Behring. Dr. Cross reports personal fees from CSL Behring and Baxter. Dr. Hogarth reports personal fees from Baxter, CSL Behring, and Grifols. Dr. Stocks reports grants to the University of Texas from Kamada, Grifols, and Talecris. Dr. Stoller has served as a scientific consultant to Grifols, CSL Behring, Baxalta, and Kamada regarding alpha-1 antitrypsin deficiency and serves on the Board of Directors of the Alpha-1 Foundation and on the Medical and Scientific Advisory Committees of the Alpha-1 Foundation and the COPD Foundation. Dr. Strange reports personal fees and stock options from Abeona, grants from Baxalta, grants and personal fees from CSL Behring, grants and personal fees from Grifols, and grants from Alpha-1 Foundation. Dr. Teckman reports he is a paid consultant for Isis Pharmaceuticals, Arrowhead Research, Alnylam Inc., Editas Inc., Proteostasis Inc., Genkyotix, Grifols, Intellia, Retrophin, RxCelerate, Velfene, and Vertex. He has received grants from the National Institutes of Health and the Alpha-1 Foundation.

Online Supplement

Search Strategy and Results Summary

The following peer-reviewed search strategy (Table I) was used to search MEDLINE with minor revisions for EMBASE, Cochrane Central Registry of Controlled Trials (CCTR) and Database of Abstracts of Reviews of Effects (DARE): (all alpha-1 antitrypsin deficiency OR familial emphysema) OR ((alpha-1 antitrypsin OR drugs) limited to treatment)).

The search for alpha-1 antitrypsin deficiency retrieved 3791 citations less duplicates for January 2002 to December 2014. Three additional hand searched articles were added. Reviewers used EndNote files for the title and abstract review. Full text articles were screened using pre-defined inclusion criteria; 34 were included in the review.

Table 1. Alpha 1-Antitrypsin Deficiency Search Strategy In-Process & Other Non-Indexed Citations, Ovid MEDLINE(R) and Ovid OLDMEDLINEa

#

Search

1

Alpha 1-Antitrypsin Deficiency/

2

((AAt or A1AT or A1-AT or A1PI or A1P1) adj deficien$).tw.

3

AATD.tw.

4

(alpha1 or alpha 1 or alpha one or alfa1 or alfa 1 or alfa one).tw.

5

(antitrypsin$ or anti-trypsin$).tw.

6

(antiproteinase or antiprotease).tw.

7

(proteinase inhibit$ or protease inhibit$).tw.

8

5 or 6 or 7

9

deficien$.tw.

10

4 and 8 and 9

11

2 or 3 or 10

12

1 or 11

13

(familial adj emphysema).tw.

14

pulmonary emphysema/ge

15

genetic emphysema.tw.

16

((inherit$ or heretitary) adj3 emphysema).tw.

17

or/13-16

18

12 or 17 [all alpha 1-antitrypsin deficiency OR genetic emphysema]

19

Alpha 1-Antitrypsin/

20

(alpha1 or alpha 1 or alpha one or alfa1 or alfa 1 or alfa one).tw.

21

(antitrypsin$ or anti-trypsin$).tw.

22

(antiproteinase or antiprotease).tw.

23

(proteinase inhibit$ or protease inhibit$).tw.

24

21 or 22 or 23

25

20 and 24

26

(A1AT or hA1AT or A1-AT or A1PI or A1P1).tw.

27

19 or 21 or 25 or 26 [all alpha 1-Antitrypsin]

28

Prolastin.mp.

29

Aralast.mp.

30

Zemaira.mp.

31

28 or 29 or 30 [alpha 1-antitrypsin drugs]

32

27 or 31 [all alpha 1-Antitrypsin OR drugs]

33

limit 18 to english

34

limit 18 to abstracts

35

33 or 34

36

limit 35 to human

37

limit 35 to animal

38

35 not 36 not 37

39

36 or 38 [all alpha 1-antitrypsin deficiency OR genetic emphysema, limited to English or English abstracts, human or non-indexed]

40

limit 32 to english

41

limit 32 to abstracts

42

40 or 41

43

limit 42 to human

44

limit 42 to animal

45

42 not 43 not 44

46

43 or 45 [all alpha 1-Antitrypsin OR drugs, limited to English or English abstracts, human or non-indexed]

47

46 not 39 [alpha 1-Antitrypsin OR drugs, but NOT alpha 1-antitrypsin deciciency, limited to English or English abstracts, human or non-indexed, all years]

48

clinical trial.pt. or random.mp. or (clinical trial or clinical trials).mp. or (tu or ad or ae).fs. or th.xs. or exp Treatment Outcome/ or (treatments adj2 (effectiv$ or efficacy)).mp. or exp cohort studies/ or exp case control studies/ or cross-sectional studies/ [treatment filter]

49

47 and 48 [alpha 1-Antitrypsin OR drugs, but NOT alpha 1-antitrypsin deficiency, limited to treatment]

50

47 not 49 [all alpha 1-Antitrypsin OR drugs that are NOT deficiency, AND NOT treatment, all years]

51

limit 50 to yr="2002-Current" [all alpha 1-Antitrypsin OR drugs that are NOT deficiency, AND NOT treatment, 2002+]

52

50 not 51 [all alpha 1-Antitrypsin OR drugs that are NOT deficiency, AND NOT treatment, before 2002]

53

39 or 49

54

limit 53 to yr="2005 -Current"

55

alpha 1-Antitrypsin Deficiency/

56

((AAt or A1AT or A1-AT or A1PI or A1P1) adj deficien$).tw.

57

AATD.tw.

58

(alpha1 or alpha 1 or alpha one or alfa1 or alfa 1 or alfa one).tw.

59

(antitrypsin$ or anti-trypsin$).tw.

61

(proteinase inhibit$ or protease inhibit$).tw.

63

deficien$.tw.

65

56 or 57 or 64

67

(familial adj emphysema).tw.

69

genetic emphysema.tw.

71

or/67-70

73

alpha 1-Antitrypsin/

75

(antitrypsin$ or anti-trypsin$).tw.

77

(proteinase inhibit$ or protease inhibit$).tw.

79

74 and 78

81

73 or 75 or 79 or 80 [all alpha 1-Antitrypsin]

83

Aralast.mp.

84

Zemaira.mp.

85

82 or 83 or 84 [alpha 1-antitrypsin drugs]

86

81 or 85 [all alpha 1-Antitrypsin OR drugs]

87

limit 72 to english

88

limit 72 to abstracts

89

87 or 88

90

limit 89 to human

91

limit 89 to animal

92

89 not 90 not 91

93

90 or 92 [all alpha 1-antitrypsin deficiency OR genetic emphysema, limited to English or English abstracts, human or non-indexed]

aAdapted for Embase, Cochrane Central Register of Controlled Trials (CCTR), Database of Abstracts of Reviews of Effects (DARE), and Cochrane Database of Systematic Reviews (CDSR) in Ovid.

 

Online Supplement: Supplemental Bibliography

Detection and Diagnosis

1. Holme J, Stockley JA, Stockley RA. Age related development of respiratory abnormalities in non-index alpha-1 antitrypsin deficient studies. Respir Med. 2013;107(3):387-393. doi: http://www.ncbi.nlm.nih.gov/pubmed/23276486

2. Ferrarotti I, Thun GA, Zorzetto M, et al. Serum levels and genotype distribution of alpha1-antitrypsin in the general population. Thorax. 2012;67(8):669-674. doi:http://www.ncbi.nlm.nih.gov/pubmed/22426792

3. Hogarth DK. Alpha one antitrypsin genotypes in an asthma referral clinic. Ann Allergy Asthma Immunol. 2012;109:A23. doi: http://www.ncbi.nlm.nih.gov/pubmed/70922512

4. Rahaghi FF, Sandhaus RA, Brantly ML, et al. The prevalence of alpha-1 antitrypsin deficiency among patients found to have airflow obstruction. COPD. 2012;9(4):352-358.doi: http://www.ncbi.nlm.nih.gov/pubmed/22506682

5. Thun GA, Ferrarotti I, Imboden M, et al. SERPINA1 PiZ and PiS heterozygotes and lung function decline in the SAPALDIA cohort. PLoS One. 2012;7(8):e42728. doi: http://www.ncbi.nlm.nih.gov/pubmed/22912729

6. Coors ME, Moseley R, McGorray S. Informed consent process in Alpha-1 testing of at-risk children: views of parents and adults tested as children. COPD. 2011;8(1):30-38.doi: http://www.ncbi.nlm.nih.gov/pubmed/21299476

7. Campos MA, Alazemi S, Zhang G, et al. Clinical characteristics of subjects with symptoms of alpha1-antitrypsin deficiency older than 60 years. Chest. 2009;135(3):600-608.doi: http://www.ncbi.nlm.nih.gov/pubmed/19017884

8. Coors ME, Levinson AH, Huitt GA. Anxiety related to genetic testing for alpha-1 antitrypsin deficiency and cystic fibrosis in COPD and/or bronchiectasis patients. Community Genet. 2008;11(3):135-140. doi: http://www.ncbi.nlm.nih.gov/pubmed/18376109

9. Bals R, Koczulla R, Kotke V, Andress J, Blackert K, Vogelmeier C. Identification of individuals with alpha-1-antitrypsin deficiency by a targeted screening program. Respir Med. 2007;101(8):1708-1714.doi: http://www.ncbi.nlm.nih.gov/pubmed/17428650

10. Carpenter MJ, Strange C, Jones Y, et al. Does genetic testing result in behavioral health change? Changes in smoking behavior following testing for alpha-1 antitrypsin deficiency. Ann Behav Med. 2007;33(1):22-28.doi: http://www.ncbi.nlm.nih.gov/pubmed/17291167

11. Eden E, Holbrook JT, Brantly ML, Turino GM, Wise RA. Prevalence of alpha-1 antitrypsin deficiency in poorly controlled asthma—results from the ALA-ACRC low-dose theophylline trial. J Asthma. 2007;44(8):605-608. doi: http://www.ncbi.nlm.nih.gov/pubmed/17943569

12. Ferrarotti I, Scabini R, Campo I, et al. Laboratory diagnosis of alpha1-antitrypsin deficiency. Transl Res. 2007;150(5):267-274. doi: http://www.ncbi.nlm.nih.gov/pubmed/17964515

13. Holme J, Stockley RA. Radiologic and clinical features of COPD patients with discordant pulmonary physiology: lessons from alpha1-antitrypsin deficiency. Chest. 2007;132(3):909-915. doi: http://www.ncbi.nlm.nih.gov/pubmed/17573491

14. Strange C, Moseley MA, Jones Y, Schwarz L, Xie L, Brantly ML. Genetic testing of minors for alpha1-antitrypsin deficiency. Arch Pediatr Adolesc Med. 2006;160(5):531-534. doi: http://www.ncbi.nlm.nih.gov/pubmed/16651497

15. Strange C, Stoller JK, Sandhaus RA, Dickson R, Turino G. Results of a survey of patients with alpha-1 antitrypsin deficiency. Respiration. 2006;73(2):185-190. doi: http://www.ncbi.nlm.nih.gov/pubmed/16141711

16. Campos MA, Wanner A, Zhang G, Sandhaus RA. Trends in the diagnosis of symptomatic patients with alpha1-antitrypsin deficiency between 1968 and 2003. Chest. 2005;128(3):1179-1186. doi: http://www.ncbi.nlm.nih.gov/pubmed/16162704

17. Stoller JK, Sandhaus RA, Turino G, Dickson R, Rodgers K, Strange C. Delay in diagnosis of alpha1-antitrypsin deficiency: a continuing problem. Chest. 2005;128(4):1989-1994. doi: http://www.ncbi.nlm.nih.gov/pubmed/16236846

18. Blanco I, Fernandez-Bustillo E, de Serres FJ, Alkassam D, Rodriguez Menendez C. PI*S and PI*Z alpha 1-antitrypsin deficiency: estimated prevalence and number of deficient subjects in Spain. Med Clin (Barc). 2004;123(20):761-765. doi:http://www.ncbi.nlm.nih.gov/pubmed/15607066

19. Chappell S, Guetta-Baranes T, Batowski K, et al. Haplotypes of the alpha-1 antitrypsin gene in healthy controls and Z deficiency patients. Hum Mutat. 2004;24(6):535-536. doi: http://www.ncbi.nlm.nih.gov/pubmed/15532029

20. Cox DW. Prenatal diagnosis for alpha1-antitrypsin deficiency. Prenat Diagn. 2004;24(6):468-470. doi: http://www.ncbi.nlm.nih.gov/pubmed/15229848

21. Kwok JSY, Lawton JWM, Yew WW, Chau CH, Lee J, Wong PC. Protease inhibitor phenotypes and serum alpha-1-antitrypsin levels in patients with COPD: a study from Hong Kong. Respirology. 2004;9(2):265-270. doi: http://www.ncbi.nlm.nih.gov/pubmed/15182280

22. Luisetti M, Seersholm N. Alpha1-antitrypsin deficiency. 1: epidemiology of alpha1-antitrypsin deficiency. Thorax. 2004;59(2):164-169. doi: http://www.ncbi.nlm.nih.gov/pubmed/14760160

23. Sibtain NA, Padley SPG. HRCT in small and large airways diseases. Eur Radiol Suppl. 2004;14 (4):L31-L43.

24. Wadsworth MEJ, Vinall LE, Jones AL, et al. Alpha1-antitrypsin as a risk for infant and adult respiratory outcomes in a national birth cohort. Am J Respir Cell Mol Biol. 2004;31(5):559-564. doi: http://www.ncbi.nlm.nih.gov/pubmed/15271689

25. Silva GE, Sherrill DL, Guerra S, Barbee RA. A longitudinal study of alpha1-antitrypsin phenotypes and decline in FEV1 in a community population. Chest. 2003;123(5):1435-1440. doi: http://www.ncbi.nlm.nih.gov/pubmed/12740258

26. Rodriguez F, Jardi R, Costa X, et al. Rapid screening for alpha1-antitrypsin deficiency in patients with chronic obstructive pulmonary disease using dried blood specimens. Am J Respir Crit Care Med. 2002;166(6):814-817. doi: http://www.ncbi.nlm.nih.gov/pubmed/12231490

27. Seersholm N. Epidemiology of emphysema in subjects with severe alpha 1-antitrypsin deficiency. Dan Med Bull. 2002;49(2):145-158. doi: http://www.ncbi.nlm.nih.gov/pubmed/12064091

Clinical Evaluation of Individuals with AATD

1. Subramanian DR, Edgar R, Ward H, Parr DG, Stockley RA. Prevalence and radiological outcomes of lung nodules in alpha 1-antitrypsin deficiency. Respir Med. 2013;107(6):863-869. doi: http://www.ncbi.nlm.nih.gov/pubmed/23337299

2. Bernspang E, Diaz S, Stoel B, Wollmer P, Sveger T, Piitulainen E. CT lung densitometry in young adults with alpha-1-antitrypsin deficiency. Respir Med. 2011;105(1):74-79. doi: http://www.ncbi.nlm.nih.gov/pubmed/20674322

3. Dye DE, Brameld KJ, Maxwell S, Goldblatt J, O’Leary P. The impact of single gene and chromosomal disorders on hospital admissions in an adult population. J Community Genet. 2011;2(2):81-90. doi: http://www.ncbi.nlm.nih.gov/pubmed/22109792

4. McGee D, Strange C, McClure R, Schwarz L, Erven M. The Alpha-1 Association Genetic Counseling Program: an innovative approach to service. J Genet Couns. 2011;20(4):330-336. doi: http://www.ncbi.nlm.nih.gov/pubmed/21424325

5. Ottaviani S, Gorrini M, Scabini R, et al. C reactive protein and alpha1-antitrypsin: relationship between levels and gene variants. Transl Res. 2011;157(6):332-338. doi: http://www.ncbi.nlm.nih.gov/pubmed/21575917

6. Shin S-Y, Choi GS, Lee KH, et al. Changes of Alpha1-Antitrypsin Levels in Allergen-induced Nasal Inflammation. Clin Exp Otorhinolaryngol. 2011;4(1):33-39. doi: http://www.ncbi.nlm.nih.gov/pubmed/21461061

7. Topic A, Prokic D, Stankovic I. Alpha-1-antitrypsin deficiency in early childhood. Fetal Pediat Pathol. 2011;30(5):312-319. doi: http://www.ncbi.nlm.nih.gov/pubmed/21609162

8. Zutler M, Quinlan PJ, Blanc PD. Alpha-1-antitrypsin deficient man presenting with lung function decline associated with dust exposure: a case report. J Med Case Reports. 2011;5:154. doi: http://www.ncbi.nlm.nih.gov/pubmed/21504560

9. Alam S, Li Z, Mahadeva R. Cigarette smoke induced oxidation of a-1 antitrypsin amplifies the pulmonary inflammatory response. Thorax. 2010;65:A29.

10. Banauch GI, Brantly M, Izbicki G, et al. Accelerated spirometric decline in New York City firefighters with alpha1-antitrypsin deficiency. Chest. 2010;138(5):1116-1124. doi: http://www.ncbi.nlm.nih.gov/pubmed/20634282

11. Duckers JM, Shale DJ, Stockley RA, et al. Cardiovascular and musculskeletal co-morbidities in patients with alpha 1 antitrypsin deficiency. Respir Res. 2010;11:173. doi: http://www.ncbi.nlm.nih.gov/pubmed/21138571

12. Eden E. Asthma and COPD in alpha-1 antitrypsin deficiency. Evidence for the Dutch hypothesis. COPD. 2010;7(5):366-374. doi: http://www.ncbi.nlm.nih.gov/pubmed/20854052

13. Gama MPR, Jonasson TH, Gama JD, et al. Extreme insulin-resistance syndrome associated with alpha-1 antitrypsin deficiency. Endocrinologist. 2010;20 (3):137-140.

14. Klitzman RL. Misunderstandings concerning genetics among patients confronting genetic disease. J Genet Couns. 2010;19(5):430-446. doi: http://www.ncbi.nlm.nih.gov/pubmed/20512408

15. Tanash HA, Nilsson PM, Nilsson J-A, Piitulainen E. Survival in severe alpha-1-antitrypsin deficiency (PiZZ). Respir Res. 2010;11:44. doi: http://www.ncbi.nlm.nih.gov/pubmed/20420704

16. Ward H, Miller MR, Stockley RA. Changes in physiological phenotypes of a-1-antitrypsin deficiency with time. Thorax. 2010; 65:A30-A31.

17. Bernspang E, Wollmer P, Sveger T, Piitulainen E. Lung function in 30-year-old alpha-1-antitrypsin-deficient individuals. Respir Med. 2009;103(6):861-865. doi: http://www.ncbi.nlm.nih.gov/pubmed/19181511

18. Campos MA, Alazemi S, Zhang G, Wanner A, Sandhaus RA. Effects of a disease management program in individuals with alpha-1 antitrypsin deficiency. COPD. 2009;6(1):31-40. doi: http://www.ncbi.nlm.nih.gov/pubmed/19229706

19. Dawkins P, Wood A, Nightingale P, Stockley R. Mortality in alpha-1-antitrypsin deficiency in the United Kingdom. Respir Med. 2009;103(10):1540-1547. doi: http://www.ncbi.nlm.nih.gov/pubmed/19443188

20. Dawkins PA, Dawkins CL, Wood AM, Nightingale PG, Stockley JA, Stockley RA. Rate of progression of lung function impairment in alpha1-antitrypsin deficiency. Eur Respir J. 2009;33(6):1338-1344. doi: http://www.ncbi.nlm.nih.gov/pubmed/19164359

21. Han MK, Bartholmai B, Liu LX, et al. Clinical significance of radiologic characterizations in COPD. COPD. 2009;6(6):459-467. http://www.ncbi.nlm.nih.gov/pubmed/19938970

22. Iwanicka-Michalowicz M, Grzelewska-Rzymowska I. Risk factors of chronic obstructive pulmonary disease in hospitalised patients. [Polish]. Pediatria i Medycyna Rodzinna. 2009;5 (1):27-32.

23. Kalsheker NA. alpha1-Antitrypsin deficiency: best clinical practice. J Clin Pathol. 2009;62(10):865-869. doi: http://www.ncbi.nlm.nih.gov/pubmed/19783716

24. Kemp SV, Polkey MI, Shah PL. The epidemiology, etiology, clinical features, and natural history of emphysema. Thorac Surg Clin. 2009;19(2):149-158. http://www.ncbi.nlm.nih.gov/pubmed/19662957

25. Segelmark M, Mohammad A. Apmis. 2009;Conference: 14th International Vasculitis and ANCA Workshop Lund Sweden. Conference Start: 20090606 Conference End: 20090609. Conference Publication: (var.pagings). 117:163.

26. Soriano JB, Miravitlles M. Your racing horses will help you to quit: a lesson for COPD and alpha1-antitrypsin deficiency research. Eur Respir J. 2009;33(6):1244-1246. doi: http://www.ncbi.nlm.nih.gov/pubmed/19483042

27. Stavngaard T, Sogaard LV, Batz M, Schreiber LM, Dirksen A. Progression of emphysema evaluated by MRI using hyperpolarized (3)He (HP (3)He) measurements in patients with alpha-1-antitrypsin (A1AT) deficiency compared with CT and lung function tests. Acta Radiol. 2009;50(9):1019-1026. doi: http://www.ncbi.nlm.nih.gov/pubmed/19863412

28. Stoller JK, Aboussouan LS. Myths and misconceptions about alpha1antitrypsin deficiency. Arch Intern Med. 2009;169 (6):546-550. doi: http://www.ncbi.nlm.nih.gov/pubmed/19307516

29. van Beek EJR, Dahmen AM, Stavngaard T, et al. Hyperpolarised 3He MRI versus HRCT in COPD and normal volunteers: PHIL trial. Eur Respir J. 2009;34(6):1311-1321. doi: http://www.ncbi.nlm.nih.gov/pubmed/19541712

30. Wanner A. COPD: new lessons from alpha1-antitrypsin deficiency? Chest. 2009;135(5):1342-1344. doi: http://www.ncbi.nlm.nih.gov/pubmed/19420201

31. Wood AM, Harrison RM, Semple S, Ayres JG, Stockley RA. Outdoor air pollution is associated with disease severity in alpha1-antitrypsin deficiency. Eur Respir J. 2009;34(2):346-353. doi: http://www.ncbi.nlm.nih.gov/pubmed/19324958

32. Yamashiro T, Matsuoka S, Estepar RSJ, et al. Quantitative airway assessment on computed tomography in patients with alpha1-antitrypsin deficiency. COPD. 2009;6(6):468-477. doi: http://www.ncbi.nlm.nih.gov/pubmed/19938971

33. Abboud RT, Vimalanathan S. Pathogenesis of COPD. Part I. The role of protease-antiprotease imbalance in emphysema. Int J Tuberc Lung Dis. 2008;12(4):361-367. doi: http://www.ncbi.nlm.nih.gov/pubmed/18371259

34. Bakker ME, Putter H, Stolk J, et al. Assessment of regional progression of pulmonary emphysema with CT densitometry. Chest. 2008;134(5):931-937. doi: http://www.ncbi.nlm.nih.gov/pubmed/18625668

35. Bilton D. Update on non-cystic fibrosis bronchiectasis. Curr Opin Pulm Med. 2008;14(6):595-599. doi: http://www.ncbi.nlm.nih.gov/pubmed/18812838

36. Carter R, Tiep BL. Patients with alpha-1 antitrypsin deficiency: Disease management considerations. Disease Management and Health Outcomes. 2008;16 (5):345-351.

37. Fregonese L, Stolk J, Frants RR, Veldhuisen B. Alpha-1 antitrypsin Null mutations and severity of emphysema. Respir Med. 2008;102(6):876-884. doi: http://www.ncbi.nlm.nih.gov/pubmed/18353624

38. Lazarus A, Myers J, Fuhrer G. Bronchiectasis in adults: a review. Postgrad Med. 2008;120(3):113-121. doi: http://www.ncbi.nlm.nih.gov/pubmed/18824830

39. Moller W, Meyer G, Kreyling WG. Advances in lung imaging techniques for the treatment of respiratory disease. Drug Discov Today Ther Strateg. 2008;5 (2):87-92.

40. Parr DG, Sevenoaks M, Deng C, Stoel BC, Stockley RA. Detection of emphysema progression in alpha 1-antitrypsin deficiency using CT densitometry; methodological advances. Respir Res. 2008;9:21. doi: http://www.ncbi.nlm.nih.gov/pubmed/18271964

41. Petty TL, Mannino DM. Will recommendations against spirometry make chronic obstructive pulmonary disease harder to treat? Ann Intern Med. 2008;149(7):512-513; author reply 513. doi: http://www.ncbi.nlm.nih.gov/pubmed/18838733

42. Quast TM, Self AR, Browning RF. Diagnostic Evaluation of Bronchiectasis. Dis Mon. 2008;54 (8):527-539. doi: http://www.ncbi.nlm.nih.gov/pubmed/18638621

43. Rodriguez E, Ferrer J, Marti S, Zock JP, Plana E, Morell F. Impact of occupational exposure on severity of COPD. Chest. 2008;134(6):1237-1243. doi: http://www.ncbi.nlm.nih.gov/pubmed/18689596

44. Silva GE, Guerra S, Keim S, Barbee RA, Sherrill DL. Longitudinal decline of diffusing capacity of the lung for carbon monoxide in community subjects with the PiMZ alpha1-antitrypsin phenotype. Chest. 2008;133(5):1095-1100. doi: http://www.ncbi.nlm.nih.gov/pubmed/18263681

45. Tanash HA, Nilsson PM, Nilsson JA, Piitulainen E. Clinical course and prognosis of never-smokers with severe alpha-1-antitrypsin deficiency (PiZZ). Thorax. 2008;63(12):1091-1095. doi: http://www.ncbi.nlm.nih.gov/pubmed/18682522

46. Yang P, Sun Z, Krowka MJ, et al. Alpha1-antitrypsin deficiency carriers, tobacco smoke, chronic obstructive pulmonary disease, and lung cancer risk. Arch Intern Med. 2008;168(10):1097-1103. doi: http://www.ncbi.nlm.nih.gov/pubmed/18504338

47. Chan ED, Kaminska AM, Gill W, et al. Alpha-1-antitrypsin (AAT) anomalies are associated with lung disease due to rapidly growing mycobacteria and AAT inhibits Mycobacterium abscessus infection of macrophages. Scand J Infect Dis. 2007;39(8):690-696. doi: http://www.ncbi.nlm.nih.gov/pubmed/17654345

48. Gupta N, Kabra M. Recent advances in approach to treatment of genetic disorders: Clinician’s perspective. Indian Pediatr. 2007;44 (5):361-369. doi: http://www.ncbi.nlm.nih.gov/pubmed/17536138

49. Hersh CP, Jacobson FL, Gill R, Silverman EK. Computed tomography phenotypes in severe, early-onset chronic obstructive pulmonary disease. COPD. 2007;4(4):331-337. doi: http://www.ncbi.nlm.nih.gov/pubmed/18027160

50. Kim W-D, Ling SH, Coxson HO, et al. The association between small airway obstruction and emphysema phenotypes in COPD. Chest. 2007;131(5):1372-1378. doi: http://www.ncbi.nlm.nih.gov/pubmed/17317735

51. Makita H, Nasuhara Y, Nagai K, et al. Characterisation of phenotypes based on severity of emphysema in chronic obstructive pulmonary disease. Thorax. 2007;62(11):932-937. doi: http://www.ncbi.nlm.nih.gov/pubmed/17573447

52. Meinke L, Chitkara R, Krishna G. Advances in the management of chronic obstructive pulmonary disease. Expert Opin Pharmacother. 2007;8(1):23-37. doi: http://www.ncbi.nlm.nih.gov/pubmed/17163804

53. Lomas DA. The selective advantage of alpha1-antitrypsin deficiency. Am J Respir Crit Care Med. 2006;173(10):1072-1077. doi: http://www.ncbi.nlm.nih.gov/pubmed/16439713

54. Mahr AD, Neogi T, Merkel PA. Epidemiology of Wegener’s granulomatosis: Lessons from descriptive studies and analyses of genetic and environmental risk determinants. Clin Exp Rheumatol. 2006;24(2 Suppl 41):S82-91. doi: http://www.ncbi.nlm.nih.gov/pubmed/16859601

55. Mayer AS, Stoller JK, Vedal S, et al. Risk factors for symptom onset in PI*Z alpha-1 antitrypsin deficiency. Int J Chron Obstruct Pulmon Dis. 2006;1(4):485-492. doi: http://www.ncbi.nlm.nih.gov/pubmed/18044105

56. O’Connor TP, Crystal RG. Genetic medicines: treatment strategies for hereditary disorders. Nat Rev Genet. 2006;7(4):261-276. doi: http://www.ncbi.nlm.nih.gov/pubmed/16543931

57. Parr DG, Dawkins PA, Stockley RA. Computed tomography: A new gold standard for the clinical assessment of emphysema. Am J Respir Crit Care Med. 2006;174 (8):954. doi: http://www.ncbi.nlm.nih.gov/pubmed/17021361

58. Wise RA. The value of forced expiratory volume in 1 second decline in the assessment of chronic obstructive pulmonary disease progression. Am J Med. 2006;119(10 Suppl 1):4-11. doi: http://www.ncbi.nlm.nih.gov/pubmed/16996894

59. McMahon MA, O’Mahony MJ, O’Neill SJ, McElvaney NG, Logan PM. Alpha-1 antitrypsin deficiency and computed tomography findings. J Comput Assist Tomogr. 2005;29(4):549-553. doi: http://www.ncbi.nlm.nih.gov/pubmed/16012317

60. Senn O, Russi EW, Imboden M, Probst-Hensch NM. alpha1-Antitrypsin deficiency and lung disease: risk modification by occupational and environmental inhalants. Eur Respir J. 2005;26(5):909-917. doi: http://www.ncbi.nlm.nih.gov/pubmed/16264055

61. Sheel AW, Road JD. Effects of physical activity on exercise capacity in twins with alpha-1 antitrypsin deficiency. Clin J Sport Med. 2005;15(3):183-185. doi: http://www.ncbi.nlm.nih.gov/pubmed/15867564

62. Stoller JK, Tomashefski J, Jr., Crystal RG, et al. Mortality in individuals with severe deficiency of alpha1-antitrypsin: findings from the National Heart, Lung, and Blood Institute Registry. Chest. 2005;127(4):1196-1204. doi: http://www.ncbi.nlm.nih.gov/pubmed/15821195

63. Yang P, Bamlet WR, Sun Z, et al. Alpha1-antitrypsin and neutrophil elastase imbalance and lung cancer risk.[Erratum appears in Chest. 2006 Jan;129(1):216 Note: Krowka, Michael J [added]]. Chest. 2005;128(1):445-452. doi: http://www.ncbi.nlm.nih.gov/pubmed/16002971

64. Shaker SB, Stavngaard T, Stolk J, Stoel B, Dirksen A. Alpha1-antitrypsin deficiency. 7: Computed tomographic imaging in alpha1-antitrypsin deficiency. Thorax. 2004;59(11):986-991. doi: http://www.ncbi.nlm.nih.gov/pubmed/15516477

65. Wewers MD. Alpha1-antitrypsin deficiency: more than a protease imbalance? Chest. 2004;125(5):1607-1609. doi: http://www.ncbi.nlm.nih.gov/pubmed/15136365

66. 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. doi: http://www.ncbi.nlm.nih.gov/pubmed/14522813

67. Corbo GM, Forastiere F, Agabiti N, et al. Passive smoking and lung function in alpha(1)-antitrypsin heterozygote schoolchildren. Thorax. 2003;58(3):237-241. doi: http://www.ncbi.nlm.nih.gov/pubmed/12612303

68. Dawkins PA, Dowson LJ, Guest PJ, Stockley RA. Predictors of mortality in alpha1-antitrypsin deficiency. Thorax. 2003;58(12):1020-1026. doi: http://www.ncbi.nlm.nih.gov/pubmed/14645964

69. Malerba M, Radaeli A, Ceriani L, Tantucci C, Grassi V. Airway hyperresponsiveness in a large group of subjects with alpha1-antitrypsin deficiency: a cross-sectional controlled study. J Intern Med. 2003;253(3):351-358. doi: http://www.ncbi.nlm.nih.gov/pubmed/12603503

70. Stolk J, Ng WH, Bakker ME, et al. Correlation between annual change in health status and computer tomography derived lung density in subjects with alpha1-antitrypsin deficiency.[Erratum appears in Thorax. 2004 Feb;59(2):179]. Thorax. 2003;58(12):1027-1030. doi: http://www.ncbi.nlm.nih.gov/pubmed/14645966

71. Takii Y, Inoue H, Karashima E, et al. Systemic vasculitis associated with alphal-antitrypsin deficiency. Intern Med. 2003;42(7):619-623. doi: http://www.ncbi.nlm.nih.gov/pubmed/12879958

72. Dowson LJ, Guest PJ, Stockley RA. The relationship of chronic sputum expectoration to physiologic, radiologic, and health status characteristics in alpha(1)-antitrypsin deficiency (PiZ). Chest. 2002;122(4):1247-1255. doi: http://www.ncbi.nlm.nih.gov/pubmed/12377849

73. Miravitlles M, Vila S, Torrella M, et al. Influence of deficient alpha1-anti-trypsin phenotypes on clinical characteristics and severity of asthma in adults. Respir Med. 2002;96(3):186-192. doi: http://www.ncbi.nlm.nih.gov/pubmed/11905553

74. Piitulainen E, Sveger T. Respiratory symptoms and lung function in young adults with severe alpha(1)-antitrypsin deficiency (PiZZ). Thorax. 2002;57(8):705-708. doi: http://www.ncbi.nlm.nih.gov/pubmed/12149531

75. Siafakas NM, Tzortzaki EG. Few smokers develop COPD. Why? Respir Med. 2002;96(8):615-624. doi: http://www.ncbi.nlm.nih.gov/pubmed/12195843

Non-specific Treatment of AATD Lung Disease

1. Tuohy MM, Remund KF, Hilfiker R, Murphy DT, Murray JG, Egan JJ. Endobronchial valve deployment in severe alpha-1 antitrypsin deficiency emphysema: a case series. Clin Respir J. 2013;7(1):45-52. doi: http://www.ncbi.nlm.nih.gov/pubmed/22236390

2. Alam S, Li Z, Janciauskiene S, Mahadeva R. Oxidation of Z alpha1-antitrypsin by cigarette smoke induces polymerization: a novel mechanism of early-onset emphysema. Am J Respir Cell Mol Biol. 2011;45(2):261-269. doi:http://www.ncbi.nlm.nih.gov/pubmed/20971880

3. El Fakir S, Nejjari C, Serhier Z, et al. Alpha 1 antitrypsin phenotypes and obstructive airway disease in subjects over 65 years of age: QUID R cohort. Age Ageing. 2011;40(5):627-629. doi: http://www.ncbi.nlm.nih.gov/pubmed/21791445

4. Karaaslan C, Hirakawa H, Yasumatsu R, et al. Elastase inhibitory activity of airway alpha1-antitrypsin is protected by treatment with a catalytic antioxidant in a baboon model of severe bronchopulmonary dysplasia. Pediatr Res. 2011;70(4):363-367. doi: http://www.ncbi.nlm.nih.gov/pubmed/21705962

5. Alam S, Li Z, Janciauskiene S, Mahadeva R. Cigarette smoke promotes polymerisation of Z alpha 1-antitrypsin. Thorax. 2010;65:A29-A30.

6. Sorheim I-C, Bakke P, Gulsvik A, et al. alpha1-Antitrypsin protease inhibitor MZ heterozygosity is associated with airflow obstruction in two large cohorts. Chest. 2010;138(5):1125-1132. doi: http://www.ncbi.nlm.nih.gov/pubmed/20595457

7. Stolk J, Cooper BG, Stoel B, et al. Retinoid treatment of Emphysema in Patients on the Alpha-1 International Registry. The REPAIR study: study design, methodology and quality control of study assessments. Therap. 2010;4(6):319-332. doi: http://www.ncbi.nlm.nih.gov/pubmed/20926506

8. Jain AB, Patel V, Apostolakos M, Sheikh B, Kashyap R, Orloff M. Effect of liver transplant on pulmonary functions in adult patients with Alpha 1 Antitrypsin (A1A) deficiency. Liver Transpl. 2009;15:S153-S154.

9. van der Meer RM, Wagena E, Ostelo RWJG, Jacobs AJE, van Schayck O. Smoking cessation for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2009(1). doi: http://www.ncbi.nlm.nih.gov/pubmed/12804448

10. Corda L, Bertella E, La Piana GE, Boni E, Redolfi S, Tantucci C. Inhaled corticosteroids as additional treatment in alpha-1-antitrypsin-deficiency-related COPD. Respiration. 2008;76(1):61-68. doi: http://www.ncbi.nlm.nih.gov/pubmed/18319586

11. Mora JI, Hadjiliadis D. Lung volume reduction surgery and lung transplantation in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2008;3(4):629-635. doi: http://www.ncbi.nlm.nih.gov/pubmed/19281079

12. Moreno P, Alvarez A, Algar FJ, et al. Incidence, management and clinical outcomes of patients with airway complications following lung transplantation. Eur J Cardiothorac Surg. 2008;34(6):1198-1205. doi: http://www.ncbi.nlm.nih.gov/pubmed/18824370

13. Boswell-Smith V, Spina D. PDE4 inhibitors as potential therapeutic agents in the treatment of COPD-focus on roflumilast. Int J Chron Obstruct Pulmon Dis. 2007;2(2):121-129. doi: http://www.ncbi.nlm.nih.gov/pubmed/18044684

14. Burton CM, Carlsen J, Mortensen J, Andersen CB, Milman N, Iversen M. Long-term Survival After Lung Transplantation Depends on Development and Severity of Bronchiolitis Obliterans Syndrome. J Heart Lung Transplant. 2007;26 (7):681-686. doi: http://www.ncbi.nlm.nih.gov/pubmed/17613397

15. Stoller JK, Gildea TR, Ries AL, Meli YM, Karafa MT. Lung Volume Reduction Surgery in Patients With Emphysema and alpha-1 Antitrypsin Deficiency. Ann Thorac Surg. 2007;83 (1):241-251. doi: http://www.ncbi.nlm.nih.gov/pubmed/17184672

16. Dauriat G, Mal H, Jebrak G, et al. Functional results of unilateral lung volume reduction surgery in alpha1-antitrypsin deficient patients. Int J Chron Obstruct Pulmon Dis. 2006;1(2):201-206. doi: http://www.ncbi.nlm.nih.gov/pubmed/18046897

17. Stavem K, Bjortuft O, Borgan O, Geiran O, Boe J. Lung transplantation in patients with chronic obstructive pulmonary disease in a national cohort is without obvious survival benefit. J Heart Lung Transplant. 2006;25(1):75-84. doi: http://www.ncbi.nlm.nih.gov/pubmed/16399534

18. Mal H, Guignabert C, Thabut G, et al. Recurrence of pulmonary emphysema in an alpha-1 proteinase inhibitor-deficient lung transplant recipient. Am J Respir Crit Care Med. 2004;170(7):811-814. doi: http://www.ncbi.nlm.nih.gov/pubmed/15184198

19. Tutic M, Bloch KE, Lardinois D, Brack T, Russi EW, Weder W. Long-term results after lung volume reduction surgery in patients with alpha1-antitrypsin deficiency. J Thorac Cardiovasc Surg. 2004;128(3):408-413. doi: http://www.ncbi.nlm.nih.gov/pubmed/15354100

20. Cassivi SD, Meyers BF, Battafarano RJ, et al. Thirteen-year experience in lung transplantation for emphysema. Ann Thorac Surg. 2002;74(5):1663-1669; discussion 1669-1670. doi: http://www.ncbi.nlm.nih.gov/pubmed/12440627

Liver Disease in AATD

1. Dawwas MF, Davies SE, Griffiths WJ, Lomas DA, Alexander GJ. Prevalence and risk factors for liver involvement in individuals with PiZZ-related lung disease. Am J Respir Crit Care Med. 2013;187(5):502-508. doi: http://www.ncbi.nlm.nih.gov/pubmed/23262512

2. Goltz D, Vossing LM, Kirfel J, Spengler U, Fischer HP. alpha1-antitrypsin PiMZ-mutation and alcoholic steatohepatitis are BI-directionally aggravating amplifiers in chronic liver disease. J Hepatol. 2013;58:S559.

3. Gorska K, Korczynski P, Struniawski R, et al. Heterozygous alpha1-antitrypsin deficiency in liver transplant candidates. Pol Arch Med Wewn. 2013;123(1-2):14-20. doi: http://www.ncbi.nlm.nih.gov/pubmed/23235492

4. Bugeja T, Gelson W, Massood A, Maguire G, Davies SE, Griffiths WJH. A high prevalence of heterozygous alpha-1 antitrypsin deficiency in patients with advanced parenchymal liver disease. J Hepatol. 2012;56:S227.

5. Nelson DR, Teckman J, Di Bisceglie AM, Brenner DA. Diagnosis and management of patients with alpha1-antitrypsin (A1AT) deficiency. Clin Gastroenterol Hepatol. 2012;10(6):575-580. doi: http://www.ncbi.nlm.nih.gov/pubmed/22200689

6. Barnes TC, Cross A, Anderson ME, Edwards SW, Moots RJ. Relative alpha1-antitrypsin deficiency in systemic sclerosis. Rheumatology (Oxford). 2011;50(8):1373-1378. doi: http://www.ncbi.nlm.nih.gov/pubmed/21454304

7. Ding J, Yannam GR, Roy-Chowdhury N, et al. Spontaneous hepatic repopulation in transgenic mice expressing mutant human alpha1-antitrypsin by wild-type donor hepatocytes. J Clin Invest. 2011;121(5):1930-1934. doi: http://www.ncbi.nlm.nih.gov/pubmed/21505264

8. Fox M, Fox J, Davies M. Diagnosis and management of chronic liver disease in older people. Rev Clin Gerontol. 2011;21(1):1-15.

9. Ezzikouri S, El Feydi AE, El Kihal L, et al. Prevalence of common HFE and SERPINA1 mutations in patients with hepatocellular carcinoma in a Moroccan population. Arch Med Res. 2008;39(2):236-241. doi: http://www.ncbi.nlm.nih.gov/pubmed/18164971

10. Stoller JK, Piliang M. Panniculitis in alpha-1 antitrypsin deficiency: A review. Clin Pulm Med. 2008;15 (2):113-117.

11. Regev A, Guaqueta C, Molina EG, et al. Does the heterozygous state of alpha-1 antitrypsin deficiency have a role in chronic liver diseases? Interim results of a large case-control study. J Pediatr Gastroenterol Nutr. 2006;43 Suppl 1:S30-35. doi: http://www.ncbi.nlm.nih.gov/pubmed/16819398

12. Scott BB, Egner W. Does alpha1-antitrypsin phenotype PiMZ increase the risk of fibrosis in liver disease due to hepatitis C virus infection? Eur J Gastroenterol Hepatol. 2006;18(5):521-523. doi: http://www.ncbi.nlm.nih.gov/pubmed/16607148

13. Bowlus CL, Willner I, Zern MA, et al. Factors associated with advanced liver disease in adults with alpha1-antitrypsin deficiency. Clin Gastroenterol Hepatol. 2005;3(4):390-396. doi: http://www.ncbi.nlm.nih.gov/pubmed/15822045

14. Hadzic N, Francavilla R, Chambers SM, Castellaneta S, Portmann B, Mieli-Vergani G. Outcome of PiSS and PiSZ alpha-1-antitrypsin deficiency presenting with liver involvement. Eur J Pediatr. 2005;164(4):250-252. doi: http://www.ncbi.nlm.nih.gov/pubmed/15711957

15. Kim JS, Tanaka N, Newell JD, et al. Nontuberculous mycobacterial infection: CT scan findings, genotype, and treatment responsiveness. Chest. 2005;128(6):3863-3869. doi: http://www.ncbi.nlm.nih.gov/pubmed/16354855

16. Rudnick DA, Perlmutter DH. Alpha-1-antitrypsin deficiency: a new paradigm for hepatocellular carcinoma in genetic liver disease. Hepatology. 2005;42(3):514-521. http://www.ncbi.nlm.nih.gov/pubmed/16044402

17. Montalto G, Cervello M, Giannitrapani L, Dantona F, Terranova A, Castagnetta LAM. Epidemiology, risk factors, and natural history of hepatocellular carcinoma. Ann N Y Acad Sci. 2002;963:13-20. doi: http://www.ncbi.nlm.nih.gov/pubmed/12095924

18. Pittschieler K. Liver involvement in alpha1-antitrypsin-deficient phenotypes PiSZ and PiMZ. Acta Paediatr. 2002;91(2):239-240. doi: http://www.ncbi.nlm.nih.gov/pubmed/11952016

Specific Treatment of AATD COPD

1. Campos M, Brantly M, Chen J, et al. Safety and tolerability of 120 mg/kg dose of alpha1-proteinase inhibitor in alpha-1 antitrypsin deficiency: A multicenter, randomized, double-blind, crossover study. Chest. 2012;142:742A. doi: http://www.ncbi.nlm.nih.gov/pubmed/71073079

2. Frenzel E, Aggarwal N, Wrenger S, Welte T, Janciauskiene S. Elastase inhibition is not a requirement for the anti-inflammatory effects of Alpha-1 Antitrypsin: Studies in neutrophil models. Pneumologie. 2012;66 (6).

3. Schmid ST, Koepke J, Dresel M, et al. The effects of weekly augmentation therapy in patients with PiZZ alpha1-antitrypsin deficiency. Int J Chron Obstruct Pulmon Dis. 2012;7:687-696. doi: http://www.ncbi.nlm.nih.gov/pubmed/23055718

4. Gotzsche PC, Johansen HK. Intravenous alpha-1 antitrypsin augmentation therapy for treating patients with alpha-1 antitrypsin deficiency and lung disease. Cochrane Database Syst Rev. 2010(7):CD007851. doi: http://www.ncbi.nlm.nih.gov/pubmed/20614465

5. McCarthy C, Dimitrov BD. Augmentation therapy for alpha-1 antitrypsin deficiency—not enough evidence to support its use yet! COPD. 2010;7(3):234; author reply 235-236. doi: http://www.ncbi.nlm.nih.gov/pubmed/20486824

6. Stockley RA, Parr DG, Piitulainen E, Stolk J, Stoel BC, Dirksen A. Therapeutic efficacy of alpha-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. doi: http://www.ncbi.nlm.nih.gov/pubmed/20920370

7. Stocks JM, Brantly ML, Wang-Smith L, et al. Pharmacokinetic comparability of Prolastin®-C to Prolastin® in alpha(1)-antitrypsin deficiency: a randomized study. BMC Clin Pharmacol. 2010;10:13. doi: http://www.ncbi.nlm.nih.gov/pubmed/20920295

8. Stocks JM, Brantly ML, Wang-Smith L, et al. Pharmacokinetic comparability of Prolastin-C to Prolastin in alpha1-antitrypsin deficiency: A randomized study. BMC Clin Pharml. 2010;10(13). doi: http://www.ncbi.nlm.nih.gov/pubmed/20920295

9. Chapman KR, Stockley RA, Dawkins C, Wilkes MM, Navickis RJ. Augmentation therapy for alpha1 antitrypsin deficiency: a meta-analysis. COPD. 2009;6(3):177-184. doi: http://www.ncbi.nlm.nih.gov/pubmed/19811373

10. Dirksen A, Piitulainen E, Parr DG, 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. doi: http://www.ncbi.nlm.nih.gov/pubmed/19196813

11. Parr DG, Dirksen A, Piitulainen E, Deng C, Wencker M, Stockley RA. Exploring the optimum approach to the use of CT densitometry in a randomised placebo-controlled study of augmentation therapy in alpha 1-antitrypsin deficiency. Respir Res. 2009;10:75. doi: http://www.ncbi.nlm.nih.gov/pubmed/19678952

12. Stoller JK, Fallat R, Schluchter MD, et al. Augmentation therapy with alpha1-antitrypsin: patterns of use and adverse events. 2003. Chest. 2009;136(5 Suppl):e30. doi: http://www.ncbi.nlm.nih.gov/pubmed/20162784

13. Tonelli AR, Rouhani F, Li N, Schreck P, Brantly ML. Alpha-1-antitrypsin augmentation therapy in deficient individuals enrolled in the Alpha-1 Foundation DNA and Tissue Bank. Int J Chron Obstruct Pulmon Dis. 2009;4:443-452. doi: http://www.ncbi.nlm.nih.gov/pubmed/20054436

14. Blanco I, Canto H, Flores J, et al. Long-term augmentation therapy with alpha-1 antitrypsin in an MZ-AAT severe persistent asthma. Monaldi Arch Chest Dis. 2008;69(4):178-182. doi: http://www.ncbi.nlm.nih.gov/pubmed/19350840

15. Heresi GA, Stoller JK. Augmentation therapy in alpha-1 antitrypsin deficiency. Expert Opin Biol Ther. 2008;8(4):515-526. doi: http://www.ncbi.nlm.nih.gov/pubmed/18352854

16. Sandhaus RA, Turino G, Stocks J, et al. alpha1-Antitrypsin augmentation therapy for PI*MZ heterozygotes: a cautionary note. Chest. 2008;134(4):831-834. doi: http://www.ncbi.nlm.nih.gov/pubmed/18842915

17. Sandstrom CS, Piitulainen E, Janciauskiene S. Augmentation therapy in emphysema patient with ZZ alpha-1-antitrypsin deficiency. Respir Med CME. 2008;1 (2):153-157.

18. Zamora NP, Pla RV, Del Rio PG, Margaleff RJ, Frias FR, Ronsano JBM. Intravenous human plasma-derived augmentation therapy in alpha 1-antitrypsin deficiency: from pharmacokinetic analysis to individualizing therapy.[Erratum appears in Ann Pharmacother. 2008 Jun;42(6):905 Note: Ronsano, Jose Bruono Montoro (corrected Ronsano, Jose Bruno Montoro)]. Ann Pharmacother. 2008;42(5):640-646. doi: http://www.ncbi.nlm.nih.gov/pubmed/18413692

19. Bernspang E, Sveger T, Piitulainen E. Respiratory symptoms and lung function in 30-year-old individuals with alpha-1-antitrypsin deficiency. Respir Med. 2007;101(9):1971-1976. doi: http://www.ncbi.nlm.nih.gov/pubmed/17532199

20. Dirksen A, Stockley RA, Piitulainen E, Shaker SB, Bernhardt T. A randomized, placebo-controlled, double-blind, exploratory study of the effect of augmentation therapy in patients with severe alpha-1 antitrysin deficiency (AATD) and chronic obstructive pulmonary disease (COPD) [Abstract]. American Thoracic Society International Conference, May. 2007.

21. Stocks JM, Brantly M. Biochemical efficacy and tolerability of a new augmentation preparation in two patients with MmaltonZ Alpha1-Antitrypsin deficiency [Abstract]. J Allergy Clin Immunol. 2007;119(1 Suppl):S301 [1178].

22. Dirksen A BT. The EXACTLIE trial a validation of computed tomography to show effects of treatment in alpha-1-antitrypsin (AAT) deficiency [Abstract]. Eur Respir J. 2006;28(Suppl 50):334s [2008].

23. Stocks JM, Brantly M, Pollock D, et al. Multi-center study: the biochemical efficacy, safety and tolerability of a new alpha1-proteinase inhibitor, Zemaira. COPD. 2006;3(1):17-23. doi: http://www.ncbi.nlm.nih.gov/pubmed/17175661

24. Gildea TR, Shermock KM, Singer ME, Stoller JK. Cost-effectiveness analysis of augmentation therapy for severe alpha1-antitrypsin deficiency. Am J Respir Crit Care Med. 2003;167(10):1387-1392. doi: http://www.ncbi.nlm.nih.gov/pubmed/12574076

25. Mullins CD, Wang J, Stoller JK. Major components of the direct medical costs of alpha1-antitrypsin deficiency. Chest. 2003;124(3):826-831. doi: http://www.ncbi.nlm.nih.gov/pubmed/12970004

26. Piitulainen E, Bernspang E, Bjorkman S, Berntorp E. Tailored pharmacokinetic dosing allows self-administration and reduces the cost of IV augmentation therapy with human alpha(1)-antitrypsin. Eur J Clin Pharmacol. 2003;59(2):151-156. doi: http://www.ncbi.nlm.nih.gov/pubmed/12728289

27. Stoller JK, Fallat R, Schluchter MD, et al. Augmentation therapy with alpha1-antitrypsin: patterns of use and adverse events. Chest. 2003;123(5):1425-1434. doi: http://www.ncbi.nlm.nih.gov/pubmed/12740257

28. Brantly M. Alpha1-antitrypsin: not just an antiprotease: extending the half-life of a natural anti-inflammatory molecule by conjugation with polyethylene glycol. Am J Respir Cell Mol Biol. 2002;27(6):652-654. doi: http://www.ncbi.nlm.nih.gov/pubmed/12444023

29. Mullins CD, Blatt L, Wang J. Societal implications of the pharmacoeconomics of alpha1-antitrypsin deficiency. Expert rev. 2002;2(3):243-249. doi: http://www.ncbi.nlm.nih.gov/pubmed/19807416

30. Stockley RA, Bayley DL, Unsal I, Dowson LJ. The effect of augmentation therapy on bronchial inflammation in alpha1-antitrypsin deficiency. Am J Respir Crit Care Med. 2002;165(11):1494-1498. doi: http://www.ncbi.nlm.nih.gov/pubmed/12045122

31. Stoller JK, Rouhani F, Brantly M, et al. Biochemical efficacy and safety of a new pooled human plasma alpha(1)-antitrypsin, Respitin. Chest. 2002;122(1):66-74. doi: http://www.ncbi.nlm.nih.gov/pubmed/12114340

Additional References

1. Bradi AC, Audisho N, Casey DK, Chapman KR. Alpha-1 antitrypsin deficiency in Canada: regional disparities in diagnosis and management. COPD. 2015;12 Suppl 1:15-21. doi: http://www.ncbi.nlm.nih.gov/pubmed/25938286

2. Chapman KR, Burdon JG, Piitulainen E, et al. Intravenous augmentation treatment and lung density in severe alpha1 antitrypsin deficiency (RAPID): a randomised, double-blind, placebo-controlled trial. Lancet. 2015;386(9991):360-368. doi: http://www.ncbi.nlm.nih.gov/pubmed/26026936

3. Mehta M, Chapman KR, Heffer M, Marras TK. Impact of pulmonary nontuberculous mycobacterial treatment on pulmonary function tests in patients with and without established obstructive lung disease. Respirology. 2015;20(6):987-993. doi: http://www.ncbi.nlm.nih.gov/pubmed/26011313

4. Schuh C, Fritscher LG, Chapman KR, Fritscher CC. The prevalence of asthma and atopy in schoolchildren from Porto Alegre, Brazil, has plateaued. Respir Med. 2015;109(3):308-311. doi: http://www.ncbi.nlm.nih.gov/pubmed/25683031

5. Tan WC, Sin DD, Bourbeau J, et al. Characteristics of COPD in never-smokers and ever-smokers in the general population: results from the CanCOLD study. Thorax. 2015;70(9):822-829. doi: http://www.ncbi.nlm.nih.gov/pubmed/26048404

6. Molloy K, Hersh CP, Morris VB, et al. Clarification of the risk of chronic obstructive pulmonary disease in alpha1-antitrypsin deficiency PiMZ heterozygotes. Am J Respir Crit Care Med. 2014;189(4):419-427. doi: http://www.ncbi.nlm.nih.gov/pubmed/24428606

7. Teckman J, Pardee E, Howell RR, et al. Appropriateness of Newborn Screening for alpha1-Antitrypsin Deficiency. J Pediatr Gastroenterol Nutr. 2014;58(2):201-205. doi: http://www.ncbi.nlm.nih.gov/pubmed/24121147

8. Teckman JH, Jain A. Advances in alpha-1-antitrypsin deficiency liver disease. Curr Gastroenterol Rep. 2014;16(1):367. doi: http://www.ncbi.nlm.nih.gov/pubmed/24338605

9. Lewis EC. Expanding the clinical indications for alpha1-antitrypsin therapy. Mol Med. 2012;18(6):957-970. doi: http://www.ncbi.nlm.nih.gov/pubmed/22634722

10. Ekeowa UI, Gooptu B, Belorgey D, et al. alpha1-Antitrypsin deficiency, chronic obstructive pulmonary disease and the serpinopathies. Clin Sci (Colch). 2009;116(12):837-850. doi: http://www.ncbi.nlm.nih.gov/pubmed/19426146

11. Greene CM, Miller SDW, Carroll T, et al. Alpha-1 antitrypsin deficiency: a conformational disease associated with lung and liver manifestations. J Inherit Metab Dis. 2008;31(1):21-34. http://www.ncbi.nlm.nih.gov/pubmed/18193338

12. Belorgey D, Hagglof P, Karlsson-Li S, Lomas DA. Protein misfolding and the serpinopathies. Prion. 2007;1(1):15-20. doi: http://www.ncbi.nlm.nih.gov/pubmed/19164889

13. Hericks AJ, Bhat A. An overview of alpha-1 antitrypsin deficiency. Mo Med. 2007;104(3):255-259. doi: http://www.ncbi.nlm.nih.gov/pubmed/17619502

14. Lomas DA. Genetic predisposition to chronic obstructive pulmonary disease: advances in alpha1-antitrypsin deficiency and the serpinopathies. Clin Med. 2007;7(5):446-447. doi: http://www.ncbi.nlm.nih.gov/pubmed/17990707

15. Mulgrew AT, Taggart CC, McElvaney NG. Alpha-1-antitrypsin deficiency: current concepts. Lung. 2007;185(4):191-201. doi: http://www.ncbi.nlm.nih.gov/pubmed/17562108

16. Rajangam S, Bijoor A. Metabolic diseases with single gene mode of inheritance - Review. Perinatology. 2007;9 (4):190-196.

17. Stolk J, Putter H, Bakker EM, et al. Progression parameters for emphysema: a clinical investigation. Respir Med. 2007;101(9):1924-1930. doi: http://www.ncbi.nlm.nih.gov/pubmed/17644366

18. Lomas DA. Parker B. Francis lectureship. Antitrypsin deficiency, the serpinopathies, and chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2006;3(6):499-501. doi: http://www.ncbi.nlm.nih.gov/pubmed/16921127

19. Churg A, Wright JL. Proteases and emphysema. Curr Opin Pulm Med. 2005;11(2):153-159. doi: http://www.ncbi.nlm.nih.gov/pubmed/15699789

20. Grigorenko EL. The inherent complexities of gene-environment interactions. J Gerontol B Psychol Sci Soc Sci. 2005;60 (SPEC. ISS.):53-64. doi: http://www.ncbi.nlm.nih.gov/pubmed/15863710

21. Gupta J, Bhadoria DP, Lal MK, et al. Association of the PIM3 allele of the alpha-1-antitrypsin gene with chronic obstructive pulmonary disease. Clin Biochem. 2005;38(5):489-491. doi: http://www.ncbi.nlm.nih.gov/pubmed/15820782

22. Lomas DA. Molecular mousetraps, alpha1-antitrypsin deficiency and the serpinopathies. Clin Med. 2005;5(3):249-257. doi: http://www.ncbi.nlm.nih.gov/pubmed/16011217

23. Spurzem JR, Rennard SI. Pathogenesis of COPD. Semin Respir Crit Care Med. 2005;26(2):142-153. doi: http://www.ncbi.nlm.nih.gov/pubmed/16088433

24. Stoller JK, Aboussouan LS. Alpha1-antitrypsin deficiency. Lancet. 2005;365(9478):2225-2236. doi: http://www.ncbi.nlm.nih.gov/pubmed/15978931

25. Crowther DC, Belorgey D, Miranda E, Kinghorn KJ, Sharp LK, Lomas DA. Practical genetics: alpha-1-antitrypsin deficiency and the serpinopathies. Eur J Hum Genet. 2004;12(3):167-172. doi: http://www.ncbi.nlm.nih.gov/pubmed/14694355

26. Lomas DA, Belorgey D, Mallya M, et al. Polymerisation underlies alpha1-antitrypsin deficiency, dementia and other serpinopathies. Front Biosci. 2004;9:2873-2891. doi: http://www.ncbi.nlm.nih.gov/pubmed/15353322

27. Lomas DA, Parfrey H. Alpha1-antitrypsin deficiency. 4: Molecular pathophysiology. Thorax. 2004;59(6):529-535. doi: http://www.ncbi.nlm.nih.gov/pubmed/15170041

28. Stoller JK. Alpha1-antitrypsin deficiency. Thorax. 2004;59(2):92-93. doi: http://www.ncbi.nlm.nih.gov/pubmed/14760142

29. Stoller JK, Snider GL, Brantly M, et al. Standards for the diagnosis and management of individuals with alpha-1 antitrypsin deficiency: Executive summary. Respir Care. 2004;48 (11):1043-1047.

30. Elzouki AN, Eriksson S. Risk of hepatobiliary disease in adults with severe alpha 1-antitrypsin deficiency (PiZZ): is chronic viral hepatitis B or C an additional risk factor for cirrhosis and hepatocellular carcinoma? Eur J Gastroenterol Hepatol. 1996;8(10):989-994. doi: http://www.ncbi.nlm.nih.gov/pubmed/8930564

31. King MB, Campbell EJ, Gray BH, Hertz MI. The proteinase-antiproteinase balance in alpha-1-proteinase inhibitor-deficient lung transplant recipients. Am J Respir Crit Care Med. 1994;149(4 Pt 1):966-971. doi: http://www.ncbi.nlm.nih.gov/pubmed/8143063

32. Guidelines for the approach to the patient with severe hereditary alpha-1-antitrypsin deficiency. American Thoracic Society. Am Rev Resp Dis. 1989;140(5):1494-1497. doi: http://www.ncbi.nlm.nih.gov/pubmed/2683912

33. Eriksson S. Alpha 1-antitrypsin deficiency and liver cirrhosis in adults. An analysis of 35 Swedish autopsied cases. Acta Medica Scand. 1987;221(5):461-467. doi: http://www.ncbi.nlm.nih.gov/pubmed/3496734

34. Eriksson S, Carlson J, Velez R. Risk of cirrhosis and primary liver cancer in alpha 1-antitrypsin deficiency. N Engl J Med. 1986;314(12):736-739. doi: http://www.ncbi.nlm.nih.gov/pubmed/3485248

1. Guidelines for the approach to the patient with severe hereditary alpha-1-antitrypsin deficiency. American Thoracic Society. Am Rev Respir Dis. 1989;140(5):1494-1497. doi: http://dx.doi.org/10.1164/ajrccm/140.5.1494

2. 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. doi: http://dx.doi.org/10.1164/rccm.168.7.818

3. Rahaghi FF, Sanhas RA, Strange C, et al. The prevalence of alpha-1 antitrypsin deficiency among patients found to have airflow obstruction. COPD. 2012; 9(4): 352-358. doi: http://dx.doi.org/10.3109/15412555.12012.669433

4. Silverman EK, Miletick JP, Pierce JA, Sherman LA, Endicott SK, Broze GJ Jr, Campbell EJ. Alpha-1-antitrypsin deficiency. High prevalence in the St. Louis area determined by direct population screening. Am Rev Respir Dis. 1989; 140 (4): 961-966. doi: http//dx.doi.org/10.1164/ajrccm/140.4.961

5. Chapman KR, Burdon JG, Piitulainen E, et al. Intravenous augmentation treatment and lung density in severe alpha1 antitrypsin deficiency (RAPID): a randomised, double-blind placebo-controlled trial. Lancet. 2015; 386(9991): 360-368. doi: http://dx.doi.org/10.1016/S0140-6736(15)60860-1

6. Aboussouan LS, Stoller JK. Detection of alpha-1 antitrypsin deficiency: a review. Respir Med. 2009;103(3):335-341. doi: http://dx.doi.org/10.1016/j.rmed.2008.10.006

7. Stoller JK, Brantly M. The challenge of detecting alpha-1 antitrypsin deficiency. COPD. 2013;10(S1):26-34. doi: http://dx.doi.org/10.3109/15412555.2013.763782

8. de Serres FJ. Worldwide racial and ethnic distribution of alpha-1antitrypsin deficiency: summary of an analysis of published genetic epidemiologic surveys. Chest. 2002;122(5):1818-1829. doi: http://dx.doi.org/10.1378/chest.122.5.1818

9. Bugeja T, Gelson W, Massood A, Maguire G, Davies SE, Griffiths WJH. A high prevalence of heterozygous alpha-1 antitrypsin deficiency in patients with advanced parenchymal liver disease. J Hepatol. 2012;56:S227. doi: http://dx.doi.org/10.1016/S0168-8278%2812%2960584-4

10. Stoller JK, Piliang M. Panniculitis in alpha-1 antitrypsin deficiency: A review. Clin Pulm Med. 2008;15 (2):113-117.doi: http://dx.doi.org/10.1097/CPM.0b013e3181672239

11. Parr DG, Guest PG, Reynolds JH, Dowson LJ, Stockley RA. Prevalence and impact of bronchiectasis in alpha1-antitrypsin deficiency. Am J Respir Crit Care Med. 2007;176 (12):1215-1221. doi: http:dx.doi.org/10.1164/rccm.200703-489OC

12. Mahr AD, Edberg JC, Stone JH, et al. Alpha-1antitrypsin deficiency-related alleles Z and S and the risk of Wegener's granulomatosis. Arthritis Rheum. 2010;62(12):3760-3767. doi: http://dx.doi.org/10.1002/art.27742

13. Morris H, Morgan MD, Wood AM, et al. ANCA-associated vasculitis is linked to carriage of the Z allele of alpha1 antitrypsin and its polymers. Ann Rheum Dis. 2011;70(10):1851-1856. doi: http://dx.doi.org/10.1136/ard.2011.153569

14. Strange C, Moseley MA, Jones Y, Schwarz L, Xie L, Brantly ML. Genetic testing of minors for alpha1-antitrypsin deficiency. Arch Pediatr Adolesc Med. 2006;160(5):531-534. doi: http://dx.doi.org/10.1001/archpedi.160.5.531

15. Teckman J, Pardee E, Howell RR, et al. Appropriateness of newborn screening for alpha1-antitrypsin deficiency. J Pediatr Gastroenterol Nutr. 2014;58(2):201-205. doi: http://dx.doi.org/10.1097/MPG.0000000000000196

16. American College of Medical Genetics. Standards and guidelines for clinical genetics laboratories: 2006 edition. American College of Medical Genetics website. https://www.acmg.negt/Pages/ACMG_Activities/stds-2002/URD.htm Published 2006. Accessed May 31, 2016.

17. Miravitlles M, Herr C, Ferrarotti I, et al. Laboratory testing of individuals with severe alpha1-antitrypsin deficiency in three European centres. Eur Respir J. 2010;35(5):960-968. doi: http://dx.doi.org/10.1183/09031936.00069709

18. Bornhorst JA, Greene DN, Ashwood ER, Grenache DG. Alpha-1antitrypsin phenotypes and associated serum protein concentrations in a large clinical population. Chest. 2013;143(4):1000-1008. doi: http://dx.doi.org/10.1378/chest.12-0564

19. Tanash HA, Nilsson PM, Nilsson JA, Piitulainen E. Clinical course and prognosis of never-smokers with severe alpha-1-antitrypsin deficiency (PiZZ). Thorax. 2008;63(12):1091-1095. doi: http://dx.doi.org/10.1136/thx.2008.095497

20. Tonelli AR, Brantly ML. Augmentation therapy in alpha-1 antitrypsin deficiency: advances and controversies. Ther Adv Respir Dis. 2010;4(5):289-312. doi: http://dx.doi.org/10.1177/1753465810373911

21. Stolk J, Putter H, Bakker EM, et al. Progression parameters for emphysema: a clinical investigation. Respir Med. 2007;101(9):1924-1930. doi: http://dx.doi.org/10.1016/j.rmed.2007.04.016

22. Dawkins PA, Dowson LJ, Guest PJ, Stockley RA. Predictors of mortality in alpha1-antitrypsin deficiency. Thorax. 2003;58(12):1020-1026. doi: http://dx.doi.org/10.1136/thorax.58.12.1020

23. Eriksson S, Carlson J, Velez R. Risk of cirrhosis and primary liver cancer in alpha 1-antitrypsin deficiency. N Engl J Med. 1986; 314:736-739. doi: http://dx.doi.org/10.1056/NEJM198603203141202

24. Nelson DR, Teckman J, Di Bisceglie AM, Brenner DA. Diagnosis and management of patients with alpha1-antitrypsin (A1AT) deficiency. Clin Gastroenterol Hepatol. 2012;10(6):575-580. doi: http://dx.doi.org/10.1016/j.cgh.2011.12.028

25. Dawwas MF, Davies SE, Griffiths WJ, Lomas DA, Alexander GJ. Prevalence and risk factors for liver involvement in individuals with PiZZ-related lung disease. Am J Respir Crit Care Med. 2013;187(5):502-508. doi: http://dx.doi.org/10.1164/rccm.201204-0739OC

26. Banga A, Gildea T, Rajeswaran J, Rokadia H, Blackstone EH, Stoller JK. The natural history of lung function after lung transplantation for alpha(1)-antitrypsin deficiency. Am J Respir Crit Care Med. 2014;190(3):274-281. doi: http://dx.doi.org/10.1164/rccm.201401-0031OC

27. Yusen RD, Edwards LB, Kucheryavaya MS, et al. The registry of the International Society of Heart and Lung Transplantation: Thirty-second official adult lung and heart-lung transplantation report--2015; Focus them: early graft failure. J Heart Lung Transplant. 2015; 34(10): 1264-1277. doi: http://dx.doi.org/10.1016/j.healun.2015.08.014

28. National Emphysema Treatment Trial Research Group. Patients at high risk of death after lung-volume-reduction surgery. N Engl J Med. 2001;345(15):1075-1083. doi: http://dx.doi.org/10.1056/nejmoa11798

29. Molloy K, Hersh CP, Morris VB, et al. Clarification of the risk of chronic obstructive pulmonary disease in alpha1-antitrypsin deficiency PiMZ heterozygotes. Am J Respir Crit Care Med. 2014;189(4):419-427. doi: http://dx.doi.org/10.1164/rccm.201311-1984OC

30. Carpenter MJ, Strange C, Jones Y, et al. Does genetic testing result in behavioral health change? Changes in smoking behavior following testing for alpha-1 antitrypsin deficiency. Ann Behav Med. 2007;33(1):22-28. doi: http://dx.doi.org/10.1207/s15324796abm3301_3

31. Stoller JK, Gildea TR, Ries AL, Meli YM, Karafa MT. Lung volume reduction surgery in patients with emphysema and alpha-1 antitrypsin deficiency. Ann Thorac Surg. 2007;83 (1):241-251. doi: http://dx.doi.org/10.1016/j.athoracsur.2006.07.080

32. Stoller JK, Tomashefski J, Jr., Crystal RG, et al. Mortality in individuals with severe deficiency of alpha1-antitrypsin: findings from the National Heart, Lung, and Blood Institute Registry. Chest. 2005;127(4):1196-1204. doi: http://dx.doi.org/10.1378/chest.127.4.1196

33. Campos MA, Kueppers F, Stocks JM, et al. Safety and pharmacokinetics of 120mg/kg versus 60mg/kg weekly intravenous infusions of alpha-1 proteinase inhibitor in alpha-1 antitrypsin deficiency: a multicenter, randomized, double-blind, crossover study (SPARK). COPD. 2013; 10(6): 687-695. doi: http://dx.doi.org/10.3109/15412555.2013.800852

34. Tonelli AR, Rouhani F, Li N, Schreck P, Brantly ML. Alpha-1-antitrypsin augmentation therapy in deficient individuals enrolled in the Alpha-1 Foundation DNA and Tissue Bank. Int J Chron Obstruct Pulmon Dis. 2009;4:443-452. doi: http://dx.doi.org/10.2147/COPD.S8577

35. Chapman KR, Stockley RA, Dawkins C, Wilkes MM, Navickis RJ. Augmentation therapy for alpha1 antitrypsin deficiency: a meta-analysis. COPD. 2009;6(3):177-184. doi: http://dx.doi.org/10.1080/15412550902905961

36. Gross B, Grebe M, Wencker M, Stoller JK, Bjursten LM, Janciauskiene S. New Findings in PiZZ alpha1-antitrypsin deficiency-related panniculitis. Demonstration of skin polymers and high dosing requirements of intravenous augmentation therapy. Dermatology. 2009;218(4):370-375. doi: http://dx.doi.org/10.1159/000202982

37. Sandhaus RA, Turino G, Stocks J, et al. Alpha-1 antitrypsin augmentation therapy for PI*MZ heterozygotes: a cautionary note. Chest. 2008;134(4):831-834. doi: http://dx.doi.org/10.1378/chest.08-0868

38. Molloy K, Hersh CP, Morris VB, et al. Clarification of the risk of chronic obstructive pulmonary disease in alpha-1 antitrypsin deficiency PiMZ heterozygotes. Am J Respir Crit Care Med. 2014; 189(4): 419-427. doi: http://dx.doi.org/10.1164/rccm.201311-1984OC.

39. Gildea TR, Shermock KM, Singer ME, Stoller JK. Cost-effectiveness analysis of augmentation therapy for severe alpha-1 antitrypsin deficiency. Am J Respir Crit Care Med. 2003;167(10):1387-1392. doi: http://dx.doi.org/10.1164/rccm.200209-1035OC

Share This Article

E-mail this article to a friend