Genetics in Asthma and COPD




Introduction


Asthma and chronic obstructive pulmonary disease (COPD) are chronic inflammatory airway diseases that result from an interaction between genetic or host susceptibility and different environmental exposures. Both airway diseases are common with an estimated 300 million individuals with asthma and 64 million individuals with COPD worldwide. Genetic variation has major effects on asthma and COPD susceptibility; however, these are not isolated to a single gene, but represent the effects of many different genes that are polymorphic (contain gene variants or polymorphisms) or undergo epigenetic regulation (changes in gene expression without changes in the genetic code, i.e., gene methylation), causing disease risk or severity ( Fig. 45-1 ). Thus, in contrast to single gene disorders such as cystic fibrosis, asthma and COPD are caused by the effects of multiple genes with smaller effects and genetically are referred to as complex genetic disorders . Recent genetic studies suggest that there may be common genetic variants that influence the susceptibility and severity of both of these “complex” obstructive airway diseases. Multiple potential environ­mental exposures interact with risk or severity genes for the development or progression of asthma, while cigarette smoke exposure is the primary environmental factor for the development of COPD.




Figure 45-1


Premise of genetic research in complex diseases.

Genetic research studies the role of genetic variability in determining risk for complex airway diseases, related phenotypes, and disease severity. Genetic risk can be altered by gene variants that directly impact biologic function or act through gene-by-environment interactions that alter gene function or expression through epigenetic mechanisms. Pharmacogenetics research studies a similar gene-by-environment interaction by analyzing the effect of genotype and exposure to a medication in determining interindividual responses to pharmacologic therapies. IgE, immunoglobulin E; SNP, single nucleotide polymorphism.


Information from genetic studies has advanced remarkably since the sequencing of the first genome in 2001 and the completion of the International HapMap project in 2005. Based on initial studies in families, it is clear that susceptibility to asthma and COPD is determined by multiple genetic loci in different genes. Since then, high-throughput genotyping techniques using chip technologies for genome-wide association studies (GWAS) have facilitated comprehensive assessment of gene variation in a larger number of subjects and, more recently, sequencing of the whole genome or of all of the exomes. In contrast to asthma, which may be recognized as early as infancy, COPD presents at a later age; therefore, family-based studies are not practical or feasible. Thus, candidate gene association and GWAS have been used to investigate the genetic basis of COPD. The exponential growth of the field of asthma and COPD genetics in the past decade has dramatically shortened the time from identification of phenotype to discovery of the gene variant in these complex airway diseases.


Different genetic studies have focused on disease susceptibility, disease severity, and therapeutic responses (i.e., pharmacogenetics). These genetic approaches require large, well-characterized populations. Unfortunately, because of the large population samples required for GWAS, the emphasis on comprehensive phenotyping has not been given priority and the results of these resource-intensive genetic studies have not always provided adequate information on subpopulations and disease heterogeneity. Overall, genomic approaches will provide a basis for the development of personalized medical strategies where molecular profiles based on variation in multiple genes or gene pathways will be used to develop a predictive genetic disease or severity score. Such a score could be used to predict which individuals are at risk for asthma or COPD and would facilitate the development of strategies to prevent disease development or progression. In those who already have these diseases, genetic profiles would provide a molecular rationale for disease heterogeneity, progression, and the basis for variable responses to therapeutic agents. These genetic profiles should facilitate precise, personalized, and patient-focused therapeutic approaches with the potential to target disease progression, maximize therapeutic efficacy, and minimize unwanted side effects or negative responses.


This chapter outlines how human genetic studies in asthma and COPD have identified gene targets, which has substantially improved understanding of the molecular basis of the development, progression, and pharmacogenetics in asthma and COPD. These genetic approaches have the potential to contribute to the development of precision or personalized medicine in obstructive airway disease. These same genetic approaches have been used in other complex diseases (e.g., cardiovascular, metabolic, cancer) to identify genetic determinants of disease susceptibility and progression.




Early Genetic Studies of Asthma Susceptibility: Family-Based Genetic Studies


Evidence That Asthma Is Heritable


A genetic basis for asthma was recognized as early as 1860 when Henry Salter reported that asthma was a heritable disease with “distinct traces of inheritance” among related individuals. Salter’s observation was confirmed in studies demonstrating that asthma prevalence was between 20% and 25% among first-degree relatives of asthma subjects, significantly higher than the approximately 5% prevalence in the general population. The clustering of asthma in families is estimated by comparing the risk for developing asthma in those related to an asthmatic proband with the risk for asthma in the general population as a lambda ratio R ). A higher λ R indicates that the disease has greater heritability and, therefore, a stronger genetic component. For instance, asthma has a λ R of 2 to 4 while an autosomal recessive disease such as cystic fibrosis has a much higher λ R . The lower λ R of a common, complex disease such as asthma is likely caused by multiple gene variants each with small effects interacting with factors not detected in heritability studies, including a diverse group of environmental exposures and disease triggers. The observed heritability for asthma demonstrated in these early studies provided the rationale for the comprehensive genetic studies discussed in this chapter.


The First Genome-Wide Screens for Susceptibility Loci: Family-Based Studies


The first genetic studies for asthma susceptibility, which were performed before the first genomes were sequenced, used family-based approaches. They used genetic variants that are equally spaced throughout the genome to identify chromosomal susceptibility regions for asthma or associated phenotypes (e.g., bronchial hyperresponsiveness, serum IgE levels, skin test atopy). In family-based genetic studies, linkage analysis is used to identify genetic loci that cosegregate or are coinherited with a trait such as asthma in families. The degree of linkage is measured as an LOD score, the “likelihood of the odds” that a genomic region and trait cosegregate ( Table 45-1 ). An LOD score of 3 or more is considered significant and is equal to a 1000 : 1 odds that a genomic region is in linkage or coinherited with a specific trait or phenotype. Family-based studies have been successful in identifying genetic diseases with an autosomal dominant or recessive mode of inheritance, but are less powerful for complex or multifactorial diseases wherein multiple genes interact to determine disease susceptibility or severity. Transmission disequilibrium testing is another family-based association test that compares the transmission of a genetic marker from both parents to their affected offspring with a disease or trait of interest (see Table 45-1 and Fig. 45-2 ).



Table 45-1

Different Methodologies * for Genetic Studies







































Methodologies Population Genome Coverage Test Measurement
Family, twin, or segregation study Families None Clustering in families to estimate heritability
Linkage study Families (pedigrees) Genome-wide Cosegregation of a marker and phenotype with the likelihood of the odds score
Family-based association test Small families (trios) Gene to genome-wide Transmission of a marker from parents to cases-controls with transmission disequilibrium testing
Candidate gene association study Cases and controls Gene Odds ratio or genetic risk and P- value
Genome-wide association study Cases and controls Genome-wide Odds ratio or genetic risk and P- value
Admixture mapping Cases and controls Gene to Genome-wide Admixture mapping association peak and P- value

* Methodologies are described by the target population analyzed, coverage ranging from the individual gene-level to genome-wide level resolution, and the analytical test measured.




Figure 45-2


Two commonly used association testing methods: family-based and case-control.

A, Family-based association testing. The basic fundamental unit of family-based association testing are the trios of affected (cases in red) probands. Trios consist of cases or affected probands and their parents, whose affected status is not necessarily known (in gray). In family-based studies, the over transmission of an allele of a polymorphism to affected probands is measured ( red arrows ). This demonstrates how a single nucleotide polymorphism (SNP) with alleles “A” or “G” shows overtransmission of the “A” allele to affected probands. The strength of the overtransmission or association at this locus is estimated using the P- value. B, Case-control association testing. Case-control association testing compares the frequency of the different alleles for this SNP in unrelated cases (in red) and controls (in blue) to determine an odds ratio (OR, red double-arrow) or genetic effect. The basic illustration demonstrates that the “A” allele is more frequent among cases compared to controls. The strength of the association at this locus is estimated using the P- value.


Early family-based genome-wide studies demonstrated that multiple genes are associated with development of asthma and of related phenotypes such as atopy and elevated serum immunoglobulin E (IgE). These studies, including those performed by the National Heart, Lung, and Blood Institute (NHLBI) Collaborative Study on the Genetics of Asthma (CSGA), provided evidence for linkage with asthma and related phenotypes in multiple different chromosomal regions: chromosome 5q, 6p, 7q, 11q, 12q, 14q, and 16q. The genetic markers identified by these family-based studies represent chromosomal regions that have subsequently been studied to identify specific risk genes responsible for these linkages ( eTable 45-1 ).




eTable 45-1

Candidate Genes for Susceptibility to Asthma and Related Phenotypes in Chromosomal Regions Identified through Linkage Studies *




































































































Gene Names Chr Pos Gene ID Associated Phenotypes References
A Disintegrin And Metalloprotease-33 20p13 ADAM33 Asthma, bronchial responsiveness, lung function, lung function rate decline
β-chain of the high-affinity receptor for IgE 11q13 FCER1B Asthma, IgE, skin test atopy
Dipeptidyl Peptidase-10 2q14 DPP10 Asthma, IgE, skin test atopy
Interleukin-4 5q31 IL4 Asthma, IgE, skin test atopy, lung function
α-chain of the Interleukin-4 Receptor 5q31 IL4R Asthma, IgE, skin test atopy, lung function
Interleukin-13 5q31 IL13 Asthma, bronchial responsiveness, IgE, skin test atopy
CD14 5q31 CD14 Asthma
Protocadherin-1 5q31 PCDH1 Bronchial responsiveness
Tumor Necrosis Factor-α 6p21 TNF Asthma, bronchial responsiveness, IgE, skin test atopy
Human Leukocyte Antigen Complex DPB1 6p21 HLA-DPB1 Asthma
Human Leukocyte Antigen Complex DQB1 6p21 HLA-DQB1 Asthma
Human Leukocyte Antigen Complex DRB1 6p21 HLA-DRB1 Asthma, skin test atopy
Human Leukocyte Antigen Complex G 6p22 HLA-G Asthma, bronchial responsiveness
Neuropeptide S Receptor-1 (NPSR1) 7p14 GPRA Asthma, IgE
PHD Finger Protein-11 13q14 PHF11 Asthma, IgE

* Candidate genes listed are within linkage regions from family-based studies or replicated in case-control association studies. Genes are summarized by full gene name, chromosomal position (Chr Pos), gene identification (ID) acronym, associated asthma or related phenotype, and references cited.



These early linkage studies demonstrated gene-gene interactions in asthma susceptibility between chromosomes 5q31, 8p23, and 12q22, and 15q13. In addition, these studies provided evidence for gene-by-environment interactions between passive cigarette smoking and asthma susceptibility at chromosomes 1p, 5q, and 9q, and 17q21. These gene-by-environment interactions provided the earliest examples of important epigenetic effects in asthma in which environmentally induced DNA modifications alter transcription without a change in gene sequence.


Early family-based linkage studies also provided evidence for genetic determinants of altered pulmonary function in asthma. In these studies, a quantitative trait locus on chromosome 5q33 and 2q32 was linked with important lung function measures. The quantitative trait locus in chromosome 2q32 was adjacent to the region linked to forced expiratory volume in 1 second/forced vital capacity (FEV 1 /FVC) in families with early-onset COPD suggesting that there may be similar genes that affect the development of both asthma and COPD.


Linkage Studies Provide Important Insight into the Complexity of Asthma Pathogenesis


The limitations of family-based linkage studies include the challenge of recruiting and characterizing related subjects and the use of genetic markers that cover genomic regions that contain hundreds of genes, requiring more detailed analysis with fine mapping or positional cloning to identify individual susceptibility genes. Another important limitation of family-based linkage studies is that they were designed to identify loci containing gene variants with a strong effect on disease susceptibility and are underpowered to detect common gene variation with a weak or modest effect. Despite these limitations, novel asthma genes have been identified (see eTable 45-1 ) which provide insight into the polygenic pathogenesis and the importance of gene-environment interactions in asthma.


Linkage Analysis and Positional Cloning Reveal Novel Asthma Susceptibility Loci


A disintegrin and metalloprotease-33 gene (ADAM33) was the first gene identified for asthma susceptibility using a family-based linkage study followed by fine mapping and association studies with cases and controls (i.e., positional cloning). It was identified by positional cloning on chromosome 20p13. ADAM33 encodes for a membrane-anchored glycoprotein that controls cell-matrix interactions and is involved in regulation of lung growth and morphogenesis. The ADAM33 protein is expressed in airway smooth muscle cells and lung fibroblasts, contains a catalytic domain with proteolytic activity and affects airway remodeling. ADAM33 is among the most replicated genes for asthma susceptibility in different racial and ethnic groups. ADAM33 gene variants have been associated with accelerated decline in lung function in asthma and in COPD, suggesting a role of this gene in the pathogenesis of progressive airflow obstruction. This is further supported by the observation that bronchoalveolar lavage concentrations of soluble ADAM33 correlate with asthma severity and inversely correlate with lung function.


Other family-based whole-genome studies used positional cloning to identify the dipeptidyl peptidase-10 ( DPP10 ) gene on chromosome 2q14 as a locus for asthma susceptibility. A s ingle nucleotide polymorphism (SNP) in DPP10 was recently identified in a GWAS of African Americans and African Caribbeans, providing additional evidence that DPP10 is an asthma susceptibility locus. Dipeptidyl peptidases cleave the terminal peptides of cytokines and chemokines and enhance airway inflammation. Several other asthma susceptibility genes have been identified using fine mapping or positional cloning techniques and include the protocadherin-1 gene (PCDH1) on chromosome 5q31, human leukocyte antigen complex G (HLA-G) on chromosome 6p22, the n europeptide S receptor-1 gene (GPRA) on chromosome 7p14, and PHD finger protein-11 (PHF11) on chromosome 13q14 (see eTable 45-1 ). The identification of asthma susceptibility loci is an example of the use of unbiased genetic techniques, that is, without preconceived notions of mechanism, to delineate novel biologic mechanisms that are important in the pathogenesis of asthma.




Candidate Gene Association Studies in Asthma


As high-throughput genotyping technologies emerged that facilitated genotyping in larger populations, a greater number of biologic candidate genes were investigated in unrelated asthma case and control populations. In contrast to family-based approaches, case-control association studies compare the frequency of the alleles of a gene variant between unrelated cases and controls (see Fig. 45-2 ). Association analysis is a very useful approach for common diseases wherein alleles confer a small disease risk. SNPs (i.e., point mutations) represent the substitution of a single nucleotide for another, resulting in variable frequencies of the variant allele in a sample population. SNPs are denoted as a reference sequence (rs) number or by the coding change at a specific codon position number for nonsynonymous SNPs (e.g., rs1042713 or Gly 16 Arg) and represent the primary gene variants used in association studies. ( Nonsynonymous refers to a change in sequence that alters the amino acid sequence of a protein; synonymous, on the other hand, is a change in sequence that does not alter the protein.)


More than 100 genes have been studied as biologic candidate genes based on plausible biologic mechanisms in asthma or locations in chromosomal regions linked to asthma (see eTable 45-1 ). For each replicated candidate gene for asthma susceptibility that has been discovered, there are studies that may not show an association, perhaps because of differences in phenotypes or differences in race between populations (see later). The most replicated candidate genes appear to be related to broad categories of lung development (e.g., ADAM33 ), the type 2 T helper cell (Th2) inflammatory pathway (IL4, IL13, IL4R), innate immunity (HLA-DRB1, HLA-DQB1, CD14), and cellular inflammation (TNF, FCER1B, DPP10) (see eTable 45-1 ). In addition, candidate genes within the glucocorticoid receptor complex pathway, the leukotriene pathway (LTC4S), the β 2 -adrenergic receptor gene (ADRB2), and the Th2 inflammatory pathway signaling (IL4R) have also been evaluated as potential pharmacogenetic loci that modify responses to pharmacologic therapy.


Highly Replicated Candidate Genes on Chromosome 5q31 and Evidence for Gene-Gene Interactions


Linkage analyses of chromosome 5q31-35 have consistently provided evidence that this region is linked to asthma or closely related phenotypes. This inflammatory gene–rich region contains multiple candidate genes related to allergic inflammation, including IL3, IL4, IL5, IL13, CD14, serine peptidase inhibitor Kazal type 5 (SPINK5), the β 2 -adrenergic receptor (ADRB2), and leukotriene C4 synthase (LTC4S). The gene encoding for IL-13 and IL-4 contains promoter SNPs and variants consistently associated with asthma and related phenotypes in multiple populations. IL-13 and IL-4 bind to the α chain of the IL-4 receptor encoded by IL4R on chromosome 16p12, which also contains variants associated with asthma and related phenotypes. The interaction of two variants in IL4R has been associated with risk for life-threatening asthma exacerbations in persistent asthmatics suggesting a role of this pathway in determining disease severity. In addition, interactions between genetic variants in IL4, IL13, and IL4R and other pathway-related genes such as STAT6 on chromosome 12q13 cumulatively determine asthma risk and serum IgE at a greater level than does each individual locus, supporting the role of multiple genes interacting in asthma susceptibility and pathogenesis. Finally, one of the first GWAS in a population of severe or difficult-to-treat asthmatics demonstrated SNPs in IL13 and a neighboring gene encoding for a DNA repair protein (RAD50) as a locus for asthma susceptibility. The interaction of variants in different Th2 inflammatory pathway signaling genes has been critical to understanding the nature of gene-gene interactions in determining asthma susceptibility and severity. Furthermore, these positive associations for the IL-4–IL-13 pathways have supported the current development of biologic therapies targeting these cytokines.


Limitations of Candidate Gene Association Studies: Lessons Learned and the Road to GWAS


All association studies, including candidate gene studies, are limited by the need to ascertain well-characterized case-control populations. An issue with early association studies was the small sample sizes (<200 cases) resulting in studies underpowered to identify gene variants with small effects and prone to false-positive results. Unrelated cases and controls have variable ancestral backgrounds, which may lead to false-positive results. SNP allele frequencies vary in different populations based on ancestry (population stratification), which may cause spurious differences in allele frequencies between cases and controls if different ethnic groups are analyzed (i.e., false positives).


The need to account for potential population stratification in genetic association studies is true for all types of association studies but was well recognized and implemented in candidate gene studies. Another limitation of all association studies is the underlying correlation of alleles in genetic loci located in a chromosomal region (i.e., linkage disequilibrium or LD). For example, if two genetic loci are physically close together, they will be inherited as one unit through multiple generations. In this case, an association study would result in positive results for both genes, making it difficult to isolate the gene or variant of interest. This is especially true for SNPs within a gene. However, in older populations of more generations or admixed population, there have been more opportunities to break the correlation, thus allowing the true risk allele or loci to be identified.




Genome-Wide Association Studies for Asthma Susceptibility


In GWAS, large numbers of SNPs are genotyped using chip technologies that cover the genome, usually with 500,000 to more than 1 million SNP genotypes (SNP variants) assessed in large case and control populations. Earlier chips contained fewer SNPs and did not provide good coverage of the genome in all races that show differences in allele frequencies based on ancestry. The overall limitation is the degree of phenotyping that can be performed in these large samples (see Fig. 45-2 ). In GWAS, the frequencies of the alleles of each SNP are compared between unrelated cases and controls on a genome-wide scale (see Fig. 45-2 ).


SNPs are the most common form of gene variation and are seen in 1 of every 100 nucleotides in the human genome; therefore, SNPs used for GWAS provide comprehensive coverage of each gene and the entire genome. The ability to genotype a larger number of SNPs rapidly and simultaneously has enabled GWAS approaches to identify a number of novel loci associated with asthma susceptibility, related phenotypes, and disease severity. In addition, whole-genome genotyping provides information about population substructure, an index of ancestral admixture. All GWAS studies discussed later have adjusted associations by population substructure to minimize confounding by population stratification ( eTable 45-2 ). A small number of studies have used genome-wide family-based association studies (i.e., transmission disequilibrium test) to minimize this source of confounding by population stratification (see Fig. 45-2 ). GWAS requires a more stringent P- value due to multiple testing of a large number of SNPs, and subsequent replication testing of significant SNP associations in independent samples is important.




eTable 45-2

Major Susceptibility Loci for Asthma Identified through GWAS *

































































































































































































Gene Names Chr Pos Gene ID Racial or Ethnic Populations Initial Reported Locus References
ORM1-Like 3 17q12 ORMDL3 European, European American, African American, African Caribbean, Hispanic, Chinese rs7216389
Gasderminlike B 17q12 GSDMB European, European American, Chinese rs2305480
Interleukin-1 Receptor 2q12 IL1RL1 European, European American, African American, African Caribbean, Hispanic rs1420101
Interleukin-18 Receptor 2q12 IL18R1 European rs3771166
Human Leukocyte Antigen Complex DQB1 6p21 HLA-DQB1 European and Japanese rs9273349
Interleukin-33 9p24 IL33 European, European American, African American, African Caribbean, Hispanic rs1342326
Interleukin-2 Receptor, β Subunit 22q12 IL2RB European rs2284033
SMAD Family Member 3 15q22 SMAD3 European rs744910
RAR-Related Orphan Receptor A 15q22 RORA European rs11071559
Thymic Stromal Lymphopoietin 5q22 TSLP European American, African American, African Caribbean, Hispanic, Japanese rs1837253
WD Repeat Domain 36 5q22 WDR36 European, East Asian rs2416257
RAD50 Homolog 5q31 RAD50 European American rs2244012
Interleukin-13 5q31 IL13 European American rs1295686
cAMP-Specific Phosphodiesterase 4D 5q11 PDE4D European, European American, Hispanic rs1588265
Pyrin and HIN Domain Family Member 1 1q23 PYHIN1 African American and African Caribbean rs1102000
Interleukin-6 Receptor 1q21 IL6R European rs4129267
GRB2-Associated Binding Protein 1 4q31 GAB1 Japanese rs1397527
Ubiquitin Specific Peptidase 38 4q31 USP38 Japanese rs7686660
GATA Binding Protein 3 10p14 GATA3 Japanese rs10508372
Ikaros Family Zinc Finger 4 12q13 IKZF4 Japanese rs1701704
Human Leukocyte Antigen Complex DPA1 6p21 HLA-DPA1 Japanese rs987870
α-1B-Adrenergic Receptor 5q33 ADRA1B African American rs10515807
Prion-Related Protein 20p12 PRNP African American rs6052761
Dipeptidyl Peptidase-10 2q14 DPP10 African American rs1435879
TNFAIP3 Interacting Protein 1 5q33 TNIP1 European American rs1422673
Human Leukocyte Antigen Complex DRA 6p21 HLA-DRA European American rs2395185

* Susceptibility loci for asthma identified through genome-wide association studies summarized by full gene name, chromosomal position (Chr Pos), gene identification (ID) acronym, racial or ethnic groups where associations were observed, initial locus (by single nucleotide polymorphism rs number) reported to be associated with asthma through GWAS, and references cited.



GWAS Identifies ORMDL3 as a Susceptibility Locus


In 2007, a GWAS for asthma susceptibility was performed in a European cohort (GABRIEL). SNPs in the ORM1-like 3 gene (ORMDL3) region on chromosome 17q12 were significantly associated with the diagnosis of childhood asthma and this finding was replicated in two independent cohorts. A second larger GWAS by the European GABRIEL consortium validated ORMDL3 as a locus for asthma risk while identifying additional susceptibility loci in the neighboring gasdermin-like genes ( GSDMB and GSDMA ) as well as other loci in the genome, including IL1RL1 / IL18RL1 (between the IL1 and IL18 receptor genes, rs3771166) on chromosome 2q12, HLA-DQB1 (rs9273349), IL33 on chromosome 9p24 (rs1342326), SMAD3 on chromosome 15q22 (rs744910), and IL2RB on chromosome 22q12 (rs2284033).


Gene variants in the ORMDL3 locus have been associated with asthma in multiple subsequent GWAS, making ORMDL3 the most replicated locus using genome-wide screening methods in case-control populations. The challenge of interpreting genetic associations in chromosome 17q12 is the strong linkage disequilibrium (LD) that exists between the variants in multiple genes that span this region. It remains unclear whether the causative gene is ORMDL3 or an adjacent gene, such as GSDMB or GSDMA .


This novel asthma susceptibility gene (ORMDL3) is a member of a class of genes that encodes transmembrane proteins anchored in the endoplasmic reticulum; however, its specific biologic role in asthma pathogenesis remains unknown. GWAS in asthma have demonstrated a predilection for genes that regulate relevant molecular pathways and are related to epithelial damage and adaptive immune response. It is likely that altered production of cytokines such as thymic stromal lymphopoietin (TSLP) and IL-33 from damaged or disrupted epithelial cells promotes Th2-mediated inflammatory responses by activating IL1RL1 receptors on mast cells, Th2 lymphocytes, and regulatory T cells.


GWAS in African Americans and Other Asthma Populations


Subsequent GWAS were performed in multiethnic populations including non-Hispanic white and African American subjects to understand the genetic diversity of populations from different ancestral backgrounds (i.e., ethnic groups) as well as to address the genetic causes of differences in the frequency and severity of asthma in different ethnicities. This is particularly important for ethnic groups such as African Americans and Puerto Ricans who have a greater asthma incidence associated with higher asthma-related morbidity and mortality. These groups may have unique genetic loci due to a higher frequency of risk alleles that determine disease susceptibility or severity. For example, African Americans are at low risk for cystic fibrosis because of the low frequency of CFTR mutations in this ancestral population while at higher risk for sickle cell disease due to increased frequency of mutations in that gene.


The EVE consortium combined 5416 asthma cases in a large, multiethnic population representing Americans of European descent, African Americans, African Caribbeans, and Hispanics (Mexican Americans and Puerto Ricans) with replication in 12,649 subjects. This large-scale multiethnic GWAS for asthma susceptibility identified genome-wide significant associations with SNPs on chromosome 17q12 (spanning IKZF3 / ZPBP2 / GSDMB / ORMDL3 ), IL1RL1, and TSLP on chromosome 5q22. Associations at these loci and a SNP in IL33 were replicated in independent cohorts. This GWAS validated the susceptibility loci identified on the chromosome 17q12 locus and IL1RL1 confirming their importance in asthma risk (see eTable 45-2 ).


The EVE consortium also identified SNPs in the pyrin and HIN domain family member 1 ( IFIX, interferon inducible nuclear protein X gene, PYHIN1 ) associated with asthma in African American and African Caribbean cohorts. The first GWAS of a population from a primarily African ancestry also identified novel susceptibility loci in the α 1B -adrenergic receptor on chromosome 5q33 (ADRA1B) and prion-related protein on chromosome 20p12 (PRNP). In addition, the DPP10 locus was associated with asthma, confirming earlier positional cloning studies.


GWAS in Asian populations confirm the importance of previously described asthma susceptibility loci in HLA-DQ, HLA-DPA1, HLA-DPB1, and TSLP in Japanese and Korean populations. In addition, five polymorphisms in chromosome 17q12 encompassing ORMDL3, GSDMB, ZPBP2, and IKZF3 were associated with asthma in a Han Chinese population (see eTable 45-2 ).


Admixture Mapping as an Alternative Approach in Admixed Populations


The colonization of the Americas by the Europeans and the subsequent African slave trade has resulted in the mixing of genetic ancestries and the complex population structures encountered in the genomes of African Americans, African Caribbeans, Mexican Americans, and Puerto Ricans. There is marked variability in the demographic histories of these admixed populations of African origin, resulting in a complex population structure and reduced LD that is not fully captured with GWAS that use genotyping platforms designed for non-Hispanic white populations. The recent mixing of these ancestries provides the rationale for genome-wide ancestry-based approaches. African Americans, on average, have an estimated 20% European ancestry (80% African) while Hispanic ethnic groups such as Puerto Ricans and Mexican Americans have a combination of three different ancestries.


Mapping by admixture linkage (admixture mapping, Fig. 45-3 ) is a genome-wide approach that is based on the principle that many SNPs show marked variable allele frequencies between different ancestral populations. In admixture mapping, estimates of ancestry at each SNP are tested for association with a phenotype, in contrast to GWAS which compares allele frequencies. Admixture mapping requires a substantially smaller number of genetic markers compared to GWAS and can evaluate regions with rare variants ( Fig 45-4 ) and other variants such as polynucleotide insertion-deletions. The optimal setting for the use of admixture mapping is in admixed populations where there are marked racial disparities in disease phenotype not attributed completely to environmental factors.




Figure 45-3


Illustration of admixture mapping.

A, The hypothesis behind mapping by admixture linkage disequilibrium or admixture mapping is that chromosomes from an admixed population (shown with red and blue genetic regions from different ancestries) contain a susceptibility allele that is more frequent in the red ancestral region versus the blue. Admixture mapping identifies increased ancestry at a susceptibility locus in cases compared to controls (region intersected by thick black line). B, Loci with significant associations between ancestry and disease risk are represented by admixture mapping peaks or chromosomal regions with an overrepresentation of ancestry from the ancestral population with the highest proportion of risk alleles at the locus containing the risk-invoking variant.

( A, Adapted from Montana G, Pritchard JK: Statistical tests for admixture mapping with case-control and cases-only data. Am J Hum Genet 75:771–789, 2004. B, From Patterson N, Hattangadi N, Lane B, et al: Methods for high-density admixture mapping of disease genes. Am J Hum Genet 74:979–1000, 2004, Fig. 1.)



Figure 45-4


Impact of genetic variants in human disease.

The “common disease–common allele” hypothesis states that multiple common genetic variants with small to modest effect sizes contribute to common disease susceptibility in an additive fashion. Genome-wide association studies (GWAS) have identified multiple common variants associated with risk for different common diseases. In contrast, the “common disease–rare allele” hypothesis states that rare genetic variants with a large effect size contribute to susceptibility for common diseases. Rare variants can only be evaluated with family-based genetic studies, DNA sequencing, and admixture mapping, and are not easily evaluated with GWAS.

(Adapted from Tsuji S: Genetics of neurodegenerative diseases: insights from high-throughput resequencing. Hum Mol Genet 19:R65–70, 2010.)


Studies of African ancestry in African Americans from the general population have suggested a role for gene variation related to African ancestry on lung function and asthma severity. Estimates of African ancestry are inversely associated with baseline lung function measures in three independent African American populations and Puerto Ricans. In African Americans, each percentage of African ancestry was associated with a 3 to 5 mL lower FEV 1 . This finding has implications for calculating predicted normal lung function in admixed ethnic groups. For example, an African American with 50% African Ancestry is likely to have an FEV 1 up to 200 mL higher compared to an African American of the same age, sex, and height with 90% African ancestry. Estimates of African ancestry have also been associated with risk for self-reported asthma and asthma exacerbations in African Americans from the United States.


Genome-wide admixture mapping studies have the potential to identify the genetic loci that account for the increased asthma incidence in those with significant African ancestry. Admixture mapping for susceptibility loci has been performed in Puerto Ricans and Mexican Americans from the Genetics of Asthma in Latino Americans (GALA) cohort, and SNPs in nearly 62 loci associated with asthma (i.e., admixture mapping peaks, see Fig. 45-3 ) have been identified. Admixture mapping in African American and Puerto Rican asthma subjects from the NHLBI-sponsored Severe Asthma Research Program (SARP), CSGA, and GALA cohorts has also identified a novel asthma susceptibility locus on chromosome 6q14.1.




GWAS and Association Studies of Severe Asthma


Initially, GWAS in asthma were designed to detect susceptibility loci, because detailed phenotyping is time intensive and costly, resulting in small case-control cohorts. Larger populations of comprehensively characterized cohorts were recruited for the NHLBI-sponsored SARP and the TENOR (The Epidemiology and Natural History of Asthma: Outcomes and Treatment Regimens) Study. A GWAS of severe asthmatics from TENOR identified multiple loci associated with asthma: rs2244012 in RAD50 adjacent to IL13 , rs1063355 in HLA-DQB1, and rs3998159 between HLA-DQB1 and HLA-DQA2 . These findings confirm the importance of Th2 cytokine and antigen presentation genes in asthma pathogenesis and potentially in asthma severity. Associations at these loci have also provided evidence for a common immunogenic pathway between asthma and autoimmune disease. For example, the IL13 asthma locus is also associated with psoriasis. A recent GWAS of children between 2 and 6 years of age from the Copenhagen Prospective Studies on Asthma in Childhood Exacerbation cohort with recurrent, severe exacerbations identified four previously identified susceptibility loci in GSDMB, IL33, RAD50, and ILRL1 as associated with risk for this severe asthma phenotype. In addition, this GWAS identified a novel locus for severe childhood asthma on the gene encoding cadherin-related family member-3 (CDRH3): a genotype-phenotype relationship that is important in determining severity risk in childhood asthma.


GWAS have also identified asthma susceptibility loci associated with lung function, a fundamental determinant of asthma severity. A SNP in the IL-6 receptor gene (rs4129267, IL6R ) has been associated with asthma in a large GWAS and was among the top SNPs associated with baseline lung function in a GWAS of the Framingham Heart Study cohort. This IL6R SNP is in LD with a coding variant (Asp 358 Ala) where the minor allele (Ala 358 ) is associated with lower baseline lung function and increased methacholine bronchial responsiveness. IL-6 is a proinflammatory cytokine expressed in inflammatory diseases such as asthma and COPD, and represents a potential therapeutic target because IL-6 receptor antagonists are available for the treatment of rheumatoid arthritis.


Li and coworkers performed a meta-analysis of 14 SNPs previously associated with lung function in GWAS of controls from the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) and SpiroMeta consortia. This analysis demonstrated that SNPs in the gene encoding for hedgehog-interacting protein (HHIP) were significantly associated with lung function in African American and non-Hispanic White asthma subjects. In addition, an increase in the number of risk alleles for SNPs in HHIP, PTCH1, and FAM13A resulted in a significant stepwise decrease in lung function. More recent studies have shown that Th1 immunity pathway genes (IL12A, IL12RB1, STAT4, IRF2) also appear to influence lung function and severity in asthma.


Elevated serum IgE is a primary associated phenotype for allergy in asthma. Two large GWAS of general population cohorts from the GABRIEL and EVE consortia have identified SNPs in FCER1A on chromosome 1q23, RAD50, STAT6, IL13, and the HLA complex associated with serum IgE concentrations, confirming prior linkage and association studies. In non-Hispanic whites, four SNPs in the chromosome 11 open reading frame and the leucine rich repeat containing 32 gene (C11orf30 LRRC32) were associated with serum IgE. The C11orf30 LRRC32 genomic region contained SNPs associated with other inflammatory diseases such as atopic dermatitis, childhood eczema, and Crohn disease.


Blood eosinophil levels and eosinophilic inflammation are related to asthma pathogenesis and severity and have been evaluated by GWAS. In a general population sample from Iceland and replication cohorts from Europe and East Asia SNPs in IL1RL1 (rs1420101), chromosome 2q13 (rs12619285), chromosome 3q21 (rs4857855), chromosome 5q31 (rs4143832), and SH2B3 on chromosome 12q24 (rs3184504) were associated with peripheral blood eosinophilia. The IL1RL1 locus has been associated with asthma and loci in WDR36 and IL33 showed suggestive associations with eosinophil counts and atopic asthma. Loci adjacent to WDR36 ( TSLP ), IL1R1, and IL33 have also been associated with asthma in prior GWAS. Fraction of exhaled nitric oxide, a biomarker of eosinophilic airway inflammation, was evaluated in a GWAS in 14 pediatric cohorts that included asthmatics and control subjects. This GWAS identified loci associated with childhood exhaled nitric oxide levels in two genes on chromosome 17q11.2-q12, LYRM9 and inducible nitric oxide synthase-2 (rs944722, NOS2 ), and adjacent to a previously replicated asthma susceptibility locus, GSDMB (rs8069176).


Chitinases are a family of conserved hydrolases that cleave chitin and mediate Th2-driven airway inflammation in murine models. YKL-40 is a chitinase-like protein that has been evaluated as a biomarker for airway inflammation and remodeling in asthma. Serum YKL-40 levels are elevated in asthmatics and have been associated with disease severity. A GWAS identified a SNP in the promoter of the chitinase 3-like 1 gene, which was associated with serum YKL-40 levels, asthma diagnosis, bronchial responsiveness, and lung function in an isolated population (Hutterites).


GWAS of lung function and inflammatory biomarkers have identified genes that impact disease severity. The impact of “lung function genes” on asthma severity provides an excellent example of how genetic markers from different genes can contribute to disease severity in an additive manner. Gene variants important in asthma susceptibility likely differ from genes that cause asthma progression and severity. This concept is outlined in Figure 45-5 , which proposes mechanisms by which susceptibility and severity genes interact in the development and progression of asthma. Thus, susceptibility to asthma is genetically determined by the gene variants identified in GWAS of asthma susceptibility. Disease progression is modulated by additional genes that determine asthma severity. For example, if the genome of an individual has gene variants conferring asthma susceptibility and also contains gene variants that encode for reduced lung function, this additive combination will result in the expression of a more severe asthma phenotype. Furthermore, genetic markers of asthma severity could be used to generate genetic profiles that predict risk for disease progression.




Figure 45-5


Genetic factors determine susceptibility and severity of asthma through independent pathogenic pathways.

This flow diagram demonstrates how susceptibility loci for asthma and related phenotypes (including allergic or atopic characteristics) contribute to risk for asthma. In turn, different genetic determinants for lung function such as loci that accelerate airway remodeling or alter early lung development in combination with altered T type 1 helper (Th1) innate immunity in response to infection result in asthma progression. Many of the genes for risk of asthma susceptibility, progression, and severity are from different gene pathways.

(Adapted from Li X, Hawkins GA, Ampleford EJ, et al: Genome-wide association study identifies Th1 pathway genes associated with lung function in asthmatic patients. J Allergy Clin Immunol 132:313–320 e15, 2013. Figure 3.)




Gene-Environment Interactions: Genetic and Epigenetic Studies for Asthma Susceptibility and Severity


Environmental factors have an important role in the pathogenesis of asthma and gene-environment interactions have been identified for asthma susceptibility and severity. Gene-environment interactions have the potential to identify factors that contribute to the missing heritability not accounted for by the effects of gene variants alone. Environmental cigarette smoke exposure is the most replicated environmental factor impacting genetic studies for asthma susceptibility. Linkage studies in families from the CSGA cohort in the United States and The Netherlands both identified a linkage signal for asthma susceptibility and bronchial responsiveness on chromosome 5q31 that was dependent on passive cigarette smoke exposure. Another linkage study in French families from the EGEA (Epidemiological Study on the Genetics and Environment of Asthma) cohort also detected linkage for markers on chromosome 17q12 that was dependent on a gene-environment interaction with smoke exposure. In a family-based association study of 36 SNPs from the chromosome 17q12 in the ORMDL3 region in the EGEA cohort, Bouzigon and coworkers demonstrated 11 SNPs associated with an increased risk for asthma; however, the risk for early-onset asthma for the SNP with the strongest association (rs8069176) was 2.9 times greater in the subgroup exposed to cigarette smoke compared with the unexposed subgroup. These effects may be mediated by epigenetic regulation and demonstrate the complexity and interaction of genetic and environmental interactions in the pathogenesis of asthma.


Dietary factors such as vitamin D deficiency are associated with asthma development and severity. A genome-wide family-based association study identified genetic risk factors that interact with serum vitamin D levels affecting asthma severity. Three SNPs in the class I MHC-restricted T cell–associated molecule gene (CRTAM) were associated with an increased frequency of asthma exacerbations with low vitamin D levels. CRTAM is expressed in lung and by natural killer T and CD8 + cells; therefore, it is plausible that these gene variants interact with vitamin D, altering the immune response to viral infections.


The list of genes associated with asthma or related phenotypes is large and demonstrates that there are a number of biologic pathways important in asthma pathogenesis (see eTables 45-1 and 45-2 ). However, with the use of systems biologic approaches, there is potential for the development of genetic risk and severity profiles that can be used for early disease diagnosis and prediction of asthma progression.




Pharmacogenetics of Asthma


Rationale for Pharmacogenetic Research in Asthma


Pharmacogenetics examines the role of gene variation in therapeutic responses to pharmacologic therapies. Pharmacogenetics is a gene-environment interaction wherein the therapeutic agent is the environmental exposure. There is evidence that genetic factors play an important role in the observed variability in therapeutic responses to different drugs. Approximately, 70% to 80% of asthmatics show varying responses to common antiasthma therapies, resulting in a marked variance in therapeutic drug responses. Genetic variation might contribute to a large percentage of the variability in drug response, whether beneficial or adverse. The rationale for pharmacogenetic studies in asthma is that genetic markers could be used to identify subgroups of nonresponders, responders, or a subgroup at risk for adverse responses.


Pharmacogenetic research in asthma is driven by two major challenges related to asthma management. First, a small subgroup of approximately 5% to 10% of asthmatics experiences uncontrolled symptoms and recurrent exacerbations despite treatment with multiple antiasthma therapies, including high doses of inhaled or oral corticosteroids. This population with refractory asthma experiences substantial morbidity and generates a major financial burden compared to those with controlled mild or inter­mittent asthma. Second, adverse effects have been associated with some asthma therapies, particularly rare, life-threatening events associated with the use β 2 -adrenergic receptor agonists (i.e., β-agonists). Pharmacogenetic studies have the potential to identify genetic markers that would personalize therapeutic approaches in individual asthmatics to optimize therapeutic response and prevent adverse side effects.


Pharmacogenetic studies of asthma therapies have been based on the primary clinical end points used in asthma clinical trials. In most pharmacogenetic studies, these predetermined clinical end points are analyzed for genetic associations with candidate gene studies or GWAS after the clinical trial is completed. These pharmacogenetic studies are important for the identification of genes or gene variants that influence drug responses. A minority of pharmacogenetic studies employs a prospective, genotype-stratified study design in which DNA is collected and genotyped for a risk variant of interest and subjects are randomized based on their genotype to treatment or placebo groups. The advantage of a prospective, genotype-stratified approach is that it can ensure sufficient statistical power to analyze less common genetic variants because the population is recruited based on a predefined risk gene variant. However, a genotype-stratified pharmacogenetic trial is feasible if only a limited number (one or two) genotypes are being considered.


Glucocorticoid Pharmacogenetics


Inhaled corticosteroids (ICS) have consistently been shown to be the most effective therapy to control persistent asthma. However, some asthmatics respond poorly to ICS. The pharmacogenetics of the glucocorticoid pathway is based on genes that code for a complex pathway consisting of the glucocorticoid biosynthetic pathway, chaperone proteins, and the cytosolic receptor heterocomplex ( eTable 45-3 ).




eTable 45-3

Pharmacogenetic Candidate Genes in Asthma *









































































































































































































Drug Classes Gene Associated Loci Study Designs Response Phenotype Sample Size References
Inhaled glucocorticoids: CRHR1 rs242941, rs1876828 Candidate gene study FEV 1 response 1117
(Fluticasone, budesonide, flunisolide, triamcinolone) STIP1 rs2236647, rs6591838, rs1011219 Candidate gene study FEV 1 response 382
TBX21 rs2240017 (His33Gln) Candidate gene study Bronchoprotection 53, 701
GLCCI1 rs37972=rs37973 GWAS FEV 1 response 1053
T Gene rs3127412, rs6456042, rs3099266, rs2305089 GWAS FEV 1 response 815
ADCY9 rs2230739 (Met 772 Ile) Candidate gene study Long-term FEV 1 response 86
Leukotriene receptor modifiers: ALOX5 Promoter repeat, rs892690, rs2029253, rs2115819 Candidate gene study FEV 1 response 114, 577
5-lipooxygenase inhibitors (ABT-761 and zileuton) LTC4S rs272431 Candidate gene study FEV 1 response 577
MRP1 rs215066, rs119774 Candidate gene study FEV 1 response 577
Cysteinyl leukotriene antagonists (montelukast) ALOX5 Promoter repeat, rs2115819 Candidate gene study FEV 1 response 61, 174
LTC4S rs730012 Candidate gene study Exacerbation risk 61
LTA4H rs266845 Candidate gene study Exacerbation risk 61
MRP1 rs119774 Candidate gene study FEV 1 response 61
Inhaled β 2 -adrenergic receptor agonists: CRHR2 rs7793837 Candidate gene study Acute FEV 1 bronchodilation 1186
2145
Short-acting β-agonists (Albuterol) ADCY9 rs2230739 (Met 772 Ile) Candidate gene study Acute FEV 1 bronchodilation 436
2145
ADRB2 rs1042713 (Gly 16 Arg) Retrospective and prospective, genotype stratified Acute FEV 1 bronchodilation 16–707
Long-term PEFR response 78
Candidate gene study Long-term PEFR response 108
ARG1 rs2781659, rs2781667 Candidate gene study Acute FEV 1 bronchodilation 200–2145
ARG2 rs7140310, rs10483801 Candidate gene study Acute FEV 1 bronchodilation 200
NOS3 rs1799983 (Asp 298 Glu) Candidate gene study Acute FEV 1 bronchodilation 81
SPATS2L rs295137 GWAS Acute FEV 1 bronchodilation 2145
Long-acting β-agonists
(salmeterol and formoterol)
ADCY9 rs2230739 (Met 772 Ile) Candidate gene study Long-term FEV 1 response 86
ADRB2 rs1042713 (Gly 16 Arg) Candidate gene study Long-term PEFR response 48–108
Retrospective and prospective, genotype-stratified prospective, genotype-stratified No effect on PEFR response 87–2655
Bronchoprotection 87, 152
Preference for montelukast or LABA as add-on to ICS 62

ICS, inhaled corticosteroids; FEV 1 , forced expiratory volume in 1 second; GWAS, genome-wide association study; LABA, long-acting β-agonists; PEFR, peak flow rate.

Reproduced from Ortega VE, Wechsler ME: Asthma pharmacogenetics: responding to the call for a personalized approach. Curr Opin Allergy Clin Immunol 13:399–409, 2013.

* Biologic candidate genes are summarized by drug class, associated polymorphisms (rs number and coding change, if relevant), study design, number of cohorts studied, population size, and response phenotype for which a pharmacogenetic effect has been detected.



A candidate gene analysis of the corticotropin-releasing hormone ( CRHR1 ) gene in more than 1000 asthma subjects from the National Institutes of Health and industry clinical trial cohorts identified two SNPs (rs242941 and rs1876828) that were associated with a change in lung function during ICS treatment. In another study, the gene coding for a glucocorticoid receptor complex chaperone protein, the heat shock organizing protein (STIP1), identified three SNPs (rs2236647, rs6591838, and rs1011219) that were also associated with a significant change in lung function during ICS treatment. The corticosteroid pathway interacts with other gene pathways that have been evaluated using a candidate gene approach in different clinical trial cohorts. The “T-box expressed in T-cells transcription factor” is encoded by TBX21 and is an important regulator of the naive T-lymphocyte development pathway. TBX21 contains a coding SNP (rs2240017, His 33 Gln) that was associated with “bronchoprotection,” that is, a reduction in bronchial hyperresponsiveness, during ICS treatment in the NHLBI Childhood Asthma Management Program (CAMP) and a Korean cohort. Adenylyl cyclase type 9 is a key enzyme in the β 2 -adrenergic receptor pathway and is encoded by ADCY9, which contains a coding SNP (rs2230739, Met 772 Ile) that has been associated with an albuterol bronchodilator response in the CAMP cohort during ICS treatment.


Use of GWAS approaches has the potential to identify novel pharmacogenetic loci for ICS response. The first GWAS evaluating ICS treatment response was performed using family-based testing in CAMP followed by association studies for replication in more than 900 asthma subjects from four independent clinical trial cohorts. A promoter SNP in the glucocorticoid-induced transcript-1 gene (rs37972 in GLCCI1 ) was associated with changes in lung function in response to ICS treatment. An in vitro study was also performed in which cells were transfected with another promoter variant in LD with rs37972 (rs37973) resulting in decreased gene expression. These studies were primarily performed in children with fewer years of exposure to corticosteroids compared to adult asthmatics. Interestingly, more recent studies using large clinical trial cohorts of adult asthmatics were not able to replicate this association. Functionally, GLCCI1 is an important regulator of apoptosis in response to glucocorticoids; therefore, this promoter variant may delay apoptosis of eosinophils during ICS therapy, thereby modulating therapeutic responses in asthma.


Another GWAS for ICS response was performed in asthma subjects from the NHLBI CAMP, Asthma Clinical Research Network (ACRN), and Childhood Asthma Research and Education Network. This GWAS identified SNPs in the T gene (rs3127412 and rs6456042) that were associated with changes in FEV 1 during ICS treatment. Subsequent detailed genotyping identified SNPs (rs3099266, rs2305089, rs1134481) within functional regions of the T gene. Pharmacogenetic loci in the glucocorticoid pathway, GLCCI1, and the T gene account for a small proportion of the interindividual variability for ICS therapeutic responses in asthma. It has yet to be determined whether these loci have additive effects with other pathway-related gene variants or are independent determinants of ICS response. Future pharmacogenetic studies in independent, large clinical trial cohorts or genotype-stratified trials are necessary to confirm and characterize these genes associated with ICS therapeutic responses and develop a genetic profile that may be used to predict therapeutic responsiveness to corticosteroids.


Cysteinyl Leukotriene Pharmacogenetics


The cysteinyl leukotriene pathway is an inflammatory pathway containing potential pharmacogenetic loci for two classes of antiasthma therapies: 5-lipoxygenase (5-LO) inhibitors (e.g., zileuton) and cysteinyl leukotriene antagonists (e.g., montelukast and zafirlukast). The cysteinyl leukotriene biosynthetic pathway is initiated by 5-LO (encoded by ALOX5 ) followed by leukotriene A 4 hydrolase (LTA4H), and leukotriene C4 synthase (LTC4S). Synthesized leukotrienes are transported to the extracellular space by multidrug resistance protein 1 ( MRP1 ) and activate cysteinyl leukotriene receptors ( CYSLTR1 and CYSLTR2 ).


ALOX5 has a tandem repeat polymorphism in its promoter that has been associated with changes in lung function during treatment with leukotriene antagonists. An analysis of the cysteinyl leukotriene pathway in asthmatics treated with zileuton also identified SNPs in ALOX5 (rs892690, rs2029253, and rs2115819), LTC4S (rs272431), and MRP1 (rs215066 and rs119774) that were associated with changes in lung function during 5-LO inhibition. Variation in leukotriene pathway genes has been reported in additional, smaller studies. These results support the finding that some of the variation in therapeutic responses to leukotriene modifiers is regulated by pharmacogenetic mechanisms (see eTable 45-3 ).


β 2 -Adrenergic Receptor Pharmacogenetics


Inhaled β-agonist treatment for asthma includes short-acting β-agonists (SABA) and long-acting β-agonists (LABA). β-Agonists activate a G protein–coupled receptor pathway via adenylyl cyclase type 9 that regulates airway smooth muscle relaxation. Inhaled β-agonists are the most commonly used treatment for asthma despite having been associated with very rare life-threatening adverse responses since the 1960s.


While various analyses have attempted to explain these “mini-epidemics” of asthma mortality, the U.S. Food and Drug Administration issued a boxed warning for all LABA-containing inhalers regarding the risk for life-threatening exacerbations. This LABA safety controversy is being evaluated in 46,800 asthma subjects who are currently being recruited for the U.S. Food and Drug Administration–mandated international LABA safety study. Pharmacogenetic studies have attempted to identify genetic markers for β-agonist response to identify a subgroup that is susceptible to these serious adverse effects. These studies have primarily focused on genes related to the β 2 -adrenergic receptor and nitric oxide synthetic pathways (see eTable 45-3 ).


The gene encoding the β 2 -adrenergic receptor (ADRB2) has a single exon and is located on chromosome 5q31 with more than 49 polymorphisms identified. The most studied of these ADRB2 polymorphisms is the common coding variant, Gly 16 Arg. In vitro, receptors expressing the Gly 16 variant show increased receptor down-regulation in response to β-agonist stimulation compared to Arg 16 .


Two early pharmacogenetic studies of ADRB2 in asthmatic children demonstrated that Arg 16 homozygotes experienced a greater bronchodilator response to a single dose of albuterol compared to Gly 16 homozygotes. This effect of Gly 16 Arg genotypes on acute SABA bronchodilator response was confirmed in small asthma populations. In contrast, pharmacogenetic studies of long-term, regular SABA treatment in two independent clinical trial cohorts demonstrated effects on therapeutic responses: Arg 16 homozygotes experienced a decline in peak flow rate (PEFR) during chronic SABA treatment while Gly 16 homozygotes did not experience changes in PEFR.


In a genotype-stratified, crossover trial, the ACRN, Gly 16 and Arg 16 homozygotes were randomized to treatment with regular albuterol or placebo. Gly 16 homozygotes showed an increase in PEFR during regular albuterol treatment while Arg 16 homozygotes showed no changes in PEFR with regular albuterol but had an increase in PEFR during as-needed albuterol treatment ( Fig. 45-6 ). In addition, regular albuterol treatment resulted in decreased rescue inhaler use and asthma symptom scores in Gly 16 homozygotes, while Arg 16 homozygotes experienced a deterioration of these secondary outcomes. Although the adverse effects of regular SABA treatment in Arg 16 homozygotes are important to understand, asthma guidelines do not recommend regular scheduled SABA in asthma. However, regular scheduled LABA is used to control asthmatics who are symptomatic on ICS alone. Thus, pharmacogenetic effects related to LABA exposure would have major therapeutic implications




Figure 45-6


The Asthma Clinical Research Network Beta Agonist Response by Genotype (BARGE) Trial.

The BARGE trial evaluated the effect of genotypes in response to regular short-acting β-agonist (albuterol) therapy or placebo given four times daily for 16-week treatment periods. For patients with the Gly/Gly genotype, the PEFR improved significantly more when they were treated with albuterol (red line) compared with placebo. In contrast, patients with the Arg/Arg genotype improved more when treated with placebo (blue line) compared with albuterol. PEFR, peak flow rate.

(Adapted from Israel E, Chinchilli VM, Ford JG, et al: Use of regularly scheduled albuterol treatment in asthma: genotype-stratified, randomised, placebo-controlled cross-over trial. Lancet 364:1505–1512, 2004. Figure 3.)


The genotypic effects of Gly 16 Arg during long-term LABA therapy were evaluated in two small trial arms from larger NHLBI ACRN clinical trials. Results from these trials showed a potential for reduced effectiveness of LABA in Arg 16 homozygotes. Subsequent prospective clinical trials and pharmacogenetic analyses with larger cohorts including two genotype-stratified trials did not show evidence for reduced therapeutic effectiveness of LABA therapy in Arg 16 homozygotes compared to Gly 16 homozygotes. Interestingly, in one of these prospective genotype-stratified trials, Gly 16 homozygotes experienced a more prolonged protection from methacholine-induced bronchoconstriction than was observed in Arg 16 homozygotes. Another genotype-stratified study randomized 62 Arg 16 homozygotes to either montelukast or LABA in addition to ICS therapy and demonstrated greater symptom control with the addition of montelukast. This study suggested that the Gly 16 Arg locus could interact with other therapies in asthma.


In contrast to a common variant such as Gly 16 Arg that likely exhibits smaller effects, it is possible that rare genetic variants with strong effects might be responsible for some of the uncommon and severe adverse responses associated with LABA treatment in asthma (see Fig. 45-4 ). A rare variant within the fourth transmembrane domain of ADRB2 , Thr 164 Ile, results in a protein with decreased β 2 -adrenergic receptor ligand binding, coupling to G s protein, and sequestration in response to SABAs and LABAs in vitro. This rare variant also significantly impairs the binding of salmeterol to its “exosite,” or secondary binding site, on the β 2 -adrenergic receptor. The Ile 164 allele was associated with an increased risk of airflow obstruction and reduced lung function in a cross-sectional population-based study of nearly 60,000 subjects. Thr 164 Ile and a 25 base pair insertion variant found only in African Americans, have also been associated with severe exacerbations requiring hospitalization in asthma patients treated with a LABA. Thr 164 Ile has also been associated with poor symptom control during LABA treatment in two independent cohorts. This finding of a rare genetic variant and adverse LABA effects has the potential to identify a very important at-risk asthma subpopulation. Thus, rare variants such as those in ADRB2 are potential biomarkers for more personalized, precise guideline-based treatment strategies in the small subset of asthmatics that have altered responsiveness to the combination therapy of a LABA with ICS. In addition, a recent study combining admixture mapping and GWAS in Puerto Ricans and Mexican asthmatic children from GALA has identified rare variants in solute carrier genes associated with bronchodilator response to SABAs, providing further evidence for the role of rare genetic variation as pharmacogenetic loci.


Association studies of candidate genes related to the β 2 -adrenergic receptor pathway have identified additional common polymorphisms that have been associated with the acute bronchodilator response to SABA. ADCY9 is a canonical β 2 -adrenergic receptor pathway gene with a common coding variant, Ile 772 Met, associated with increased acute FEV 1 bronchodilator response to albuterol during ICS therapy in children from the CAMP cohort. The effect of Met 772 Ile genotypes on the bronchodilator response to β-agonists during ICS therapy was also reported in a Korean population treated with a LABA (formoterol). CRHR2 is another pathway-related gene with SNPs that have been associated with the SABA response. CRHR2 encodes the corticotropin-releasing hormone receptor-2, a G-coupled protein receptor that regulates relaxation of airway smooth muscle through a signaling pathway similar to that of the β 2 -adrenergic receptor (via activation of adenylyl cyclase and protein kinase A). Five SNPs in CRHR2 were associated with acute bronchodilator responses to SABA in asthma subjects from three independent clinical trial cohorts.


Association studies of genes indirectly related to the β 2 -adrenergic receptor pathway have identified variants within the nitric oxide biosynthetic pathway associated with acute SABA bronchodilator response. An association study of 111 genes from the β 2 -adrenergic receptor and glucocorticoid pathways in subjects from the CAMP cohort and three independent trial cohorts identified a SNP in arginase 1 (ARG1), rs2781659, which was associated with an acute bronchodilator response. SNPs in ARG2 have also been associated with a SABA bronchodilator response. Arginase 1 and 2 metabolize l -arginine, which is a natural substrate for nitric oxide synthase, resulting in the production of nitric oxide. Nitric oxide is an endogenous bronchodilator; therefore, ARG1 and ARG2 polymorphisms may alter airway smooth muscle relaxation during β-agonist treatment.


Finally, a GWAS reported by Himes and colleagues identified a novel genetic locus for acute SABA bronchodilator responses. The primary GWAS analyzed 1644 asthmatics from six clinical trial cohorts with replication assessed in 1051 subjects from SARP and a Dutch asthma cohort. A SNP within the promoter region of the spermatogenesis associated, serine-rich 2–like gene (rs295137 in SPATS2L ) was associated with the acute bronchodilator response to albuterol. In human airway smooth muscle cells, the functional importance of SPATS2L in the β 2 -adrenergic receptor pathway was confirmed after a knockdown of this gene resulted in increased β 2 -adrenergic receptor expression. The investigators also confirmed previous associations for three candidate genes for albuterol bronchodilator response: ADCY9, CRHR2, and ARG1 .


Limitation of Current Pharmacogenetic Associations and Future Directions


Most of the pharmacogenetic associations for therapeutic response do not account for a significant amount of the variability in drug responses. In part, this is because multiple genes influence these responses. In addition, pharmacogenetic analysis was not the primary outcome of most of these studies and sample sizes were too small to assess single or multiple gene interactions. Furthermore, when the combination of a LABA and ICS are used, polymorphisms in the β 2 -adrenergic receptor pathway might result in a negative treatment response that interacts with polymorphisms in the same pathway or an alternative (i.e., glucocorticoid) pathway to obscure associations.


Pharmacogenetic studies have the potential to identify the subgroup of asthmatics who are more or less responsive to biologic therapies currently under development. Biologic therapies are more expensive and have the potential to cause unwanted adverse responses. Thus, pharmacogenetic therapeutic “biomarkers” that identify susceptible asthmatics should facilitate development and registration of biologic therapies. Pitrakinra is a molecular inhibitor of the IL4α receptor subunit that inhibits both IL-4 and IL-13, which are important in the regulation of Th2 allergic inflammation. In a phase 2b clinical trial, specific variants in the gene coding for the IL4α receptor subunit (IL4RA) have been associated with changes in antigen-induced bronchial hyperresponsiveness in atopic asthmatics treated with pitrakinra, and improvements in asthma exacerbation frequency. These results are an excellent example of the use of a pharmacogenetic “biomarker” to identify the subset (approximately 35 percent) of responders to this IL4α receptor antagonist.


Pharmacogenetic studies in asthma had initially been limited to candidate gene studies in smaller clinical trial cohorts; however, investigators have more recently performed GWAS which have identified novel pharmacogenetic loci (see eTable 45-3 ). In vitro studies have been used to validate the function of these SNPs. Future pharmacogenetic studies in independent, large clinical trial cohorts or genotype-stratified trials will be necessary to evaluate the importance of these gene variants, to confirm previous associations, and to determine whether these polymorphisms act independently or have additive effects on therapeutic responses to ICS, inhaled β-agonists, leukotriene modifiers, and other, newer asthma therapies. Understanding the pharmacologic and genetic basis of therapeutic responsiveness in asthma may lead to the development of more effective therapeutic agents. These approaches, when performed in adequately powered pharmacogenetic trials using unbiased methodology (GWAS), have delineated genetic determinants of pharmacologic treatment, with both beneficial and adverse responses, in other complex diseases such as those currently under investigation in the National Institutes of Health Pharmacogenomics Research Network. Thus, future precise therapeutic approaches in asthma will include the use of genetic pharmacogenetic profiles.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jul 21, 2019 | Posted by in CARDIOLOGY | Comments Off on Genetics in Asthma and COPD

Full access? Get Clinical Tree

Get Clinical Tree app for offline access