Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Nov 1.
Published in final edited form as: J Allergy Clin Immunol Pract. 2021 Sep 4;9(11):3887–3897. doi: 10.1016/j.jaip.2021.08.027

“Non-respiratory comorbidities in asthma”

Juan Carlos Cardet 1,*, Adeeb A Bulkhi 1,2,*, Richard F Lockey 1,3
PMCID: PMC8631133  NIHMSID: NIHMS1745370  PMID: 34492402

Abstract

Asthma is a chronic heterogeneous airway disease. Common comorbid conditions are often disproportionately present in severe asthma. Optimal care of patients with asthma requires the recognition and treatment of these comorbid conditions. This review outlines the pathophysiologic mechanisms between non-respiratory comorbid conditions and asthma and their effect on asthma outcomes. They include: type 2 diabetes mellitus, hypertension, atherosclerotic cardiovascular disease, adrenal and thyroid gland diseases, pregnancy, osteoporosis, adverse effects from medications, and mental health disorders. Studies indicate how poor glycemic control of type 2 diabetes mellitus is not only associated with greater healthcare utilization but poorer asthma outcomes. Also, a large healthcare claims database indicates that a substantial proportion of pregnant women have uncontrolled asthma and are prescribed suboptimal controller therapy. Additional data about these non-respiratory comorbidities and medications known to benefit both non-respiratory comorbidities and asthma are necessary.

Keywords: Diabetes mellitus, insulin resistance, hba1c, hypertension, atherosclerotic cardiovascular disease, adrenal disease, thyroid disease, pregnancy, osteoporosis, adverse effects from medications, mental health disorders, panic disorders, anxiety, depression, phenotype, endotype, airway obstructive disease, spirometry, pulmonary function tests, bronchoconstriction, airway hyperresponsiveness, airway smooth muscle, t helper cells, methacholine, randomized controlled trials, exacerbation prone asthma, glucocorticoids, beta agonists, beta blockers, ace inhibitors, sex hormones

Introduction:

Asthma is a common heterogeneous chronic airway disease constituted by multiple phenotypes and endotypes (1). Comorbid conditions are often disproportionately present in those with more severe disease (2). Optimal treatment of subjects with asthma requires the recognition and management of these comorbid conditions (3). This review focuses on shared pathophysiologic mechanisms between non-respiratory comorbid conditions and asthma, and the effect that these comorbidities have on clinical outcomes. The non-respiratory comorbid conditions addressed include type 2 diabetes mellitus, hypertension, atherosclerotic cardiovascular disease, adrenal and thyroid gland diseases, pregnancy, osteoporosis, adverse effects from medications, and mental health disorders (Table 1). There is also a bidirectional relationship, however, this article concentrates on the effects of these conditions on asthma outcomes and not the reverse.

Table 1.

Summary of non-respiratory comorbidities of asthma

Comorbidity Major findings References
Diabetes mellitus In vitro and in vivo:
 ○ Insulin heightens airway hyperresponsiveness, potentially through increased laminin and β-catenin expression.
Clinical studies in humans:
 ○ Higher Th1/Th2 ratios in peripheral blood lymphocytes among obese adolescents with asthma relative to non-obese adolescents with and without asthma
 ○ Higher IL6 levels are seen in subjects with asthma and those with the metabolic syndrome and may aggravate pathologic features of asthma.
 ○ Systemic inflammation from DM2 may worsen asthma
 ○ DM2 is associated with a greater incidence of asthma and worse asthma morbidity outcomes.
 ○ Dual anti-asthma and anti-diabetes medications are being investigated (e.g. metformin, GLP1RA).
8, 9, 12, 13, 16, 19, 21, 23
Hypertension In vitro and in vivo:
 ○ Hypertension is characterized by higher levels of IL17+ T cells, and IL17 induces AHR in mice.
Clinical studies in humans:
 ○ Hypertension is associated with greater rescue inhaler use and greater emergency room visits or hospitalizations for asthma
 ○ Dual anti-asthma and anti-hypertensive medications are being investigated (e.g. angiotensin receptor blockers)
11, 31, 37, 40
Atherosclerotic cardiovascular disease In vitro and in vivo:
 ○ IL1- and IL17-mediated inflammation and mast cells are implicated in atherosclerotic plaque formation and in subsets of asthma.
Clinical studies in humans:
 ○ Subjects with asthma compared to those without asthma are at higher risk of developing atherosclerotic cardiovascular complications.
 ○ Whether statins are beneficial in asthma remains inconclusive.
 ○ The anti-IL1β monoclonal antibody canakinumab improves cardiovascular outcomes but needs to be tested for efficacy in asthma
44, 46, 49, 52
Adrenal disorders In vitro and in vivo:
 ○ Glucocorticoids attenuate inflammation through genomic and non-genomic mechanisms.
Clinical studies in humans:
 ○ Case reports link Addison’s disease to asthma onset as well as deterioration of pre-existing asthma.
 ○ Subjects taking cytochrome P450 inhibitors are at greater risk of developing Cushing syndrome from use of ICS
56, 58, 62, 67, 68
Thyroid diseases In vitro and in vivo:
 ○ Thyroid hormones have an unclear role in asthma pathophysiology due to contrasting effects on airway structural and inflammatory cells.
Clinical studies in humans:
 ○ Subjects with autoimmune thyroiditis versus normals have greater odds of having asthma.
66, 71, 76
Pregnancy In vitro and in vivo:
 ○ Estrogen receptor-α signaling results in type-2 airway inflammation, mast cell activation, mucin hypersecretion.
 ○ Conversely, estrogen reduces airway hyperresponsiveness through airway smooth muscle relaxation.
 ○ Progesterone attenuates airway remodeling and glucocorticoid resistance.
Clinical studies in humans:
 ○ Pregnancy induces physiologic pulmonary function changes to accommodate for the increased maternal cardiometabolic demands.
 ○ A large proportion of pregnant women have uncontrolled asthma. Smoking and suboptimal pharmacotherapy are modifiable risk factors for uncontrolled asthma.
 ○ A greater susceptibility to viral respiratory infections in pregnant women with asthma relative to those without asthma may underlie their risk of uncontrolled asthma during pregnancy.
84, 87, 89, 90, 93, 98
Osteoporosis • Subjects with asthma at moderate to high risk of osteoporosis due to glucocorticoid use should have DEXA scan screens.
• Regular exercise, vitamin D and calcium supplementations, fall prevention strategies, smoking cessation, reductions in alcohol consumption are some of the cornerstones to maintaining adequate bone health.
102105
Medications • Beta blockers (especially non-cardio-selective ones) are not recommended in severe and uncontrolled asthma, but the literature supporting this recommendation is equivocal.
• Atopy and asthma may increase the risk of ACE inhibitor-induced cough, and the risk may be higher for severe asthma.
107, 110, 114, 118, 119
Mental health disorders In vitro and in vivo:
 ○ Serotonin has an unclear role in asthma pathophysiology due to contrasting effects on airway structural and inflammatory cells.
Clinical studies in humans:
 ○ Anxiety and panic disorders are more prevalent among subjects with asthma versus the general population.
 ○ Anxiety and depression are associated with poor asthma control and greater risk of asthma exacerbations.
124, 126, 128, 132, 133

ACE: angiotensin converting enzyme; AHR: airway hyperresponsiveness; DEXA: dualenergy x-ray absorptiometry; DM2: diabetes mellitus type 2; GLP1RA: glucagon-like peptide 1 receptor agonists ICS: inhaled corticosteroids; IL: interleukin; Th: T helper cell

Type 2 Diabetes

Contributory mechanisms:

Hyperinsulinemia characterizes type 2 diabetes mellitus (DM2) and may worsen asthma pathobiology. Experimentally, exogenous insulin administration potentiates vagally-induced bronchoconstriction in vivo in both obese and lean rats and ex vivo in airway smooth muscle (ASM) and tracheal rings derived from these animals when exposed to either methacholine or serotonin(4). This effect may be mediated by insulin-induced increased laminin expression, important glycoprotein components of extracellular matrices in ASM(5). Intranasal insulin administration in mice also increases airway hyperresponsiveness (AHR) and collagen deposition in the lung, the latter possibly due to PI3K/Akt signaling mediated by increased β-catenin expression(6). Insulin binds the insulin receptor but can also bind the insulin-like growth factor receptor 1 (IGFR1)(7), and IGFR1 knockout mice exhibit attenuated AHR and mucus secretion. Although there are no data that show that insulin binding of IGFR1 associates with AHR, this is possibly another signaling pathway linking DM2 with asthma(8).

Human data link DM2 and asthma through multiple mechanisms. Obese adolescents with asthma have higher T helper cell (Th)1/Th2 ratios relative to non-obese adolescents with and without asthma, and this skewing correlates with insulin resistance(9), which associates with lung function impairment. In turn, Th1-mediated inflammation is increased in subsets of severe asthma (7), suggesting that insulin resistance may contribute to asthma morbidity among obese subjects with non-Th2-mediated asthma. Non-Th1 and non-Th2 pathways are also at play in the DM2-asthma connection. Interleukin (IL)-6 is a pluripotent cytokine that induces mucus hypersecretion (10) and Th17 differentiation, both features frequently observed in the airways of subjects with severe asthma(11). IL6 is also overexpressed in obesity, insulin resistance and the metabolic syndrome(12). Peters et al of the National Heart, Lung, and Blood Institute Severe Asthma Research Program-3 (SARP3) network showed that serum IL6 elevations are associated with a persistent exacerbation-prone phenotype over 3 years of follow-up(13). Finally, DM2 may lead to the development or worsening of asthma through systemic low-grade inflammation(14).

Effects on clinical asthma outcomes:

DM2 is associated with a greater incidence of asthma and worse asthma morbidity outcomes. In a large prospective cohort, individuals with hyperglycemia or DM2 had 43% greater odds of incident asthma through a mean of 11 years of follow up(15). Obese individuals with insulin resistance, relative to those without, have a greater prevalence of current asthma symptoms(16). Insulin resistance associates with poorer lung function among adolescent subjects with and without asthma(17). Individuals with glycated hemoglobin (HbA1C) in the pre-diabetic or diabetic range versus those in the normal range have 27%−33% higher asthma exacerbation rates(18), worse lung function, and 68% greater odds of asthma hospitalization (19). As noted for IL6 levels, DM2 is overrepresented among subjects with severe asthma with a persistent exacerbation-prone phenotype over 3 years of follow-up(13). Zhang et al found that diabetic versus non-diabetic subjects with asthma have longer lengths of stay and greater costs and readmission risks in a large hospitalization healthcare database(20). Their longer length of stay may be due to a higher risk of non-respiratory complications, specifically, acute kidney injury, sepsis, and encephalopathy, potentially secondary to systemic glucocorticoid therapy, especially in the presence of DM2. These results suggest that DM2 and insulin resistance are detrimental to subjects with asthma.

Antidiabetic Agents and Asthma Outcomes:

There is considerable interest to develop interventions that can concomitantly control DM2 and asthma. Metformin is a biguanide that reduces insulin resistance and is first-line therapy for DM2. In vitro, metformin inhibits ASM proliferation through AMP-activated protein kinase activation(21) and may have anti-asthmatic therapeutic potential. Wu et al, in a retrospective analysis of a national administrative claims database, found that among individuals with asthma and diabetes, metformin use was associated with an 8% reduction in asthma exacerbations, specifically in those that incur greater healthcare utilization (ER visits, hospitalizations)(22).

Glucagon-like peptide-1 receptor agonists (GLP1RA) are indicated for the treatment of DM2. They also attenuate airway inflammation in murine animal models(23). Foer et al, in a retrospective analysis of electronic medical records of an academic medical center, found that subjects with DM2 and asthma who were prescribed GLP1RA had significantly lower asthma exacerbation rates versus those prescribed other anti-diabetic medications(24). However, epidemiological data must be interpreted with caution before attributing a causal therapeutic benefit to a medication. For example, thiazolidinediones were promising hypoglycemic asthma therapies(25) but did not demonstrate efficacy.(26). Randomized controlled trials (RCTs) are needed to determine whether metformin and GLP1RAs are efficacious to improve asthma outcomes.

Hypertension

Contributory mechanisms:

Several shared immune mechanisms underlie the pathogenesis of both hypertension and asthma(27), particularly for non-T2 asthma. Angiotensin II is an important vasopressor that leads to the secretion of IL17A (28). IL17A blocks vasodilation induced by endothelium-derived nitric oxide(29). Furthermore, IL17A knockout mice do not exhibit sustained hypertension(30) or related cardiovascular pathology(28). In turn, IL17A contributes to AHR in mice(31) and is elevated in bronchoalveolar airway lavage fluid of subjects with severe asthma(11). Hypertension is associated with Th1-skewing and increased interferon (IFN)-y production from rat splenic T cells(32). In addition, IFN-y is elevated in the bronchoalveolar lavage of subjects with severe asthma(33). Animal models implicate inflammasome activation and IL1 signaling in systemic hypertension(34), where its pharmacologic inhibition normalizes systemic blood pressure(35). In turn, inflammasome gene expression is elevated in the sputum from subjects with severe neutrophilic asthma versus controls without asthma (36). A large proportion of subjects with both asthma and hypertension have elevated markers of systemic inflammation such as C reactive protein(37).

Effects on clinical asthma outcomes:

Hypertension is associated with worse asthma morbidity and outcomes, although few studies document this association. Hypertension was associated with greater odds of rescue inhaler use, emergency room visits or hospitalizations, and corticosteroids bursts in a case-control study of 117,922 subjects with asthma in a large healthcare database (38). Hypertension, like DM2, was overrepresented in exacerbation-prone versus exacerbation-resistant asthma in a 3-year study (13). A large database analysis of subjects older than 60 years found that those with versus those without asthma had 48% greater odds of hypertension, but this study did not control for obesity which may confound this association(39). A cross-sectional analysis of a Korean nationally representative cohort found that the prevalence of asthma, only among women, is overrepresented among hypertensives(40). These clinical findings suggest that hypertension may be an important comorbidity within particular asthma populations; determining which requires further clarification.

As noted for DM2, it would be clinically valuable to identify interventions that can concomitantly control both hypertension and asthma. Angiotensin receptor blockers could be dual therapeutics for both asthma and hypertension considering their effects on vasculature, AHR and inflammation. One small clinical trial documented a modest improvement in responsiveness to methacholine bronchoprovocation among subjects with asthma administered an angiotensin receptor blocker. However, more clinically relevant outcomes have not been documented(41). A small crossover trial with nifedipine did not improve asthma symptoms or lung function despite the theoretical benefit of bronchodilation from a calcium-channel blocker(42). See section below on “medications and asthma” for a discussion on beta adrenergic blockers and ACE inhibitors.

Atherosclerotic cardiovascular disease

Contributory mechanisms:

Atherosclerotic cardiovascular disease (CVD), as hypertension, shares immunologic mechanistic underpinnings with asthma(43). A prominent role for the inflammasome/IL1 signaling was clinically demonstrated with the reduction in adverse CVD events among subjects administered canakinumab, an anti-IL1β monoclonal antibody(44), a mechanism as noted above, shared by some subjects with severe asthma.

Experimentally, mice genetically engineered for CVD susceptibility exhibit abundant Th17 infiltration in atherosclerotic plaques(45), reduced by anti-IL17 antibody administration(46). Mast cells are causally implicated in atherosclerotic plaques(47). They also orchestrate asthmatic airway inflammation and hyperresponsiveness. ASM proliferation is common to both AHR and atherosclerotic lesions (48).

Effects on clinical asthma outcomes:

Clinical data point to an association between CVD and asthma morbidity. A Dutch pharmacy database survey of 2,312 subjects with asthma indicates that CVD was disproportionately prevalent among difficult-to-control versus more treatable asthma subjects(49). A meta-analysis of 495,024 asthma subjects in 7 studies reported a 42% increased hazard of CVD for subjects with asthma versus those without asthma (50). A cross-sectional study of 173,209 Korean subjects showed greater odds of ischemic heart disease among subjects with asthma versus those without asthma, but only among those older than 53 years(51). Not all of these studies adjusted for obesity, which as noted above, may confound the relationship between CVD and asthma.

Little is known about interventions for a concomitant secondary prevention of CVD and asthma morbidity. Statin drugs are the mainstay preventive intervention for CVD, but a 2020 Cochrane review did not conclude that statin therapy was efficacious for asthma(52). However, a 2021 meta-analysis of 11 RCTs and 8 observational asthma studies suggest that statin drugs can improve asthma control and reduce exacerbations(53). As noted above, canakinumab improves CVD outcomes but has not been tested in asthma RCTs.

Adrenal glucocorticoids and asthma

Adrenal hormones have been known to have a positive effect on asthma since the early 1900s(54, 55). The adrenal zona fasciculata produces glucocorticoids (GC) regulated by the hypothalamic-pituitary-adrenal axis. GC inhibit most inflammatory cells through genomic and non-genomic mechanisms(56, 57). These include transcriptional effects via the GC receptor and GC-responsive elements, which induce the expression of potent anti-inflammatory mediators and transcription factors, (e.g., the inhibitor alpha of nuclear factor kappa-light-chain-enhancer of activated B cells (IκBα), interleukin (IL)-10, and annexin-1), and repress the expression of pro-inflammatory molecules, (e.g., nuclear factor kappa-light-chain-enhancer of activated B cells (NκFB) and activator protein-1). Non-genomic anti-inflammatory mechanisms of GCs include: mitogen activation protein kinase inhibition; mRNA destabilization; competition for co-activators; and disruption of DNA binding of transcription factors (56, 58). GC prevent the release of pro-bronchoconstrictive mediators from mast cells (59). They also enhance the expression and function of beta 2 adrenergic receptors in the airways and thus enhance bronchodilation (6062).

Relationship with clinical asthma outcomes

Adrenal suppression is a potential side effect of abrupt cessation of systemic GCs (63), a side effect occasionally observed with high dose inhaled corticosteroids (ICS)(64). A few cases link asthma onset to GC-naïve subjects following the onset of Addison’s disease (65). A deterioration of pre-existing asthma also has been reported following the diagnosis of Addison’s disease(66). Cushing syndrome is a known side effect of systemic corticosteroid therapy but occurs uncommonly with ICS due to lower plasma concentrations. However, subjects on cytochrome P450 inhibitors, such as ritonavir, are at greater risk of developing Cushing syndrome secondary to the greater plasma ICS concentrations reached due to reduced ICS metabolism (67, 68).

Thyroid disorders and asthma

Thyroid hormonal effects on asthma

Thyroid hormones play an unclear role in asthma pathobiology. In vitro triiodothyronine (T3) and thyroxine (T4) do not directly affect ASM cells, but T4>T3 act synergistically with transforming growth factor (TGF)-β1 to induce ASM proliferation. Conversely, thyroid hormones reduce TGF-β1-induced ASM contractility(69). T3 downregulates airway epithelial cell mucin gene and protein expression via retinoic acid receptor inhibition(70), while thyroid stimulating hormone upregulates submucosal gland hypertrophy and hyperplasia(71).

Experimental thyroidectomy in rat models of asthma reduces serum IgE levels and airway leukocytic infiltrates. Both are restored with thyroid hormone supplementation(72). These data suggest that the relationship between the thyroid hormonal axis and airway structural and inflammatory cells may be important, but its role in asthma pathobiology remains unclear.

Relationship with clinical asthma outcomes

Case reports link hyperthyroidism with asthma severity and exacerbation frequency(7375). Subjects with autoimmune thyroid disorders versus normals have a 3-fold higher odds of having asthma (76). Anti-thyroid peroxidase antibodies (anti-TPO) are frequently detected in subjects with autoimmune hyper- and hypo-thyroidism and are also more commonly noted in subjects with asthma versus healthy controls(77). These subjects frequently also express anti-cytokeratin 18 autoantibodies(78). This suggests that both thyroiditis and asthma in this subset may have a common autoimmune etiology.

Pregnancy

Sex hormonal effects on asthma:

Sex hormones exert profound effects on airway structural and inflammatory cells (79). These effects are pro-asthmatic in susceptible subjects, although predictors for this susceptibility remain unknown. The reversal in greater asthma prevalence by gender, boys more than girls during childhood, then girls more than boys after puberty, is hypothesized to be due to sex hormonal changes(80). Pregnancy also involves dramatic increases and fluctuations of several sex hormones(81). These hormonal changes are hypothesized to affect asthma based on in vitro and in vivo studies. However, sex hormone effects appear to be context-dependent, with both asthma-inducing and -protective effects depending on the cell type. Signaling through the estrogen receptor alpha subtype (ER-a) results in type-2 (T2) airway inflammation, which frequently characterizes asthma.

For example, experimentally, ER-a signaling results in IL4- and GATA-3 expression in CD4+ T helper (Th) cells(82) and alternative macrophage differentiation(83). ER-a signaling also leads to interleukin-(IL)-33/innate lymphoid cell (ILC)-2 mediated, allergen-induced eosinophilic airway inflammation in mice, an effect not observed in ER-a knockout mice(84). Likewise, estrogen activates mast cells(85), prime drivers of allergic inflammation implicated in asthma(86). Airway epithelial cells exposed to estrogen hypersecrete mucin(87). Conversely, estrogen reduced airway hyperresponsiveness (AHR) to methacholine in ovariectomized rats(88), an effect potentially mediated by ASM relaxation(89). Progesterone also seems to protect against asthma, attenuating airway remodeling and GC resistance in mice exposed to ozone(90).

Cellular functional studies that can recreate the hormonal conditions of pregnancy are difficult to design, so the net clinical effect of sex hormones on asthma has not been recapitulated by a mechanistic, reductionist in vitro approach.

Clinical asthma outcomes during pregnancy:

Human pregnancy induces physiologic pulmonary function changes to accommodate for the increased cardiometabolic demands on the mother(91). These include an increase in minute ventilation due to a rise in tidal volume and a reduction in functional residual capacity as pregnancy progresses. Many pregnant women perceive these respiratory changes as dyspnea(92), a perception usually heightened with the presence of asthma. Kircher et al reported a “rule of thirds” effect, with improvement, deterioration, and no change each occurring in 1/3 of subjects in a secondary analysis of prospective data collected from 671 pregnant women with asthma in the Kaiser-Permanente network (93). The only significantly associated variable to improvement in asthma symptoms during pregnancy was the concomitant improvement in rhinitis symptoms. A cross-sectional analysis of two large US healthcare claims databases (n=604,420 and n=2,071,359) published in 2019 identified that a substantial proportion of pregnant women with asthma have uncontrolled symptoms during pregnancy (16.5% and 28.0%, respectively) (94). The authors identified that many of these women with uncontrolled asthma were not prescribed long-acting beta agonists (38.4% and 43.3%, respectively), suggesting that the management of asthma during pregnancy can be optimized at the healthcare level. An analysis from a large longitudinal cohort of pregnant women with mild persistent asthma versus non-asthma women who underwent serial spirometric monitoring was published in 2021(95). It showed that while both pregnant women with and without asthma experience a significant decrease in FVC throughout gestation, only women without asthma experience a significant decrease in FEV1. This finding may be due to the high proportion of women with asthma (69%) that were prescribed an ICS + long-acting beta agonist combination therapy and to frequent medical visits—features that may be unique to this cohort and perhaps not representative of the general population. Asthma control did not fluctuate substantially from baseline and paralleled the stability of FEV1 regardless of the initial level of asthma control. These findings contrast those of prior reports which showcase loss of asthma control and a worsening FEV1 in pregnant women with asthma(96) and emphasizes the need for optimal pharmacotherapeutic management of asthma during pregnancy. Clinicians should recognize that the risk of uncontrolled asthma is more concerning to both mother and fetus than the risk of using inhaled medications for asthma.

To this effect, a prospective study of n=85 inner-city pregnant women with asthma was published in 2021 and showed that integrating a subspecialty asthma clinic visit to routine prenatal care significantly improved asthma control (97). The improvement in asthma control was likely related to optimization of these subjects’ controller therapy regimen as ~80% of them were routinely using rescue therapy alone.

Grzeskowiak et al prospectively followed 189 pregnant women with asthma and determined that smoking, older maternal age, and ICS use were predictors of uncontrolled asthma, but not asthma exacerbations(98). However, the association between ICS and uncontrolled asthma may have been confounded by severity. In a prospective study of 285 subjects, pregnant women with asthma versus those without asthma were more likely to experience respiratory viral infections and more likely to develop uncontrolled asthma, highlighting the possibility that pregnancy leads to loss of asthma control through greater susceptibility to viral respiratory tract infections(99). Asthma control returns to pre-pregnancy levels post-partum in 73% of women(100). Anecdotal evidence also suggests a subgroup of women develop asthma post-partum.

Osteoporosis

Contributory mechanisms:

The deficient bone mass characteristic of osteoporosis is due to excessive osteolysis and inadequate osteogenesis. Osteoporosis predisposes to fractures and osteoporosis-induced vertebral compression fractures lead to kyphosis, which can mechanically impair lung function. The systemic GC frequently prescribed to subjects with asthma increase osteocyte and osteoblast apoptosis, inhibit collagen formation, increase osteoclast activity, and decrease osteoclast apoptosis(101), increasing the risk of osteoporosis. GC also causes feedback inhibition of the anterior pituitary gonadotropic hormones, which are osteogenic. They stimulate collagen formation and decrease osteoblast apoptosis. Finally, GC reduces calcium reabsorption from the kidneys and absorption from the intestine, which leads to decreased circulatory calcium levels and thus increased parathyroid hormone production(102). Elevated parathyroid hormone levels lead to further bone demineralization through bone calcium release(103).

Relationship with clinical asthma outcomes:

Guidelines suggest risk stratifying subjects to low, moderate, or high risk of osteoporosis based on formulation and duration of GC use (104106) and on other factors, e.g., gender, smoking history, obesity, and other inflammatory or endocrinopathy conditions(102). Subjects with moderate to high risk need dual-energy x-ray absorptiometry (DEXA) bone scans to assess bone density(107). Table 2 summarizes osteoporosis screening recommendations.

Table 2.

Recommendations for initial fracture assessment after starting GC

Children younger than 18 years Adults between 18 and 40 years of age Adults older than 40 years
Clinical risk assessment for fractures within 6 months after starting GC
No further initial assessment BMD testing needed for those with a history of osteoporosis- related fracture OR other significant risk factors Fracture risk assessment using FRAX* and BMD testing within 6 months
Clinical fracture risk assessment every 12 months
Clinical fracture risk evaluation every 12 months only Any of the following requires BMD testing every 2–3 years regardless of osteoporosis treatment status:
  1. History of osteoporosis fracture.
  2. Z score < −3 at hip or spine.
  3. >10%/year loss of BMD at hip or spine.
  4. Very high dose GCs
  5. Other osteoporosis risk factors
For subjects without prior treatment for osteoporosis: --BMD testing every 1–3 years For subjects being treated for osteoporosis: --BMD testing every 2–3 years only if any of the following are present:
1. History of osteoporosis fracture >= 18 months after treatment initiation
2. Poor medication adherence
3. Very high dose GCs
4. Other osteoporosis risk factors --Otherwise, no testing is indicated
For subjects who have completed treatment for osteoporosis: --BMD testing every 2–3 years

High dose GC equivalent to prednisone ≥30 mg/day and a cumulative dose of >5 g in the past year;

*

FRAX (https://www.shef.ac.uk/FRAX/tool.jsp) is a fracture risk assessment tool than incorporates GC dose and risk factors. BMD: bone mineral density; GC: glucocorticosteroids . Adapted from Buckley et al. (104).

Regular exercise, vitamin D and calcium supplementations, fall prevention strategies, smoking cessation, and reductions in alcohol consumption are some of the cornerstones to maintain bone health.(102). Bisphosphonates and newer monoclonal agents like denosumab should be considered as prophylaxis, especially during prolonged GC use and to treat osteoporosis (102).

Medications and asthma

Beta adrenergic blockers

Beta adrenergic blockers (BB) are indicated for the treatment of arrhythmias, congestive heart failure, hypertension, and migraines, among other diseases. They are not recommended for use with severe and uncontrolled asthma. This recommendation originated from the observation that bronchoconstriction occurs due to BB use, particularly non-cardio-selective BBs, among healthy volunteers and subjects with asthma administered propranolol (110, 111) (108). Current literature on BB that does not account for their cardioselectivity is still mixed on whether they worsen asthma (108, 109).

BB putatively cause bronchoconstriction by antagonizing the pro-bronchodilating β2 adrenergic receptor, resulting in unopposed pro-bronchoconstrictive cholinergic signaling (108). But a meta-analysis found that subjects with asthma tolerate cardio-selective BB, especially low doses(112). Non-selective BB impose a higher risk of bronchoconstriction for which a gradual up-titrated dose and initial concomitant use of a long-acting muscarinic antagonist to prevent unopposed cholinergic tone is recommended. Similarly, a meta-analysis found that exposure to topical non-cardio-selective BB eye drops was associated with decreases in FEV1 (113).

BB also are theorized to be of benefit for asthma through an up-regulation of β2 adrenergic receptor density on ASM. This theory is based on the experimental observation that the extended administration of BB in murine models of asthma reduces AHR and airway inflammation(114).

Subjects with asthma exhibited reduced AHR in a small open-label pilot study of subjects chronically exposed to BB(115), however, this effect was not confirmed in more rigorously controlled trials (116). Among subjects with mild to moderate asthma chronically administered ICS and long-acting muscarinic antagonists, those on a BB exhibited similar AHR to those on a placebo(117, 118).

Angiotensin converting enzyme (ACE) inhibitors

ACE inhibitors are indicated for the treatment of congestive heart failure, hypertension, and chronic kidney disease. These drugs inhibit the renin angiotensin aldosterone system (RAAS) which regulates systemic vascular tone and blood pressure. ACE inhibitor-induced cough occurs in 4–35% of subjects, which can mimic cough-variant asthma. The presence of atopy and asthma may increase this risk(119), and the risk may be higher for more severe asthma(120). ACE inhibitors may worsen asthma control but not induce asthma de novo (121).

Mental health disorders

Contributory mechanisms:

Dysregulation of the serotonin system is essential for neural processes implicated in anxiety, depression, phobia, and suicidality(122) and also modulates immune responses. However, the data on the relationship between serotonin and asthma are mixed. Experimentally, alveolar macrophages (AM) exposed in vitro to serotonin secrete more type 2 inflammatory cytokines, which are commonly implicated in asthma pathobiology(123, 124) (125). Conversely, 5HT decreases the expression of T cell-derived type 2 inflammatory mediators, including monocyte chemotactic protein-1, IL-5 and IL-13, and granulocyte-macrophage colony-stimulating factor. This suggests that this effect may depend on cell types(126). 5HT induces ASM contraction. Effects on AHR and inflammation in animal models of asthma are mixed(127).

Effects on clinical asthma outcomes

Anxiety, panic disorder and agoraphobia are more prevalent in subjects with asthma versus the general population(128) (129) and are associated with higher exacerbation rates(130). The prevalence of anxiety disorder is 19% and depression disorder 7% in the general population in the USA,(131) versus 34% and 5–10% among subjects with asthma, respectively(132, 133). Many subjects have difficulty distinguishing between anxiety and asthma as the cause of their dyspnea, which complicates the management of asthma through misdirected therapy (134). Anxiety also associates with dysfunctional breathing, an altered breathing pattern that results in dyspnea at rest often despite normal lung function (135). Further, the hyperventilation experienced by subjects with anxiety and other panic disorders can cause increased inhalation of cold air, which is bronchoconstrictive(125). Poor asthma control test correlates with higher stress, anxiety and depression scores(136). Subjects with comorbid asthma and depression versus those without depression have a 2-fold increased risk of asthma-related ER visits (137). The relationship between asthma and depression is bidirectional, as subjects with asthma versus those without are also at 1.8-fold higher odds of developing depression (138). Subjects with asthma and co-morbid anxiety exhibit worse quality-of-life in domains both related and unrelated to asthma(139, 140). In randomized trial comparing the effects of the anti-depressant escitalopram to placebo among subjects with asthma and major depression, a subset of the intervention group experienced significant improvements in asthma symptoms and reductions in oral corticosteroid usage compared to placebo (141). This finding suggests that appropriate treatment of comorbid mental health disorders is important for optimal asthma care.

Conclusion:

Non-respiratory comorbidities frequently associate with worse asthma control and greater risks of exacerbation and thus pose challenges for its optimal management. A multidisciplinary approach that addresses these comorbidities may result in better clinical asthma outcomes. However, additional prospective data about these non-respiratory comorbidities are needed. For example, although uncontrolled hyperglycemia associates with worse clinical asthma outcomes, there is no prospective data to show that improved glycemic control also improves clinical asthma outcomes. Clinicians are therefore advised to use the best available evidence to optimize the management of both asthma and non-respiratory comorbidities. Likewise, trials that study the effect on clinical asthma outcomes of medications known to benefit both non-respiratory comorbidities and asthma are equally important and needed.

Acknowledgments

Funding:

This work was conducted with the support of grant K23AI125785 from NIAID and the Allergic Respiratory Diseases Research Award AI-835475 from ALA/AAAAI to JCC; by generous contributions by the Culverhouse family fund in Tampa to JCC and RFL, and by the James A. Haley Veterans’ Affairs Hospital to RFL.

Abbreviations:

5-HT2A

5-hydroxytryptamine 2A

ACTH

adrenocorticotropic hormone

AHR

airway hyperresponsiveness

AM

alveolar macrophage

Anti-TPO

anti–thyroid peroxidase antibodies

ASM

airway smooth muscle

BB

beta adrenergic blockers

CRH

corticotrophin releasing hormone

CVD

atherosclerotic cardiovascular disease

DM2

type 2 diabetes mellitus

DEXA

Dual-energy X-ray Absorptiometry

ER-a

estrogen receptor alpha subtype

GC

glucocorticoids

GM-CSF

granulocyte macrophage colony-stimulating factor

GLP1RA

glucagon-like peptide-1 receptor agonists

HbA1C

glycated hemoglobin A1C

ICS

inhaled corticosteroid

IFN

interferon

IGFR1

insulin-like growth factor receptor 1

ILC

innate lymphoid cell

IκBα

inhibitor alpha of nuclear factor kappa-light-chain-enhancer of activated B cells

IL

interleukin

MCP-1

monocyte chemotactic protein-1

NκFB

nuclear factor kappa-light-chain-enhancer of activated B cells

PGE2

prostaglandin-E2

RCT

randomized controlled trials

SARP3

Severe Asthma Research Program-3

T2

type 2 inflammation

T3

triiodothyronine

T4

thyroxine (T4)

Th

T helper cell

TGF

transforming growth factor

TNF

tumor necrosis factor

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

COI:

JCC reports receiving honoraria from Astra Zeneca and Genentech for work in advisory committees. AAB reports receiving honoraria from GSK and Sanofi for work in advisory committees. RFL does not have any conflicts of interest to report.

References:

  • 1.Kaur R, Chupp G. Phenotypes and endotypes of adult asthma: Moving toward precision medicine. J Allergy Clin Immunol 2019;144(1):1–12. [DOI] [PubMed] [Google Scholar]
  • 2.Denlinger LC, Phillips BR, Ramratnam S, Ross K, Bhakta NR, Cardet JC, et al. Inflammatory and Comorbid Features of Patients with Severe Asthma and Frequent Exacerbations. Am J Respir Crit Care Med 2017;195(3):302–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Federico MJ, Denlinger LC, Corren J, Szefler SJ, Fuhlbrigge AL. Exacerbation-Prone Asthma: A Biologic Phenotype or a Social Construct. The Journal of Allergy and Clinical Immunology: In Practice 2021. [DOI] [PubMed] [Google Scholar]
  • 4.Proskocil BJ, Calco GN, Nie Z. Insulin acutely increases agonist-induced airway smooth muscle contraction in human and rat. Am J Physiol Lung Cell Mol Physiol 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Dekkers BG, Schaafsma D, Tran T, Zaagsma J, Meurs H. Insulin-induced laminin expression promotes a hypercontractile airway smooth muscle phenotype. Am J Respir Cell Mol Biol 2009;41(4):494–504. [DOI] [PubMed] [Google Scholar]
  • 6.Singh S, Bodas M, Bhatraju NK, Pattnaik B, Gheware A, Parameswaran PK, et al. Hyperinsulinemia adversely affects lung structure and function. Am J Physiol Lung Cell Mol Physiol 2016;310(9):L837–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Boucher J, Tseng YH, Kahn CR. Insulin and insulin-like growth factor-1 receptors act as ligand-specific amplitude modulators of a common pathway regulating gene transcription. J Biol Chem 2010;285(22):17235–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Pineiro-Hermida S, Gregory JA, Lopez IP, Torrens R, Ruiz-Martinez C, Adner M, et al. Attenuated airway hyperresponsiveness and mucus secretion in HDM-exposed Igf1r-deficient mice. Allergy 2017;72(9):1317–26. [DOI] [PubMed] [Google Scholar]
  • 9.Rastogi D, Fraser S, Oh J, Huber AM, Schulman Y, Bhagtani RH, et al. Inflammation, metabolic dysregulation, and pulmonary function among obese urban adolescents with asthma. Am J Respir Crit Care Med 2015;191(2):149–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Neveu WA, Allard JB, Dienz O, Wargo MJ, Ciliberto G, Whittaker LA, et al. IL-6 is required for airway mucus production induced by inhaled fungal allergens. J Immunol 2009;183(3):1732–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Irvin C, Zafar I, Good J, Rollins D, Christianson C, Gorska MM, et al. Increased frequency of dual-positive TH2/TH17 cells in bronchoalveolar lavage fluid characterizes a population of patients with severe asthma. J Allergy Clin Immunol 2014;134(5):1175–86 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Lumeng CN, Saltiel AR. Inflammatory links between obesity and metabolic disease. J Clin Invest 2011;121(6):2111–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Peters MC, Mauger D, Ross KR, Phillips B, Gaston B, Cardet JC, et al. Evidence for Exacerbation-Prone Asthma and Predictive Biomarkers of Exacerbation Frequency. Am J Respir Crit Care Med 2020;202(7):973–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kankaanranta H, Kauppi P, Tuomisto LE, Ilmarinen P. Emerging Comorbidities in Adult Asthma: Risks, Clinical Associations, and Mechanisms. Mediators Inflamm 2016;2016:3690628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Brumpton BM, Camargo CA Jr., Romundstad PR, Langhammer A, Chen Y, Mai XM. Metabolic syndrome and incidence of asthma in adults: the HUNT study. Eur Respir J 2013;42(6):1495–502. [DOI] [PubMed] [Google Scholar]
  • 16.Cardet JC, Ash S, Kusa T, Camargo CA Jr., Israel E. Insulin resistance modifies the association between obesity and current asthma in adults. Eur Respir J 2016;48(2):403–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Forno E, Han YY, Muzumdar RH, Celedon JC. Insulin resistance, metabolic syndrome, and lung function in US adolescents with and without asthma. J Allergy Clin Immunol 2015;136(2):304–11 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wu TD, Brigham EP, Keet CA, Brown TT, Hansel NN, McCormack MC. Association Between Prediabetes/Diabetes and Asthma Exacerbations in a Claims-Based Obese Asthma Cohort. J Allergy Clin Immunol Pract 2019;7(6):1868–73 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yang G, Han YY, Forno E, Yan Q, Rosser F, Chen W, et al. Glycated Hemoglobin A1c, Lung Function, and Hospitalizations Among Adults with Asthma. J Allergy Clin Immunol Pract 2020;8(10):3409–15 e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Zhang P, Lopez R, Attaway AH, Georas SN, Khatri SB, Abi-Saleh S, et al. Diabetes Mellitus Is Associated with Worse Outcome in Patients Hospitalized for Asthma. J Allergy Clin Immunol Pract 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pan Y, Liu L, Zhang Q, Shi W, Feng W, Wang J, et al. Activation of AMPK suppresses S1P-induced airway smooth muscle cells proliferation and its potential mechanisms. Mol Immunol 2020;128:106–15. [DOI] [PubMed] [Google Scholar]
  • 22.Wu TD, Keet CA, Fawzy A, Segal JB, Brigham EP, McCormack MC. Association of Metformin Initiation and Risk of Asthma Exacerbation. A Claims-based Cohort Study. Ann Am Thorac Soc 2019;16(12):1527–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Toki S, Goleniewska K, Reiss S, Zhang J, Bloodworth MH, Stier MT, et al. Glucagon-like peptide 1 signaling inhibits allergen-induced lung IL-33 release and reduces group 2 innate lymphoid cell cytokine production in vivo. J Allergy Clin Immunol 2018;142(5):1515–28 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Foer D, Beeler PE, Cui J, Karlson EW, Bates DW, Cahill KN. Asthma Exacerbations in Type 2 Diabetics with Asthma on Glucagon-like Peptide-1 Receptor Agonists. Am J Respir Crit Care Med 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Rinne ST, Feemster LC, Collins BF, Au DH, Perkins M, Bryson CL, et al. Thiazolidinediones and the risk of asthma exacerbation among patients with diabetes: a cohort study. Allergy Asthma Clin Immunol 2014;10(1):34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kaler M, Barochia AV, Weir NA, Cuento RA, Stylianou M, Roth MJ, et al. A randomized, placebo-controlled, double-blinded, crossover trial of pioglitazone for severe asthma. J Allergy Clin Immunol 2017;140(6):1716–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wenzel U, Turner JE, Krebs C, Kurts C, Harrison DG, Ehmke H. Immune Mechanisms in Arterial Hypertension. J Am Soc Nephrol 2016;27(3):677–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Li Y, Wu Y, Zhang C, Li P, Cui W, Hao J, et al. gammadeltaT Cell-derived interleukin-17A via an interleukin-1beta-dependent mechanism mediates cardiac injury and fibrosis in hypertension. Hypertension 2014;64(2):305–14. [DOI] [PubMed] [Google Scholar]
  • 29.Nguyen H, Chiasson VL, Chatterjee P, Kopriva SE, Young KJ, Mitchell BM. Interleukin-17 causes Rho-kinase-mediated endothelial dysfunction and hypertension. Cardiovasc Res 2013;97(4):696–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Madhur MS, Lob HE, McCann LA, Iwakura Y, Blinder Y, Guzik TJ, et al. Interleukin 17 promotes angiotensin II-induced hypertension and vascular dysfunction. Hypertension 2010;55(2):500–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Chesne J, Braza F, Chadeuf G, Mahay G, Cheminant MA, Loy J, et al. Prime role of IL-17A in neutrophilia and airway smooth muscle contraction in a house dust mite-induced allergic asthma model. J Allergy Clin Immunol 2015;135(6):1643–e3. [DOI] [PubMed] [Google Scholar]
  • 32.Shao J, Nangaku M, Miyata T, Inagi R, Yamada K, Kurokawa K, et al. Imbalance of T-cell subsets in angiotensin II-infused hypertensive rats with kidney injury. Hypertension 2003;42(1):31–8. [DOI] [PubMed] [Google Scholar]
  • 33.Raundhal M, Morse C, Khare A, Oriss TB, Milosevic J, Trudeau J, et al. High IFN-gamma and low SLPI mark severe asthma in mice and humans. J Clin Invest 2015;125(8):3037–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sun HJ, Ren XS, Xiong XQ, Chen YZ, Zhao MX, Wang JJ, et al. NLRP3 inflammasome activation contributes to VSMC phenotypic transformation and proliferation in hypertension. Cell Death Dis 2017;8(10):e3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Krishnan SM, Ling YH, Huuskes BM, Ferens DM, Saini N, Chan CT, et al. Pharmacological inhibition of the NLRP3 inflammasome reduces blood pressure, renal damage, and dysfunction in salt-sensitive hypertension. Cardiovasc Res 2019;115(4):776–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rossios C, Pavlidis S, Hoda U, Kuo CH, Wiegman C, Russell K, et al. Sputum transcriptomics reveal upregulation of IL-1 receptor family members in patients with severe asthma. J Allergy Clin Immunol 2018;141(2):560–70. [DOI] [PubMed] [Google Scholar]
  • 37.Christiansen SC, Zuraw BL. Treatment of Hypertension in Patients with Asthma. N Engl J Med 2019;381(11):1046–57. [DOI] [PubMed] [Google Scholar]
  • 38.Christiansen SC, Schatz M, Yang SJ, Ngor E, Chen W, Zuraw BL. Hypertension and Asthma: A Comorbid Relationship. J Allergy Clin Immunol Pract 2016;4(1):76–81. [DOI] [PubMed] [Google Scholar]
  • 39.Bozek A, Rogala B, Bednarski P. Asthma, COPD and comorbidities in elderly people. J Asthma 2016;53(9):943–7. [DOI] [PubMed] [Google Scholar]
  • 40.Lee HS, Park YM, Han K, Pekler G, Lee SS, Yoo S, et al. Sex-specific association between asthma and hypertension in nationally representative young Korean adults. Sci Rep 2017;7(1):15667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Myou S, Fujimura M, Kamio Y, Kita T, Watanabe K, Ishiura Y, et al. Effect of candesartan, a type 1 angiotensin II receptor antagonist, on bronchial hyper-responsiveness to methacholine in patients with bronchial asthma. Br J Clin Pharmacol 2002;54(6):622–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Patakas D, Maniki E, Tsara V, Dascalopoulou E. Nifedipine treatment of patients with bronchial asthma. J Allergy Clin Immunol 1987;79(6):959–63. [DOI] [PubMed] [Google Scholar]
  • 43.Gurgone D, McShane L, McSharry C, Guzik TJ, Maffia P. Cytokines at the Interplay Between Asthma and Atherosclerosis? Front Pharmacol 2020;11:166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med 2017;377(12):1119–31. [DOI] [PubMed] [Google Scholar]
  • 45.Gao Q, Jiang Y, Ma T, Zhu F, Gao F, Zhang P, et al. A critical function of Th17 proinflammatory cells in the development of atherosclerotic plaque in mice. J Immunol 2010;185(10):5820–7. [DOI] [PubMed] [Google Scholar]
  • 46.Wang L, Gao S, Xu W, Zhao S, Zhou J, Wang N, et al. Allergic asthma accelerates atherosclerosis dependent on Th2 and Th17 in apolipoprotein E deficient mice. J Mol Cell Cardiol 2014;72:20–7. [DOI] [PubMed] [Google Scholar]
  • 47.Kovanen PT, Bot I. Mast cells in atherosclerotic cardiovascular disease - Activators and actions. Eur J Pharmacol 2017;816:37–46. [DOI] [PubMed] [Google Scholar]
  • 48.Liu CL, Zhang JY, Shi GP. Interaction between allergic asthma and atherosclerosis. Transl Res 2016;174:5–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hekking PP, Amelink M, Wener RR, Bouvy ML, Bel EH. Comorbidities in Difficult-to-Control Asthma. J Allergy Clin Immunol Pract 2018;6(1):108–13. [DOI] [PubMed] [Google Scholar]
  • 50.Liu H, Fu Y, Wang K. Asthma and risk of coronary heart disease: A meta-analysis of cohort studies. Ann Allergy Asthma Immunol 2017;118(6):689–95. [DOI] [PubMed] [Google Scholar]
  • 51.Wee JH, Park MW, Min C, Byun SH, Park B, Choi HG. Association between asthma and cardiovascular disease. Eur J Clin Invest 2021;51(3):e13396. [DOI] [PubMed] [Google Scholar]
  • 52.Naing C, Ni H. Statins for asthma. Cochrane Database Syst Rev 2020;7:CD013268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sunata K, Kabata H, Kuno T, Takagi H, So M, Masaki K, et al. The effect of statins for asthma. A systematic review and meta-analysis. J Asthma 2021:1–10. [DOI] [PubMed] [Google Scholar]
  • 54.Schleimer RP. Interactions between the hypothalamic-pituitary-adrenal axis and allergic inflammation. J Allergy Clin Immunol 2000;106(5 Suppl):S270–4. [DOI] [PubMed] [Google Scholar]
  • 55.Arthur G. Epinephrine: a short history. Lancet Respir Med 2015;3(5):350–1. [DOI] [PubMed] [Google Scholar]
  • 56.Bellavance MA, Rivest S. The HPA - Immune Axis and the Immunomodulatory Actions of Glucocorticoids in the Brain. Front Immunol 2014;5:136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Xing Y, Edwards MA, Ahlem C, Kennedy M, Cohen A, Gomez-Sanchez CE, et al. The effects of ACTH on steroid metabolomic profiles in human adrenal cells. Journal of Endocrinology 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Rhen T, Cidlowski JA. Antiinflammatory action of glucocorticoids--new mechanisms for old drugs. N Engl J Med 2005;353(16):1711–23. [DOI] [PubMed] [Google Scholar]
  • 59.Peachell P. Regulation of mast cells by beta-agonists. Clin Rev Allergy Immunol 2006;31(2–3):131–42. [DOI] [PubMed] [Google Scholar]
  • 60.Baraniuk JN, Ali M, Brody D, Maniscalco J, Gaumond E, Fitzgerald T, et al. Glucocorticoids induce beta2-adrenergic receptor function in human nasal mucosa. Am J Respir Crit Care Med 1997;155(2):704–10. [DOI] [PubMed] [Google Scholar]
  • 61.Mak JC, Nishikawa M, Barnes PJ. Glucocorticosteroids increase beta 2-adrenergic receptor transcription in human lung. Am J Physiol 1995;268(1 Pt 1):L41–6. [DOI] [PubMed] [Google Scholar]
  • 62.Mak JC, Chuang TT, Harris CA, Barnes PJ. Increased expression of G protein-coupled receptor kinases in cystic fibrosis lung. Eur J Pharmacol 2002;436(3):165–72. [DOI] [PubMed] [Google Scholar]
  • 63.Broersen LH, Pereira AM, Jorgensen JO, Dekkers OM. Adrenal Insufficiency in Corticosteroids Use: Systematic Review and Meta-Analysis. J Clin Endocrinol Metab 2015;100(6):2171–80. [DOI] [PubMed] [Google Scholar]
  • 64.Choi IS, Sim DW, Kim SH, Wui JW. Adrenal insufficiency associated with long-term use of inhaled steroid in asthma. Ann Allergy Asthma Immunol 2017;118(1):66–72 e1. [DOI] [PubMed] [Google Scholar]
  • 65.Harris PW, Collins JV. Bronchial asthma with Addison’s disease. Lancet 1971;1(7713):1349–50. [DOI] [PubMed] [Google Scholar]
  • 66.Saraclar Y, Turktas I, Adalioglu G, Tuncer A. Bronchial asthma with Addison’s disease. Respiration 1993;60(4):241–2. [DOI] [PubMed] [Google Scholar]
  • 67.Arrington-Sanders R, Hutton N, Siberry GK. Ritonavir-fluticasone interaction causing Cushing syndrome in HIV-infected children and adolescents. Pediatr Infect Dis J 2006;25(11):1044–8. [DOI] [PubMed] [Google Scholar]
  • 68.Figueiredo J, Serrado M, Khmelinskii N, do Vale S. Iatrogenic Cushing syndrome and multifocal osteonecrosis caused by the interaction between inhaled fluticasone and ritonavir. BMJ Case Rep 2020;13(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Dekkers BG, Naeimi S, Bos IS, Menzen MH, Halayko AJ, Hashjin GS, et al. L-thyroxine promotes a proliferative airway smooth muscle phenotype in the presence of TGF-beta1. Am J Physiol Lung Cell Mol Physiol 2015;308(3):L301–6. [DOI] [PubMed] [Google Scholar]
  • 70.Gray T, Nettesheim P, Basbaum C, Koo J. Regulation of mucin gene expression in human tracheobronchial epithelial cells by thyroid hormone. Biochem J 2001;353(Pt 3):727–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tamada T, Sasaki T, Saitoh H, Ohkawara Y, Irokawa T, Sasamori K, et al. A novel function of thyrotropin as a potentiator of electrolyte secretion from the tracheal gland. Am J Respir Cell Mol Biol 2000;22(5):566–73. [DOI] [PubMed] [Google Scholar]
  • 72.Manzolli S, Macedo-Soares MF, Vianna EO, Sannomiya P. Allergic airway inflammation in hypothyroid rats. J Allergy Clin Immunol 1999;104(3 Pt 1):595–600. [DOI] [PubMed] [Google Scholar]
  • 73.Hoffman DA, McConahey WM. THYROTOXICOSIS AND ASTHMA. The Lancet 1982. [DOI] [PubMed] [Google Scholar]
  • 74.Luong KVQ, Nguyen LTH. Hyperthyroidism and asthma. Journal of Asthma 2000. [DOI] [PubMed] [Google Scholar]
  • 75.Ayres J, Clark TJ. Asthma and the thyroid. Lancet 1981;2(8255):1110–1. [DOI] [PubMed] [Google Scholar]
  • 76.Fekri MS, Shokoohi M, Gozashti MH, Esmailian S, Jamshidian N, Shadkam-Farokhi M, et al. Association between anti-thyroid peroxidase antibody and asthma in women. Iranian Journal of Allergy, Asthma and Immunology 2012. [PubMed] [Google Scholar]
  • 77.El-Aziz MHA, Rafaat MM, Sabry I, Yousef M, Mandour A, editors. Study of Thyroid AutoAntibodies in Patients with Bronchial Asthma and Allergic Rhinitis 2010. [Google Scholar]
  • 78.Mohammad HA, Abdelfattah MT, Ali LH, Morsi ZI. Association between anti-thyroid peroxidase and anti-cytokeratin 18 autoantibodies and bronchial asthma in women. Egyptian Journal of Chest Diseases and Tuberculosis 2016. [Google Scholar]
  • 79.Townsend EA, Miller VM, Prakash YS. Sex differences and sex steroids in lung health and disease. Endocr Rev 2012;33(1):1–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Dharmage SC, Perret JL, Custovic A. Epidemiology of Asthma in Children and Adults. Front Pediatr 2019;7:246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Schock H, Zeleniuch-Jacquotte A, Lundin E, Grankvist K, Lakso HA, Idahl A, et al. Hormone concentrations throughout uncomplicated pregnancies: a longitudinal study. BMC Pregnancy Childbirth 2016;16(1):146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lambert KC, Curran EM, Judy BM, Milligan GN, Lubahn DB, Estes DM. Estrogen receptor alpha (ERalpha) deficiency in macrophages results in increased stimulation of CD4+ T cells while 17beta-estradiol acts through ERalpha to increase IL-4 and GATA-3 expression in CD4+ T cells independent of antigen presentation. J Immunol 2005;175(9):5716–23. [DOI] [PubMed] [Google Scholar]
  • 83.Campbell L, Emmerson E, Williams H, Saville CR, Krust A, Chambon P, et al. Estrogen receptor-alpha promotes alternative macrophage activation during cutaneous repair. J Invest Dermatol 2014;134(9):2447–57. [DOI] [PubMed] [Google Scholar]
  • 84.Cephus JY, Gandhi VD, Shah R, Brooke Davis J, Fuseini H, Yung JA, et al. Estrogen receptor-alpha signaling increases allergen-induced IL-33 release and airway inflammation. Allergy 2021;76(1):255–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Zaitsu M, Narita S, Lambert KC, Grady JJ, Estes DM, Curran EM, et al. Estradiol activates mast cells via a non-genomic estrogen receptor-alpha and calcium influx. Mol Immunol 2007;44(8):1977–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Cahill KN, Katz HR, Cui J, Lai J, Kazani S, Crosby-Thompson A, et al. KIT Inhibition by Imatinib in Patients with Severe Refractory Asthma. N Engl J Med 2017;376(20):1911–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Tam A, Wadsworth S, Dorscheid D, Man SF, Sin DD. Estradiol increases mucus synthesis in bronchial epithelial cells. PLoS One 2014;9(6):e100633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Degano B, Prevost MC, Berger P, Molimard M, Pontier S, Rami J, et al. Estradiol decreases the acetylcholine-elicited airway reactivity in ovariectomized rats through an increase in epithelial acetylcholinesterase activity. Am J Respir Crit Care Med 2001;164(10 Pt 1):1849–54. [DOI] [PubMed] [Google Scholar]
  • 89.Townsend EA, Thompson MA, Pabelick CM, Prakash YS. Rapid effects of estrogen on intracellular Ca2+ regulation in human airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2010;298(4):L521–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zhang X, Bao W, Fei X, Zhang Y, Zhang G, Zhou X, et al. Progesterone attenuates airway remodeling and glucocorticoid resistance in a murine model of exposing to ozone. Mol Immunol 2018;96:69–77. [DOI] [PubMed] [Google Scholar]
  • 91.Bonham CA, Patterson KC, Strek ME. Asthma Outcomes and Management During Pregnancy. Chest 2018;153(2):515–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Gluck JC. The change of asthma course during pregnancy. Clin Rev Allergy Immunol 2004;26(3):171–80. [DOI] [PubMed] [Google Scholar]
  • 93.Kircher S, Schatz M, Long L. Variables affecting asthma course during pregnancy. Ann Allergy Asthma Immunol 2002;89(5):463–6. [DOI] [PubMed] [Google Scholar]
  • 94.Cohen JM, Bateman BT, Huybrechts KF, Mogun H, Yland J, Schatz M, et al. Poorly Controlled Asthma During Pregnancy Remains Common in the United States. J Allergy Clin Immunol Pract 2019;7(8):2672–80 e10. [DOI] [PubMed] [Google Scholar]
  • 95.Jensen ME, Robijn AL, Gibson PG, Oldmeadow C, Managing Asthma in Pregnancy study collaborative g, Breathing for Life Trial collaborative g, et al. Longitudinal Analysis of Lung Function in Pregnant Women with and without Asthma. J Allergy Clin Immunol Pract 2021;9(4):1578–85 e3. [DOI] [PubMed] [Google Scholar]
  • 96.Schatz M, Zeiger RS, Hoffman CP. Intrauterine growth is related to gestational pulmonary function in pregnant asthmatic women. Kaiser-Permanente Asthma and Pregnancy Study Group. Chest 1990;98(2):389–92. [DOI] [PubMed] [Google Scholar]
  • 97.Yoo EJ, Most JF, Lee NL, McWilliams T, Plante LA, Schulman ES. Improving asthma symptoms among inner-city women during pregnancy: a prospective cohort intervention. J Allergy Clin Immunol Pract 2021. [DOI] [PubMed] [Google Scholar]
  • 98.Grzeskowiak LE, Smith B, Roy A, Dekker GA, Clifton VL. Patterns, predictors and outcomes of asthma control and exacerbations during pregnancy: a prospective cohort study. ERJ Open Res 2016;2(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Murphy VE, Powell H, Wark PAB, Gibson PG. A prospective study of respiratory viral infection in pregnant women with and without asthma. Chest 2013;144(2):420–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Schatz M, Harden K, Forsythe A, Chilingar L, Hoffman C, Sperling W, et al. The course of asthma during pregnancy, post partum, and with successive pregnancies: a prospective analysis. J Allergy Clin Immunol 1988;81(3):509–17. [PubMed] [Google Scholar]
  • 101.Canalis E, Mazziotti G, Giustina A, Bilezikian JP. Glucocorticoid-induced osteoporosis: pathophysiology and therapy. Osteoporos Int 2007;18(10):1319–28. [DOI] [PubMed] [Google Scholar]
  • 102.Weare-Regales N, Hudey SN, Lockey RF. Practical Guidance for Prevention and Management of Glucocorticoid-Induced Osteoporosis for the Allergist/Immunologist. J Allergy Clin Immunol Pract 2021. [DOI] [PubMed] [Google Scholar]
  • 103.Lane NE. Glucocorticoid-Induced Osteoporosis: New Insights into the Pathophysiology and Treatments. Curr Osteoporos Rep 2019;17(1):1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Buckley L, Guyatt G, Fink HA, Cannon M, Grossman J, Hansen KE, et al. 2017 American College of Rheumatology Guideline for the Prevention and Treatment of Glucocorticoid-Induced Osteoporosis. Arthritis Rheumatol 2017;69(8):1521–37. [DOI] [PubMed] [Google Scholar]
  • 105.Camacho PM, Petak SM, Binkley N, Diab DL, Eldeiry LS, Farooki A, et al. American Association of Clinical Endocrinologists/American College of Endocrinology Clinical Practice Guidelines for the Diagnosis and Treatment of Postmenopausal Osteoporosis-2020 Update. Endocr Pract 2020;26(Suppl 1):1–46. [DOI] [PubMed] [Google Scholar]
  • 106.Schousboe JT, Shepherd JA, Bilezikian JP, Baim S. Executive summary of the 2013 International Society for Clinical Densitometry Position Development Conference on bone densitometry. J Clin Densitom 2013;16(4):455–66. [DOI] [PubMed] [Google Scholar]
  • 107.Bledsoe L, Alessi K, Toro JB, Giordano B, Hanypsiak BT. Fragility Fractures: Diagnosis and Treatment. Am J Orthop (Belle Mead NJ) 2018;47(12). [DOI] [PubMed] [Google Scholar]
  • 108.Morales DR, Lipworth BJ, Donnan PT, Jackson C, Guthrie B. Respiratory effect of beta-blockers in people with asthma and cardiovascular disease: population-based nested case control study. BMC Med 2017;15(1):18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Huang KY, Tseng PT, Wu YC, Tu YK, Stubbs B, Su KP, et al. Do beta-adrenergic blocking agents increase asthma exacerbation? A network meta-analysis of randomized controlled trials. Sci Rep 2021;11(1):452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Molis MA, Molis WE. Exercise-induced bronchospasm. Sports Health 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Woods BD, Sladen RN. Perioperative considerations for the patient with asthma and bronchospasm. British Journal of Anaesthesia 2009. [DOI] [PubMed] [Google Scholar]
  • 112.Morales DR, Jackson C, Lipworth BJ, Donnan PT, Guthrie B. Adverse respiratory effect of acute beta-blocker exposure in asthma: a systematic review and meta-analysis of randomized controlled trials. Chest 2014;145(4):779–86. [DOI] [PubMed] [Google Scholar]
  • 113.Morales DR, Dreischulte T, Lipworth BJ, Donnan PT, Jackson C, Guthrie B. Respiratory effect of beta-blocker eye drops in asthma: population-based study and meta-analysis of clinical trials. Br J Clin Pharmacol 2016;82(3):814–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Callaerts-Vegh Z, Evans KL, Dudekula N, Cuba D, Knoll BJ, Callaerts PF, et al. Effects of acute and chronic administration of beta-adrenoceptor ligands on airway function in a murine model of asthma. Proc Natl Acad Sci U S A 2004;101(14):4948–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Hanania NA, Singh S, El-Wali R, Flashner M, Franklin AE, Garner WJ, et al. The safety and effects of the beta-blocker, nadolol, in mild asthma: an open-label pilot study. Pulm Pharmacol Ther 2008;21(1):134–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Short PM, Williamson PA, Anderson WJ, Lipworth BJ. Randomized placebo-controlled trial to evaluate chronic dosing effects of propranolol in asthma. Am J Respir Crit Care Med 2013;187(12):1308–14. [DOI] [PubMed] [Google Scholar]
  • 117.De Graaf L. Salbutamol. Nursing 2018. [Google Scholar]
  • 118.Ullmann N, Caggiano S, Cutrera R. Salbutamol and around. Italian Journal of Pediatrics 2015. [Google Scholar]
  • 119.Yilmaz I, Turk M, Baran Ketencioglu B, Cetinkaya Z, Tutar N, Oymak FS, et al. The presence of underlying asthma should be investigated in patients diagnosed with ACE inhibitor induced cough. Clin Respir J 2020;14(4):382–8. [DOI] [PubMed] [Google Scholar]
  • 120.Morales DR, Lipworth BJ, Donnan PT, Wang H. Intolerance to angiotensin converting enzyme inhibitors in asthma and the general population: a UK population-based cohort study. The Journal of Allergy and Clinical Immunology: In Practice 2021(In press). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Yilmaz I. Angiotensin-Converting Enzyme Inhibitors Induce Cough. Turk Thorac J 2019;20(1):36–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Lemonde S, Turecki G, Bakish D, Du L, Hrdina PD, Bown CD, et al. Impaired repression at a 5-hydroxytryptamine 1A receptor gene polymorphism associated with major depression and suicide. J Neurosci 2003;23(25):8788–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Betz M, Fox BS. Prostaglandin E2 inhibits production of Th1 lymphokines but not of Th2 lymphokines. J Immunol 1991;146(1):108–13. [PubMed] [Google Scholar]
  • 124.Shibata Y, Henriksen RA, Honda I, Nakamura RM, Myrvik QN. Splenic PGE2-releasing macrophages regulate Th1 and Th2 immune responses in mice treated with heat-killed BCG. J Leukoc Biol 2005;78(6):1281–90. [DOI] [PubMed] [Google Scholar]
  • 125.Menard G, Turmel V, Bissonnette EY. Serotonin modulates the cytokine network in the lung: involvement of prostaglandin E2. Clin Exp Immunol 2007;150(2):340–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Kang BN, Ha SG, Bahaie NS, Hosseinkhani MR, Ge XN, Blumenthal MN, et al. Regulation of serotonin-induced trafficking and migration of eosinophils. PLoS One 2013;8(1):e54840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Nau F Jr., Miller J, Saravia J, Ahlert T, Yu B, Happel KI, et al. Serotonin 5-HT(2) receptor activation prevents allergic asthma in a mouse model. Am J Physiol Lung Cell Mol Physiol 2015;308(2):L191–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Shavitt RG, Gentil V, Mandetta R. The association of panic/agoraphobia and asthma: Contributing factors and clinical implications. General Hospital Psychiatry 1992;14(6):420–3. [DOI] [PubMed] [Google Scholar]
  • 129.Bussing R, Burket RC, Kelleher ET. Prevalence of anxiety disorders in a clinic-based sample of pediatric asthma patients. Psychosomatics 1996;37(2):108–15. [DOI] [PubMed] [Google Scholar]
  • 130.Kim J-Y, Ko I, Kim MS, Kim DW, Cho B-J, Kim D-K. Relationship of Chronic Rhinosinusitis with Asthma, Myocardial Infarction, Stroke, Anxiety, and Depression. J Allergy Clin Immunol Pract 2020;8(2):721–7.e3. [DOI] [PubMed] [Google Scholar]
  • 131.School HM. National Comorbidity Survey (NCS) 2007 [Data Table 2: 12-month prevalence DSM-IV/WMH-CIDI disorders by sex and cohort Available from: https://www.hcp.med.harvard.edu/ncs/index.php.
  • 132.Weiser EB. The Prevalence of Anxiety Disorders Among Adults with Asthma: A Meta-Analytic Review. Journal of Clinical Psychology in Medical Settings 2007;14(4):297–307. [Google Scholar]
  • 133.Scott KM, Von Korff M, Ormel J, Zhang MY, Bruffaerts R, Alonso J, et al. Mental disorders among adults with asthma: results from the World Mental Health Survey. Gen Hosp Psychiatry 2007;29(2):123–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.McLeish AC, Luberto CM, O’Bryan EM. Anxiety Sensitivity and Reactivity to Asthma-Like Sensations Among Young Adults With Asthma. Behav Modif 2016;40(1–2):164–77. [DOI] [PubMed] [Google Scholar]
  • 135.Denton E, Bondarenko J, Tay T, Lee J, Radhakrishna N, Hore-Lacy F, et al. Factors Associated with Dysfunctional Breathing in Patients with Difficult to Treat Asthma. J Allergy Clin Immunol Pract 2019;7(5):1471–6. [DOI] [PubMed] [Google Scholar]
  • 136.Sastre J, Crespo A, Fernandez-Sanchez A, Rial M, Plaza V, investigators of the CSG. Anxiety, Depression, and Asthma Control: Changes After Standardized Treatment. J Allergy Clin Immunol Pract 2018;6(6):1953–9. [DOI] [PubMed] [Google Scholar]
  • 137.Ahmedani BK, Peterson EL, Wells KE, Williams LK. Examining the relationship between depression and asthma exacerbations in a prospective follow-up study. Psychosom Med 2013;75(3):305–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Choi S, Kim SH, Lee JS. Association between depression and asthma in Korean adults. Allergy Asthma Proc 2017;38(3):37–46. [DOI] [PubMed] [Google Scholar]
  • 139.Lehrer P, Feldman J, Giardino N, Song HS, Schmaling K. Psychological aspects of asthma. Journal of Consulting and Clinical Psychology 2002. [DOI] [PubMed] [Google Scholar]
  • 140.Lee YC, Lee CTC, Lai YR, Chen VCH, Stewart R. Association of asthma and anxiety: A nationwide population-based study in Taiwan. Journal of Affective Disorders 2016. [DOI] [PubMed] [Google Scholar]
  • 141.Brown ES, Sayed N, Van Enkevort E, Kulikova A, Nakamura A, Khan DA, et al. A Randomized, Double-Blind, Placebo-Controlled Trial of Escitalopram in Patients with Asthma and Major Depressive Disorder. J Allergy Clin Immunol Pract 2018;6(5):1604–12. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES