Next Article in Journal
Coexisting Congenital Mesoblastic Nephroma and Lissencephaly: Unique Case Report with Pathological Analysis and Its Clinical Significance
Next Article in Special Issue
Severe Asthma and Active SARS-CoV-2 Infection: Insights into Biologics
Previous Article in Journal
Ezetimibe Anticancer Activity via the p53/Mdm2 Pathway
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Connections and Unmet Needs: Severe Asthma Biologics and Osteoporosis

by
Fabiana Furci
1,
Marco Umberto Scaramozzino
2,*,
Giuseppe Rocco Talesa
3 and
Corrado Pelaia
4
1
Provincial Healthcare Unit, Section of Allergology, 89900 Vibo Valentia, Italy
2
“La Madonnina” Outpatient Clinic, 89100 Reggio Calabria, Italy
3
Department of Orthopaedics and Traumatology, San Matteo Degli Infermi Hospital, 06049 Spoleto, Italy
4
Department of Medical and Surgical Sciences, University of Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2025, 13(1), 197; https://doi.org/10.3390/biomedicines13010197
Submission received: 23 December 2024 / Revised: 8 January 2025 / Accepted: 9 January 2025 / Published: 15 January 2025

Abstract

:
Asthma is a chronic inflammatory disease with the main anti-inflammatory drugs for better disease control being steroids or corticosteroids. The use of steroids in asthma patients, in particular in uncontrolled asthma patients, is associated with an increased risk of osteoporosis and fragility fractures. A single oral corticosteroid course increases the risk of osteoporosis and the continual use of inhaled corticosteroids is correlated over time to an increased risk for both bone conditions. With the use of new, available biologic therapies for asthma, perhaps even anticipating the times of their use in therapeutic management, in the current guidelines and with targeted strategies of prevention it may be possible to improve asthma management, preventing some comorbidities, such as osteoporosis.

1. Introduction

Asthma and osteoporosis are two medical conditions that may appear distinct, but which are interconnected through a series of pathophysiological mechanisms and shared risk factors. This article explores the links between asthma and osteoporosis, focusing on how chronic inflammation, glucocorticoid use, and other common risk factors contribute to the development of both diseases. Additionally, therapeutic implications and management strategies for patients suffering from both conditions are discussed. Asthma is a chronic inflammatory disease of the airways characterized by reversible bronchial obstruction, bronchial hyperreactivity, and the recurring symptoms of wheezing, dyspnoea, chest tightness, and cough. Asthma management is based on international guidelines that include the use of bronchodilators and inhaled corticosteroids (ICSs) to control inflammation and prevent exacerbations [1]. Osteoporosis is a systemic skeletal disease characterized by reduced bone mass and the deterioration of bone microarchitecture, leading to increased bone fragility and fracture risk. The risk factors for osteoporosis include advanced age, menopause, calcium and vitamin D deficiency, and prolonged use of glucocorticoids [2]. Chronic inflammation present in asthma can negatively impact bone health. Pro-inflammatory cytokines such as IL-6 and TNF-α, which are elevated in asthma, can stimulate bone resorption, reducing bone mineral density (BMD) [3]. Glucocorticoids, frequently used in asthma treatment, are well known for their adverse effects on bone mass. Chronic use of glucocorticoids can decrease intestinal calcium absorption and inhibit bone formation, significantly increasing the risk of osteoporosis and fractures [4]. Studies have shown that both oral corticosteroids (OCSs) and ICSs can contribute to bone loss, although the effects are more pronounced with OCSs [5,6], which accelerate bone loss and hence, increase the risk of fracture in patients taking > 7.5 mg of prednisolone daily [7]. However, evidence highlights that low- and medium-dose ICSs are safe and have little to no effect on BMD and metabolism [8].
A sedentary lifestyle is a risk factor for both conditions. Regular physical activity, particularly weight-bearing and resistance exercises, can improve both pulmonary function and bone health [9]. A diet poor in calcium and vitamin D can contribute to osteoporosis and worsen general health, negatively affecting asthma management. Proper nutrition is essential for maintaining good bone health and preventing chronic inflammation [10]. Some genetic factors may predispose individuals to both conditions. Ongoing research continues to explore the genetic links between asthma and osteoporosis, aiming to identify common genes that may influence the development of these diseases [11]. Asthma management should include the prudent use of glucocorticoids, balancing respiratory benefits with the risk of osteoporosis. Using inhalers at the minimal effective dose and introducing alternative medications where possible can help mitigate this risk. Additionally, the use of drugs such as bisphosphonates may be indicated to prevent bone loss in patients requiring long-term glucocorticoids [12]. For asthma patients using glucocorticoids, supplementation with calcium and vitamin D may be indicated. This approach can help maintain BMD and reduce the risk of osteoporosis. Tailored exercise programs will help. Exercise not only helps maintain good bone health but can also improve asthma control, reducing the frequency and severity of exacerbations.
The use of biological therapies has revolutionized the treatment of severe asthma. These therapies target specific pathways involved in the inflammatory process, offering a more personalized approach to asthma management. Biologics such as IL-5 and IL-5R (mepolizumab, reslizumab, and benralizumab) and the IL-4 receptor subunit α of IL-4 shared by the IL-4 and IL-13 receptor complexes, thereby inhibiting IL-4 and IL-13 signaling (dupilumab), have shown significant efficacy in reducing asthma exacerbations, improving lung function, and enhancing the quality of life (QoL) for patients with severe asthma. The introduction of these biologics also has implications for bone health. By potentially reducing the need for systemic glucocorticoids, biologics may help mitigate the associated risk of osteoporosis [13] (Figure 1). Considering that asthma patients exposed to OCSs or high-dose ICSs are more susceptible to bone comorbidities, asthma biologics have reported a safer profile, which, together with the efficacy of biologics, can make corticosteroids (CSs) the last therapeutic choice after biologics. As reported by Domingo C. et al., asthma biologics, such as mepolizumab, induce a de-escalation effect in OCS maintenance dose and a reduction in cumulative CS exposure in many severe asthma patients [14].
In particular, the reduction or avoidance of chronic OCS use in asthma management correlates with a reduction in the risk of osteoporosis and fractures [3,14]. Indeed, osteoporosis prevention treatment is recommended for patients (with additional risk factors) aged < 40–50 years in treatment with prednisolone > 7.5 mg/day or aged > 50 years receiving ≥ 5 mg/day [15]. Between January 2015 and February 2021, in a propensity score-matched, prospective cohort study using data from the International Severe Asthma Registry in which severe asthma patients and high OCS (HOCS) exposure (long-term OCSs for ≥1 year or ≥4 courses of rescue OCSs within a 12-month period) were identified, the initiation of biologics reported improvements in asthma outcomes, such as exacerbation rate, OCS exposure, and healthcare resource utilization. Patients treated with biologics had twice as high a chance of achieving a daily long-term OCS dose of less than 5 mg and a four times higher chance of reducing their total OCS dose by more than 75% from baseline than patients not treated with severe asthma biologics [16].
As reported by Chalictsios VV. et al., asthma patients treated with OCS or high ICS doses have a higher risk of presenting bone comorbidities and, therefore, striking the right balance between the efficacy and safety of steroids in these patients is important to improve QoL.
The use of glucocorticoids can induce osteoporosis, the most common adverse effect of corticosteroids. Osteoporosis is characterized by the structural deterioration of bone tissue and low bone mass, with consequent bone fragility and increased risk of fracture. Decreased BMD is related to a higher fracture risk. Fractures are related to morbidity, mortality, and increased healthcare costs [3]. Thus, the pharmacoeconomic impact of biological therapies in asthma management is substantial. While these treatments are often more expensive upfront compared to traditional therapies, they can reduce healthcare costs in the long term by decreasing the frequency of severe asthma exacerbations, hospitalizations, and the overall burden of the disease. Furthermore, by potentially reducing the need for systemic glucocorticoids, biologics may lower the incidence of glucocorticoid-induced osteoporosis, thereby reducing the costs related to osteoporosis management and fracture treatment [17]. It is crucial to regularly monitor BMD in asthmatic patients undergoing long-term glucocorticoid therapy. This can be carried out through exams such as dual-energy X-ray absorptiometry (DEXA), which allows the early identification of bone loss and intervention with preventive measures. The integrated management of asthma and osteoporosis requires a multidisciplinary approach that considers the interconnection between the two conditions. A thorough understanding of the common mechanisms and preventive strategies can improve the QoL for patients affected by both diseases. Collaboration between pulmonologists, endocrinologists, and other specialists is essential to develop personalized treatment plans that address the specific needs of patients. Ultimately, the management of asthma and osteoporosis must consider the interplay between these conditions. Ensuring optimal asthma control while minimizing the adverse effects of treatment on bone health requires careful consideration of medication choices, lifestyle modifications, and regular monitoring. Reviewing the CS dose and using the lowest dose possible is a key strategy in asthma management [4]. The advent of biological therapies, considered as OCS-sparing agents, provides new opportunities for managing severe asthma more effectively and with potentially fewer side effects, highlighting the importance of ongoing research and innovation in the treatment of these chronic conditions.

2. Materials and Methods

2.1. Osteoporosis and the IL-31/33 Axis

Bronchial asthma is a chronic respiratory disease characterized by the inflammation and hyperreactivity of the airways. The interleukins IL-31 and IL-33 play a crucial role in the pathological mechanisms that govern this condition [4]. IL-31, produced mainly by T helper 2 (Th2) cells, is involved in modulating the inflammatory response. This cytokine is known to promote the activation of mast cell and basophil cells, facilitating allergic inflammation and contributing to the production of other pro-inflammatory cytokines and chemokines, which in turn amplify asthma symptoms [17,18,19]. Its secretion is increased in conditions of severe asthma, suggesting a direct link between elevated IL-31 levels and disease severity [20]. On the other hand, IL-33, a high alarmin released by damaged epithelial cells, plays a critical role in the activation of immune system cells, including T cells and mast cells [21]. Activation occurs in situations of environmental stress or tissue injury, stimulating a robust inflammatory response. IL-33 induces the production of other Th2 cytokines, such as IL-4 and IL-13, which are important for the allergic response and chronic inflammation in the airways. Interactions between IL-31 and IL-33 lead to further amplification of inflammation, contributing to the pathogenesis of asthma [22]. This chronic inflammation is not only limited to the airways, but also causes systemic effects that can affect bone metabolism. Pro-inflammatory cytokines, activated by the mediators of the IL-31/IL-33 axis, can stimulate osteoclastogenesis, i.e., the formation of osteoclasts, cells responsible for bone resorption. As a result, asthma patients with sustained inflammation may be at increased risk of developing osteoporosis [23]. Furthermore, prolonged use of CS for asthma control further compromises BMD, making patients vulnerable to bone fractures [24]. Therefore, it is critical for pulmonary physicians to integrate an osteoporosis risk assessment into the management of asthma patients, as chronic inflammation and CS treatment may interact negatively to impact bone health. The identification of the IL-31/IL-33 axis as a risk factor for osteoporosis not only improves the understanding of the pathophysiology of asthma, but also highlights the importance of therapeutic strategies that consider both respiratory and systemic symptoms. Preventive strategies such as the adoption of a healthy lifestyle, combined with scrupulous pharmacological management, can have their say in mitigating the side effects linked to osteoporosis in these patients [6,25]. Continued research on the effects of IL-31 and IL-33 in asthma may lead to new therapeutic innovations, thus improving the QoL of asthma patients and reducing the risk of associated complications (Figure 2 and Figure 3) [26].

2.2. Overview of Available OCS and Conversion of Doses of OCS

The main treatment in asthma is the use of CS given their capacity to act on inflammation which is typical of asthma. It is important to understand the correct use of the available corticosteroids and their pharmacokinetics. An OCS conversion table can be found at the end of the paragraph (Table 1) [27].
Cortisone: This hormone acts on the intermediate metabolism, increasing the availability of glucose, and inducing proteolysis and lipolysis, thus placing the organism in an energetically active state, which is useful for overcoming many stressful conditions; it alters the normal fluid and electrolyte balance, increasing the reabsorption of sodium and the excretion of potassium and calcium. Finally, it induces vasoconstriction at the microcirculation level. Indicated in the treatment of acute and chronic inflammatory pathologies that require systemic therapies with OCS, it can also be useful in the management of symptoms in allergic and neoplastic conditions [28].
Hydrocortisone: Hydrocortisone is the main glucocorticoid produced by the adrenal cortex. It works by binding to its receptors present inside the cells, promoting the expression of some specific genes with the consequent activation of molecules with anti-inflammatory action and the simultaneous inhibition of molecules with pro-inflammatory activity. It is used against inflammation, allergic reactions, diseases involving collagen, asthma, adrenal insufficiency, some forms of cancer, Addison’s disease, and autoimmune diseases such as arthritis, lupus, psoriasis, ulcerative colitis, and Crohn’s disease [29].
Deflazacort: This is a corticosteroid which, after oral intake, is absorbed rapidly and completely in the intestine with peak plasma levels reached in 1–2 h. Subsequently, the original compound is deacetylated at position 21 to be transformed into an active metabolite characterized by high binding affinity for tissue glucocorticoid receptors. The absolute oral bioavailability is 68%, the binding to plasma proteins is 39.8%, while the almost complete elimination of the active metabolite occurs within 24 h mainly through urine. It is a drug that has indications in the treatment of various pathologies including sarcoidosis, juvenile chronic arthritis, polymyalgia rheumatica, and rheumatoid arthritis [30].
Prednisolone: This works by preventing the release of molecules that trigger inflammation. It is used in the treatment of many different disorders associated with inflammation, for example, allergies, ulcerative colitis, arthritis, lupus, psoriasis, or respiratory problems [31].
Prednisone: This is a synthetic hormone that belongs to the corticosteroid group, and has properties to reduce pain, swelling, stiffness, redness and heat in affected tissues. It is used in some rheumatological conditions such as rheumatoid arthritis, acute gouty arthritis, Still’s disease, ankylosing spondylitis, systemic lupus erythematosus, dermatomyositis, bronchial asthma, atopic and contact dermatitis, in sarcoidosis, to treat some diseases of the blood, as palliative therapy for some tumors, in addition to therapies for gastrointestinal pathologies [32].
Methylprednisolone: Methylprednisolone is a powerful anti-inflammatory that, similarly to other corticosteroid drugs, appears to inhibit the release of arachidonic acid, the precursor of important inflammatory mediators. Oral methylprednisolone is used in the treatment of adrenal insufficiency; joint pathologies, such as psoriatic arthritis and rheumatoid arthritis; skin conditions such as severe psoriasis, pemphigus, allergic diseases; eye disorders such as diffuse posterior uveitis, optic neuritis, iritis, and iridocyclitis; lung diseases such as sarcoidosis and emphysema; blood pathologies and tumors such as leukemia; intestinal diseases such as ulcerative colitis and regional enteritis; brain disorders (e.g., tuberculous meningitis); and other pathologies [33].
Triamcinolone: This acts from a molecular point of view, determining the expression of the lipocortin enzyme, capable of inhibiting phospholipase A2 and consequently reducing the availability of arachidonic acid. The reduced concentration of the starting substrate translates into a modest synthesis of inflammatory mediators such as leukotrienes, prostaglandins, and prostacyclins with the consequent control of the entire inflammatory process and related tissue damage. Once its activity is complete, the active ingredient is metabolized in the liver, mainly through hydroxylation processes, and subsequently excreted via the kidneys. It can be used to treat many different conditions: allergies, disorders of the skin and related tissues (hair and nails) such as psoriasis, rheumatic diseases, ulcerative colitis (chronic inflammatory bowel disease), and some respiratory diseases [34].
Betamethasone: This belongs to the class of long-acting glucocorticoids. The main indications are for the treatment of bronchial asthma, severe allergic diseases, rheumatoid arthritis, collagenopathies, inflammatory skin diseases, tumors of the lymphatic system such as acute and chronic malignant hemolymphopathies, Hodgkin’s disease, nephrotic syndrome, ulcerative colitis and Crohn’s disease, pemphigus, sarcoidosis, rheumatic carditis, ankylosing spondylitis, hemolytic anemia, agranulocytosis, and thrombocytopenic purpura [35].
Dexamethasone: This is a synthetic hormone with an anti-inflammatory activity approximately 7 times more powerful than prednisolone and 30 times more powerful than hydrocortisone. The molecule exerts its action on the hypothalamic–pituitary–adrenal axis by binding to specific receptors on the cellular plasma membrane. In other tissues, the molecule passes cell membranes and diffuses into the cytosol, combining with specific cytoplasmic receptors, the glucocorticoid receptor. The complex that is formed enters the cell nucleus and stimulates protein synthesis. Like other adrenocortical steroids, dexamethasone has anti-allergic, anti-shock, antitoxic, antifebrile, and immunosuppressive properties. Characteristically, this molecule predominantly presents glucocorticoid activity; its ability to determine the renal retention of sodium and water (i.e., mineralocorticoid activity) is vastly lower than that of other molecules of the same class. After oral administration, dexamethasone is rapidly absorbed from the gastrointestinal tract. Following intravenous injection, peak plasma levels are reached in 5 min. Its half-life in plasma is approximately 190 min. It may be more interesting to consider the biological half-life of the molecule, which is equal to 36–54 h, which makes dexamethasone a substance that is indicated for the treatment of pathologies that require prolonged glucocorticoid action such as in patients suffering from rheumatoid arthritis, bronchial asthma, purpura, undergoing chemotherapy, etc. [36].

2.3. The Impact of Severe Asthma Biologics in Sparing Steroid Therapy

Severe asthma biologics that act on the different steps of type 2 inflammation are a revolutionary strategy to treat the disease. When the diagnosis of asthma is confirmed and comorbidities have been addressed, such as rhinitis, rhinosinusitis, nasal polyps, and gastroesophageal reflux, severe asthma is classified as “asthma which requires treatment with high dose ICSs plus a second controller (and/or systemic corticosteroids) to prevent it from becoming ‘uncontrolled’ or which remains ‘uncontrolled’ despite this therapy.” Patients affected by uncontrolled asthma present high rates of exacerbations with the need for glucocorticoid therapy.
There are now several mAb therapies (“biologics”) available to treat severe asthma. These drugs all reduce the exacerbation rate [37]. Once comorbidities have been addressed, and excluding those patients who are poorly adherent to inhaled therapy, the prevalence of severe asthma affects approximately 3.7% of the asthma population [1], resulting in significant asthma-related healthcare costs, frequent exacerbations, impairment of QoL, missed work/school days, and frequent healthcare utilization. Severe asthma exacerbations need OCS or may need maintenance oral corticosteroids (mOCSs) to control the disease. The long-term use of OCS is associated with significant long-term morbidity, such as adrenal suppression, osteopenia and osteoporosis, increased risk of type II diabetes, cataracts, and obesity [38]. Asthma can be considered the result of the combination of genetic susceptibility with external factors such as allergens, microbes, pollutants, and other triggers that, acting on the airway epithelium, induce the release of “alarmins”, including interleukin (IL)-25, IL-33, and thymic stromal lymphopoietin (TSLP), which result in the production of T2 cytokines from cells including Th2 cells and type 2 innate lymphoid cells (ILC2) [39]. Severe asthma biologics that are essential in the exacerbation pathogenesis of asthma act on one or more mediators or cells within this pathway. Type 2 cytokines that play a key role in asthma disease include IL-4, related to IgE production from B-cell;, IL-5, related to the eosinophil cycle; and IL-13, related to mucus hypersecretion and airway hyperresponsiveness [40].
Omalizumab (“Xolair”), the first mAb approved to treat severe asthma, with an antiviral response and a reduction in virus-induced exacerbations, binds to free circulating IgE, inducing the inhibition of attachment to its receptor (FCεRI) and a reduction in downstream effects, including mast cell degranulation and the expression of inflammatory cytokines such as IL-3, IL-4, IL-5, IL-6, and IL-13 [41]. Various real-world, observational trials reported mOCS dose reduction in patients treated with omalizumab [42,43,44,45]. In a systematic review of 42 observational or registry trials, the authors highlighted a mean OCS dose reduction of 68% at 12 months, with a broad range from −78% to −12% [46]. The large “eXpeRience” registry reported on over 260 patients on baseline OCS and reported that 57% either reduced or stopped mOCS with 1 year of omalizumab treatment. Moreover, it was reported that the mean prednisolone dose at the end of 1 year was 7.7 mg compared to 15.5 mg at baseline [47]. In a randomized trial, a major reduction or interruption of mOCS in patients treated with omalizumab rather than with optimal standard care was described [48].
Mepolizumab (“Nucala”), the first mAb introduced for severe eosinophilic asthma, acts on the IL-5 cytokine involved in the development, migration, and survival of eosinophils [38,40]. The phase 3 MENSA trial reported a reduction in exacerbations (52%) in patients treated with mepolizumab compared to patients treated with placebo [47] and, moreover, a significant reduction in OCS dose in patients dependent on daily OCS to maintain asthma control. The SIRIUS trial reported that at 24 weeks, 14% of the asthma patients in mepolizumab treatment were able to stop prednisolone with a general median reduction of 50% OCS use [48].
A retrospective analysis of 99 patients in mepolizumab treatment reported a 50% reduction in exacerbation rate, with a reduction in prednisolone dose of at least 50% [49]. Regarding Reslizumab (“Cinqaero”), a recombinant humanized IgG4 mAb that acts on IL-5, efficacy in terms of reduction in mOCS use has not been formally reported. Indeed, mepolizumab, benralizumab, and dupilumab are the only severe asthma biologics for which a significant reduction in daily OCS use has been reported.
Benralizumab (“Fasenra”), which acts on the IL-5 pathway through ligation to the α subunit of the IL-5 receptor (IL-5R-α), induces cell-mediated cytotoxicity and consequent cell apoptosis (eosinophils and basophils) on which IL-5R-α is expressed [50,51]. In the ZONDA study, which enrolled OCS-dependent asthma patients, a 50% reduction in OCS dose in benralizumab-treated patients was reported compared with placebo. Moreover, a reduction of 70% in exacerbations was described [52]. In the open-label PONENTE study, it was reported that in 598 OCS-dependent patients treated with benralizumab over 80% were able to stop the use of steroids, or achieved a dosage of 5 mg or less if the reason for stopping the reduction was adrenal insufficiency rather than asthma [53].
Dupilumab (“Dupixent”), an mAb targeted against the α subunit of the IL-4 receptor, has also reported a significant reduction in mOCS use in asthma. The LIBERTY ASTHMA VENTURE trial reported an overall median prednisolone dose reduction of 50% compared to placebo, and 69% reduced their dose below 5 mg·day 1 compared with 33% of the placebo group [54]. Moreover, in this trial, a significant reduction in corticosteroid dose in both low (<300 cells·μL−1) and high (≥300 cells·μL−1) blood eosinophil groups was seen, although the greatest response was reported in the asthma patients with higher baseline blood eosinophil counts [55].
Tezepelumab is an mAb that targets the epithelial alarmin TSLP, which is released by epithelial cells in response to various pro-inflammatory stimuli and related to both T2 cytokine expression and asthma severity in asthma [56,57]. In the SOURCE trial, with patients with OCS-dependent asthma, randomized to tezepelumab or placebo, the investigators reported that the trial did not meet the primary endpoint of reducing daily OCS whilst maintaining asthma control [58,59].

3. Discussion

Asthma patients are characterized by an increased risk of osteoporosis and fragility fractures, in particular of vertebra and forearm/wrist fractures, compared with the general population. The mechanism of risk for osteoporosis in asthma patients can be divided into asthma-induced osteoporosis and corticosteroid-induced osteoporosis. The direct effect of asthma leading to osteoporosis is related to disease inflammation (e.g., neutrophil-mediated inflammation, TNF-α and IL-6). TNF-α, an inhibitor of osteogenesis and an activator of osteoclastogenesis, is involved in neutrophil migration, which is associated with a decrease in BMD. IL-6 is also an important inflammatory factor involved in neutrophil-inflammation-mediated osteoporosis in asthma patients. The overexpression of IL-6 induces an inhibition of the differentiation of osteoblasts, with an increased expression of RANKL and an increase in osteoclast activity [60].
Knowledge of the risk of CS-related toxicities correlated to OCS or parenteral CS therapies for the management of acute severe asthma exacerbations is an important factor to consider in terms of asthma management and safety. It has been reported that the risk of osteoporosis diagnosis and fracture increases per 1 g in systemic CS exposure in asthma patients [38]. Therefore, the use of anti-osteoporotic treatment is essential in adult patients with high cumulative CS exposure (>5 g/year) [15,61]. Moreover, an analysis published in 2021 highlighted how there is an inter-person variability in the risk of OCS-related toxicity among severe asthma patients, as measured by the Glucocorticoid Toxicity Index (GTI), with only a modest correlation between recent OCS exposure and GTI score [62]. This inter-variability appears to be part of the heterogeneity of patients suffering from asthma, with their different phenotypes and endotypes and varying responses to treatments, including biological therapies, which have shown a more favorable safety profile compared to therapies that correlate to multi-comorbidities [62,63]. Indeed, as reported by Cutroneo PM et al. conducting an overview of the safety data of severe asthma biologics in VigiBase, the World Health Organization global pharmacovigilance database, the most frequently reported suspected adverse drug reactions are the well-known adverse effects such as general disorders, injection-site reactions, nasopharyngitis, headache, and hypersensitivity, while others, such as asthma exacerbation or therapeutic failure, may be related to the indication of use. The authors reported that a significant confounder in the analysis conducted is represented by the concomitant or recent use of OCSs by severe asthma patients [64]. Albeit, after starting biologics, OCS sparing up to withdrawal can be considered a primary outcome in the management of severe asthma patients to minimize side effects, and the frequent or continuous use of OCSs or long-term high-dose ICSs could be associated with many potential adverse events, such as fluid retention, bone damage, elevated blood sugars, and psychiatric problems. These could be clinically relevant, in particular when high doses are administered for a prolonged period of time [65]. The reduction or suppression of OCSs during severe asthma biological therapy may unmask some comorbidity symptoms or patients’ underlying diseases (e.g., psoriasis and multiple inflammatory diseases), known as glucocorticoid deprivation syndrome [64].
From the knowledge of the molecules involved in asthmatic inflammation, such as IL-6, it is possible to understand the pivotal role of severe asthma biological therapies both in terms of asthma and in relation to pathologies such as osteoporosis which correlate with asthma inflammation [14,66]. Indeed, as reported by Perlato M. et al., considering that in severe asthma patients, apart from the phenomenon known as “airway remodeling”, the prolonged use of OCS induces “patient remodeling”, with the manifestation of physical changes (e.g., suppression of hypothalamic–pituitary–adrenal axis, diabetes, adrenal insufficiency, obesity, buffalo hump, striae rubrae, and osteoporosis), the use of biologics can improve asthma control and avoid or limit the inflammation-induced “airway remodeling” and the OCS-induced “patient remodeling” [67].
The core of the multidisciplinary approach to airway diseases is a person-centered model of care focused on the “Treatable Traits” that are causing symptoms and impacts. This approach is integral to addressing the complexity and heterogeneity of severe asthma patients [68].
In severe asthma, multidisciplinary care that includes diverse healthcare professionals (e.g., endocrinologists, allergists, otolaryngologists, and gastroenterologists) that involves systematic or multidimensional assessment is recommended by international guidelines [37,69]. Observational and registry studies, and a meta-analysis, reported that these approaches to severe asthma management induce an improvement in asthma control (SMD 0.36 (95% CI 0.23–0.59)) and in asthma QoL (SMD 0.36 (95% CI 0.22–0.50)), reduce acute attacks (SMD −0.34 (95% CI −0.44– −0.23)), and reduce OCS use (reduction of 11.5 mg on average) [70,71].

4. Conclusions

Physicians managing severe asthma should remain vigilant in assessing the CS-related toxicity risk in patients receiving biologics or long-term OCS, especially in patients with additional osteoporotic risk factors. A timely diagnosis is necessary in order to increase the possibility of having important comorbidities following prolonged use of oral steroids and reduce the costs of healthcare spending due to excessive use of OCS and hospitalization of patients suffering from severe asthma.

Author Contributions

Conceptualization, F.F.; resources, M.U.S., C.P. and F.F.; writing—original draft preparation, F.F. and M.U.S.; writing—review and editing, F.F., G.R.T., M.U.S. and C.P.; drawing of the figures, F.F., G.R.T. and M.U.S.; project administration, C.P. and F.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Global Initiative for Asthma. Global Strategy for Asthma Management and Prevention 2024. Update May 2024. Available online: http://www.ginasthma.org/ (accessed on 31 December 2024).
  2. Kearney, D.M.; Lockey, R.F. Osteoporosis and asthma. Ann. Allergy Asthma Immunol. 2006, 96, 769–774. [Google Scholar] [CrossRef] [PubMed]
  3. Chalitsios, C.V.; Shaw, D.E.; McKeever, T.M. Corticosteroids and bone health in people with asthma: A systematic review and meta-analysis. Respir. Med. 2021, 181, 106374. [Google Scholar] [CrossRef] [PubMed]
  4. Chalitsios, C.V.; McKeever, T.M.; Shaw, D.E. Incidence of osteoporosis and fragility fractures in asthma: A UK population-based matched cohort study. Eur. Respir. J. 2021, 57, 2001251. [Google Scholar] [CrossRef] [PubMed]
  5. Heffler, E.; Madeira, L.N.G.; Ferrando, M.; Puggioni, F.; Racca, F.; Malvezzi, L.; Passalacqua, G.; Canonica, G.W. Inhaled Corticosteroids Safety and Adverse Effects in Patients with Asthma. J. Allergy Clin. Immunol. Pract. 2018, 6, 776–781. [Google Scholar] [CrossRef] [PubMed]
  6. Smith, B.J.; Phillips, P.J.; Heller, R.F. Asthma and chronic obstructive airway diseases are associated with osteoporosis and fractures: A literature review. Respirology 1999, 4, 101–109. [Google Scholar] [CrossRef]
  7. Eastell, R. Management of corticosteroid-induced osteoporosis. UK Consensus Group. Meeting on Osteoporosis. J. Intern. Med. 1995, 237, 439–447. [Google Scholar] [CrossRef]
  8. Woodcock, A. Effects of inhaled corticosteroids on bone density and metabolism. J. Allergy Clin. Immunol. 1998, 101, S456–S459. [Google Scholar] [CrossRef]
  9. Chalitsios, C.V.; Shaw, D.E.; McKeever, T.M. Risk of osteoporosis and fragility fractures in asthma due to oral and inhaled corticosteroids: Two population-based nested case-control studies. Thorax 2021, 76, 21–28. [Google Scholar] [CrossRef]
  10. Cardet, J.C.; Bulkhi, A.A.; Lockey, R.F. Nonrespiratory Comorbidities in Asthma. J. Allergy Clin. Immunol. Pract. 2021, 9, 3887–3897. [Google Scholar] [CrossRef]
  11. Weare-Regales, N.; Hudey, S.N.; Lockey, R.F. Practical Guidance for Prevention and Management of Glucocorticoid-Induced Osteoporosis for the Allergist/Immunologist. J. Allergy Clin. Immunol. Pract. 2021, 9, 1841–1850. [Google Scholar] [CrossRef]
  12. Carpaij, O.A.; van den Berge, M. The asthma-obesity relationship: Underlying mechanisms and treatment implications. Curr. Opin. Pulm. Med. 2018, 24, 42–49. [Google Scholar] [CrossRef] [PubMed]
  13. Maslan, J.; Mims, J.W. What is asthma? Pathophysiology, demographics, and health care costs. Otolaryngol. Clin. N. Am. 2014, 47, 13–22. [Google Scholar] [CrossRef] [PubMed]
  14. Domingo, C.; Sogo, A.; Casado, E.; Martinez-Moragon, E.; Blanco-Aparicio, M.; Carrillo, T. Potential impact of mepolizumab in stepping down anti-osteporotic treatment in corticosteroid-dependent asthma. Front. Pharmacol. 2023, 14, 1183156. [Google Scholar] [CrossRef] [PubMed]
  15. Buckley, L.; Guyatt, G.; Fink, H.A.; Cannon, M.; Grossman, J.; Hansen, K.E.; Humphrey, M.B.; Lane, N.E.; Magrey, M.; Miller, M.; et al. American college of rheumatology guideline for the prevention and treatment of glucocorticoid-induced osteoporosis. Arthritis Rheumatol. 2017, 69, 1521–1537. [Google Scholar] [CrossRef]
  16. Chen, W.; Tran, T.N.; Sadatsafavi, M.; Murray, R.; Wong, N.C.B.; Ali, N.; Ariti, C.; Bulathsinhala, L.; Gil, E.G.; Fitzgerald, J.M.; et al. Impact of Initiating Biologics in Patients with Severe Asthma on Long-Term Oral Corticosteroids or Frequent Rescue Steroids (GLITTER): Data From the International Severe Asthma Registry. J. Allergy Clin. Immunol. Pract. 2023, 11, 2732–2747. [Google Scholar] [CrossRef]
  17. González-Barcala, F.J.; Muñoz-Gall, X.; Mariscal, E.; García, A.; Yang, S.; van de Wetering, G.; Izquierdo-Alonso, J.L. Cost-effectiveness analysis of anti-IL-5 therapies of severe eosinophilic asthma in Spain. J. Med. Econ. 2021, 24, 874–882. [Google Scholar] [CrossRef]
  18. Virchow, J.C.; Barnes, P.J. Asthma. Semin. Respir. Crit. Care Med. 2012, 33, 577. [Google Scholar]
  19. Barnes, P.J. Cellular and molecular mechanisms of asthma and COPD. Clin. Sci. 2017, 131, 1541–1558. [Google Scholar] [CrossRef]
  20. Barnes, P.J.; Adcock, I.M. How do corticosteroids work in asthma? Ann. Intern. Med. 2003, 139, 359–370. [Google Scholar] [CrossRef]
  21. Ginaldi, L.; De Martinis, M.; Ciccarelli, F.; Saitta, S.; Imbesi, S.; Mannucci, C.; Gangemi, S. Increasedlevels of interleukin 31 (IL-31) in osteoporosis. BMC Immunol. 2015, 16, 60. [Google Scholar] [CrossRef]
  22. De Martinis, M.; Sirufo, M.M.; Suppa, M.; Ginaldi, L. IL-33/IL-31 Axis in Osteoporosis. Int. J. Mol. Sci. 2020, 21, 1239. [Google Scholar] [CrossRef] [PubMed]
  23. Sirufo, M.M.; Suppa, M.; Ginaldi, L.; De Martinis, M. Does Allergy Break Bones? Osteoporosis and Its Connection to Allergy. Int. J. Mol. Sci. 2020, 21, 712. [Google Scholar] [CrossRef] [PubMed]
  24. Malerba, M.; Romanelli, G.; Grassi, V. Glucocorticoid-induced osteoporosis in asthma and respiratory diseases. Front. Horm. Res. 2002, 30, 86–93. [Google Scholar] [PubMed]
  25. Barnes, P.J. Pathophysiology of asthma. Br. J. Clin. Pharmacol. 1996, 42, 3–10. [Google Scholar] [CrossRef]
  26. Chung, K.F.; Barnes, P.J. Role of inflammatory mediators in asthma. Br. Med. Bull. 1992, 48, 135–148. [Google Scholar] [CrossRef]
  27. Parente, L. Deflazacort: Therapeutic index, relative potency and equivalent doses versus other corticosteroids. BMC Pharmacol. Toxicol. 2017, 18, 1. [Google Scholar] [CrossRef]
  28. Cortisone. Summary of Product Characteristics. Available online: https://it.wikipedia.org/wiki/Cortisone (accessed on 31 December 2024).
  29. Hydrocortisone. Summary of Product Characteristics. Available online: https://it.wikipedia.org/wiki/Idrocortisone (accessed on 31 December 2024).
  30. Deflazacort. Summary of Product Characteristics. Available online: https://it.wikipedia.org/wiki/Deflazacort (accessed on 31 December 2024).
  31. Prednisolone. Summary of Product Characteristics. Available online: https://it.wikipedia.org/wiki/Prednisolone (accessed on 31 December 2024).
  32. Prednisone. Summary of Product Characteristics. Available online: https://it.wikipedia.org/wiki/Prednisone (accessed on 31 December 2024).
  33. Methylprednisolone. Summary of Product Characteristics. Available online: https://it.wikipedia.org/wiki/Metilprednisolone (accessed on 31 December 2024).
  34. Triamcinolone. Summary of Product Characteristics. Available online: https://it.wikipedia.org/wiki/Triamcinolone_acetonide (accessed on 31 December 2024).
  35. Betamethasone. Summary of Product Characteristics. Available online: https://it.wikipedia.org/wiki/Betametasone (accessed on 31 December 2024).
  36. Dexamethasone. Summary of Product Characteristics. Available online: https://it.wikipedia.org/wiki/Desametasone (accessed on 31 December 2024).
  37. Chung, K.F.; Wenzel, S.E.; Brozek, J.L.; Bush, A.; Castro, M.; Sterk, P.J.; Adcock, I.M.; Bateman, E.D.; Bel, E.H.; Bleecker, E.R.; et al. International ERS/ATS guidelines on definition, evaluation and treatment of severe asthma. Eur. Respir. J. 2014, 43, 343–373. [Google Scholar] [CrossRef]
  38. Price, D.B.; Trudo, F.; Voorham, J.; Xu, X.; Kerkhof, M.; Ling Zhi Jie, J.; Tran, T.N. Adverse outcomes from initiation of systemic corticosteroids for asthma: Long-term observational study. J. Asthma Allergy 2018, 11, 193–204. [Google Scholar] [CrossRef]
  39. Fahy, J.V. Type 2 inflammation in asthma—Present in most, absent in many. Nat. Rev. Immunol. 2015, 15, 57–65. [Google Scholar] [CrossRef]
  40. Lambrecht, B.N.; Hammad, H.; Fahy, J.V. The cytokines of asthma. Immunity 2019, 50, 975–991. [Google Scholar] [CrossRef]
  41. Teach, S.J.; Gill, M.A.; Togias, A.; Sorkness, C.A.; Arbes, S.J., Jr.; Calatroni, A.; Wildfire, J.J.; Gergen, P.J.; Cohen, R.T.; Pongracic, J.A.; et al. Preseasonal treatment with either omalizumab or an inhaled corticosteroid boost to prevent fall asthma exacerbations. J. Allergy Clin. Immunol. 2015, 136, 1476–1485. [Google Scholar] [CrossRef] [PubMed]
  42. Adachi, M.; Kozawa, M.; Yoshisue, H.; Milligan, K.L.; Nagasaki, M.; Sasajima, T.; Miyamoto, T.; Ohta, K. Real-world safety and efficacy of omalizumab in patients with severe allergic asthma: A long-term post-marketing study in Japan. Respir. Med. 2018, 141, 56–63. [Google Scholar] [CrossRef] [PubMed]
  43. Gevaert, P.; Omachi, T.A.; Corren, J.; Mullol, J.; Han, J.; Lee, S.E.; Kaufman, D.; Ligueros-Saylan, M.; Howard, M.; Zhu, R. Efficacy and safety of omalizumab in nasal polyposis: 2 randomized phase 3 trials. J. Allergy Clin. Immunol. 2020, 146, 595–605. [Google Scholar] [CrossRef] [PubMed]
  44. Cox, L.; Platts-Mills, T.A.; Finegold, I.; Schwartz, L.B.; Simons, F.E.R.; Wallace, D.V. American Academy of Allergy, Asthma & Immunology/American College of Allergy, Asthma and Immunology joint task force report on omalizumab-associated anaphylaxis. J. Allergy Clin. Immunol. 2007, 120, 1373–1377. [Google Scholar] [PubMed]
  45. Smith, D.A.; Minthorn, E.A.; Beerahee, M. Pharmacokinetics and pharmacodynamics of mepolizumab, an anti-interleukin-5 monoclonal antibody. Clin. Pharmacokinet. 2011, 50, 215–227. [Google Scholar] [CrossRef]
  46. Flood-Page, P.T.; Menzies-Gow, A.N.; Kay, A.B.; Robinson, D.S. Eosinophil’s role remains uncertain as anti–interleukin-5 only partially depletes numbers in asthmatic airway. Am. J. Respir. Crit. Care Med. 2003, 167, 199–204. [Google Scholar] [CrossRef]
  47. Ortega, H.G.; Liu, M.C.; Pavord, I.D.; Brusselle, G.G.; FitzGerald, J.M.; Chetta, A.; Humbert, M.; Katz, L.E.; Keene, O.N.; MENSA Investigators; et al. Mepolizumab treatment in patients with severe eosinophilic asthma. N. Engl. J. Med. 2014, 371, 1198–1207. [Google Scholar] [CrossRef]
  48. Bel, E.H.; Wenzel, S.E.; Thompson, P.J.; Prazma, C.M.; Keene, O.N.; Yancey, S.W.; Ortega, H.G.; Pavord, I.D. SIRIUS Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic asthma. N. Engl. J. Med. 2014, 371, 1189–1197. [Google Scholar] [CrossRef]
  49. Kavanagh, J.E.; d’Ancona, G.; Elstad, M.; Green, L.; Fernandes, M.; Thomson, L.; Roxas, C.; Dhariwal, J.; Nanzer, A.M.; Kent, B.D.; et al. Real-world effectiveness and the characteristics of a ‘super-responder’ to mepolizumab in severe eosinophilic asthma. Chest 2020, 158, 491–500. [Google Scholar] [CrossRef]
  50. Ghazi, A.; Trikha, A.; Calhoun, W.J. Benralizumab—A humanized mAb to IL-5Rα with enhanced antibody-dependent cell-mediated cytotoxicity—A novel approach for the treatment of asthma. Expert. Opin. Biol. Ther. 2012, 12, 113–118. [Google Scholar] [CrossRef]
  51. Kolbeck, R.; Kozhich, A.; Koike, M.; Peng, L.; Andersson, C.K.; Damschroder, M.M.; Reed, J.L.; Woods, R.; Dall’acqua, W.W.; Stephens, G.L. MEDI-563, a humanized anti-IL-5 receptor alpha mAb with enhanced antibody-dependent cell-mediated cytotoxicity function. J. Allergy Clin. Immunol. 2010, 125, 1344–1353. [Google Scholar] [CrossRef] [PubMed]
  52. Nair, P.; Wenzel, S.; Rabe, K.F.; Bourdin, A.; Lugogo, N.L.; Kuna, P.; Barker, P.; Sproule, S.; Ponnarambil, S.; Goldamn, M.; et al. Oral glucocorticoid–sparing effect of benralizumab in severe asthma. N. Engl. J. Med. 2017, 376, 2448–2458. [Google Scholar] [CrossRef] [PubMed]
  53. Menzies-Gow, A.; Gurnell, M.; Heaney, L.G.; Corren, J.; Bel, E.H.; Maspero, J.; Harrison, T.; Jackson, D.J.; Price, D.; Lugogo, N.; et al. Oral corticosteroid elimination via a personalised reduction algorithm in adults with severe, eosinophilic asthma treated with benralizumab (PONENTE): A multicentre, open-label, single-arm study. Lancet Respir. Med. 2022, 10, 47–58. [Google Scholar] [CrossRef] [PubMed]
  54. Rabe, K.F.; Nair, P.; Brusselle, G.; Maspero, J.F.; Castro, M.; Sher, L.; Zhu, H.; Hamilton, J.D.; Swanson, B.N.; Khan, A.; et al. Efficacy and safety of dupilumab in glucocorticoid-dependent severe asthma. N. Engl. J. Med. 2018, 378, 2475–2485. [Google Scholar] [CrossRef] [PubMed]
  55. Kavanagh, J.E.; Hearn, A.P.; Jackson, D.J. A pragmatic guide to choosing biologic therapies in severe asthma. Breathe 2021, 17, 210144. [Google Scholar] [CrossRef]
  56. Ziegler, S.F.; Roan, F.; Bell, B.D.; Stoklasek, T.A.; Kitajima, M.; Han, H. The biology of thymic stromal lymphopoietin (TSLP). Adv. Pharmacol. 2013, 66, 129–155. [Google Scholar]
  57. Ying, S.; O’Connor, B.; Ratoff, J.; Meng, Q.; Mallett, K.; Cousins, D.; Robinson, D.; Zhang, G.; Lee, T.H.; Corrigan, C. Thymic stromal lymphopoietin expression is increased in asthmatic airways and correlates with expression of Th2-attracting chemokines and disease severity. J. Immunol. 2005, 174, 8183–8190. [Google Scholar] [CrossRef]
  58. Wechsler, M.E.; Colice, G.; Griffiths, J.M.; Almqvist, G.; Skärby, T.; Piechowiak, T.; Kaur, P.; Bowen, K.; Hellqvist, A.; Mo, M.; et al. SOURCE: A phase 3, multicentre, randomized, double-blind, placebo-controlled, parallel group trial to evaluate the efficacy and safety of tezepelumab in reducing oral corticosteroid use in adults with oral corticosteroid dependent asthma. Respir. Res. 2020, 21, 264. [Google Scholar] [CrossRef]
  59. AstraZeneca. Update on SOURCE Phase III Trial for Tezepelumab in Patients with Severe, Oral Corticosteroid-Dependent Asthma. Updated: 22 December 2020. Available online: https://www.astrazeneca.com/media-centre/press-releases/2020/update-on-source-phase-iii-trial-for-tezepelumab-in-patients-with-severe-oral-corticosteroid-dependent-asthma.html (accessed on 14 September 2021).
  60. MA, Y.; Qiu, S.; Zhou, R. Osteoporosis in Patients With Respiratory Diseases. Front. Physiol. 2022, 13, 939253. [Google Scholar] [CrossRef]
  61. Naranjo Hernández, A.; Díaz Del Campo Fontecha, P.; Aguado Acín, M.P.; Arboleya Rodríguez, L.; Casado Burgos, E.; Castañeda, S.; Arestè, J.F.; Gifre, L.; Gòmez Vaquero, C.; Rodrìguez, G.C.; et al. Recommendations by the Spanish society of rheumatology on osteoporosis. Reumatol. Clin. Engl. 2019, 15, 188–210. [Google Scholar] [CrossRef]
  62. McDowell, P.J.; Stone, J.H.; Zhang, Y.; Honeyford, K.; Dunn, L.; Logan, R.J.; Butler, C.A.; McGarvey, L.P.A.; Heaney, L.G. Quantification of glucocorticoid-associated morbidity in severe asthma using the Glucocorticoid Toxicity Index. J. Allergy Clin. Immunol. Pract. 2021, 9, 365–372. [Google Scholar] [CrossRef] [PubMed]
  63. Portacci, A.; Dragonieri, S.; Carpagnano, G.E. Super-Responders to Biologic Treatment in Type 2-High Severe Asthma: Passing Fad or a Meaningful Phenotype? J. Allergy Clin. Immunol. Pract. 2023, 11, 1417–1420. [Google Scholar] [CrossRef] [PubMed]
  64. Caminati, M.; Vaia, R.; Furci, F.; Guarnieri, G.; Senna, G. Uncontrolled Asthma: Unmet Needs in the Management of Patients. J Asthma. Allergy 2021, 14, 457–466. [Google Scholar] [CrossRef] [PubMed]
  65. Cutroneo, P.M.; Arzeton, E.; Furci, F.; Scapini, F.; Bulzomì, M.; Luxi, N.; Caminati, M.; Senna, G.; Moretti, U.; Trifirò, G. Safety of Biological Therapies for Severe Asthma: An Analysis of Suspected Adverse Reactions Reported in the WHO Pharmacovigilance Database. BioDrugs 2024, 38, 425–448. [Google Scholar] [CrossRef]
  66. Canonica, G.W.; Blasi, F.; Paggiaro, P.; Senna, G.; Passalacqua, G.; Spanevello, A.; Aliberti, S.; Bagnasco, D.; Bonavia, M.; Bonini, M.; et al. Oral CorticoSteroid sparing with biologics in severe asthma: A remark of the Severe Asthma Network in Italy (SANI). World Allergy Organ. J. 2020, 13, 100464. [Google Scholar] [CrossRef]
  67. Perlato, M.; Mecheri, V.; Vivarelli, E.; Accinno, M.; Vultaggio, A.; Matucci, A. “Patient remodeling” as a consequence of uncontrolled and prolonged OCS use in severe asthma: How biologic therapy can reverse a dangerous trend. J. Asthma. 2024, 61, 72–75. [Google Scholar] [CrossRef]
  68. McDonald, V.M.; Harrington, J.; Clark, V.L.; Gibson, P.G. Multidisciplinary care in chronic airway diseases: The Newcastle model. ERJ Open Res. 2022, 8, 00215–02022. [Google Scholar] [CrossRef]
  69. Caminati, M.; Cegolon, L.; Bacchini, M.; Segala, N.; Dama, A.; Bovo, C.; Olivieri, B.; Furci, F.; Senna, G. The potential role of local pharmacies to assess asthma control: An Italian cross-sectional study. BMC Public Health 2021, 21, 19. [Google Scholar] [CrossRef]
  70. Denton, E.; Lee, J.; Tay, T.; Radhakrishna, N.; Hore-Lacy, F.; Mackay, A.; Hoy, R.; Dabscheck, E.; O’Hehir, R.E.; Hew, M. Systematic assessment for difficult and severe asthma improves outcomes and halves oral corticosteroid burden independent of monoclonal biologic use. J. Allergy Clin. Immunol. Pract. 2020, 8, 1616–1624. [Google Scholar] [CrossRef]
  71. Clark, V.L.; Gibson, P.G.; Genn, G.; Hiles, S.A.; Pavord, I.D.; McDonald, V.M. Multidimensional assessment of severe asthma: A systematic review and meta-analysis. Respirology 2017, 22, 1262–1275. [Google Scholar] [CrossRef]
Figure 1. Flow chart for severe asthma management.
Figure 1. Flow chart for severe asthma management.
Biomedicines 13 00197 g001
Figure 2. The role of IL-31 in bone remodeling. IL-31, mainly produced by T helper 2 memory cells (Th2 CD45RO+), promotes osteoclastogenesis by inducing the differentiation of osteoclast progenitors (OCPs) into mature osteoclasts (OCs). IL-31, in synergy with reactive oxygen species (ROS), also stimulates osteoclastogenesis indirectly by inducing antigen-presenting cells (APCs), monocytes, and T helper 1 and 17 lymphocytes (Th1/Th17) to produce chemokines, metalloproteinases, and inflammatory cytokines, which in turn increase the production of IL-31. All of these events lead to the development of osteoporosis.
Figure 2. The role of IL-31 in bone remodeling. IL-31, mainly produced by T helper 2 memory cells (Th2 CD45RO+), promotes osteoclastogenesis by inducing the differentiation of osteoclast progenitors (OCPs) into mature osteoclasts (OCs). IL-31, in synergy with reactive oxygen species (ROS), also stimulates osteoclastogenesis indirectly by inducing antigen-presenting cells (APCs), monocytes, and T helper 1 and 17 lymphocytes (Th1/Th17) to produce chemokines, metalloproteinases, and inflammatory cytokines, which in turn increase the production of IL-31. All of these events lead to the development of osteoporosis.
Biomedicines 13 00197 g002
Figure 3. The role of IL-33 in bone remodeling. After inflammatory stimulation, the stromal cells produce IL-33, which directly blocks osteoclast (OC) formation from hemopoietic myeloid cells (HMCs), shifting the osteoclast precursor differentiation towards cytokine-producing macrophages and antigen-presenting cells (APCs). Damaged osteocytes (OCys), in addition to inducing inflammation, also release IL-33, which in turn stimulates the mineralization of the bone matrix. IL-33 acts directly by stimulating mature osteoblasts (OBs) and their bone marrow progenitors, the mesenchymal stem cells (MSCs), inducing their differentiation and maturation. Despite the production of the receptor activator of nuclear factor kappa-Β ligand (RANKL) and the partial blockage of the production of osteoprotegerin (OPG) by activated osteoblasts, the prevalent effect of IL-33 on the skeleton is anti-osteoporotic. Arrows with a “forbidden” signal mean that il-33 has anti-osteoporotic activities.
Figure 3. The role of IL-33 in bone remodeling. After inflammatory stimulation, the stromal cells produce IL-33, which directly blocks osteoclast (OC) formation from hemopoietic myeloid cells (HMCs), shifting the osteoclast precursor differentiation towards cytokine-producing macrophages and antigen-presenting cells (APCs). Damaged osteocytes (OCys), in addition to inducing inflammation, also release IL-33, which in turn stimulates the mineralization of the bone matrix. IL-33 acts directly by stimulating mature osteoblasts (OBs) and their bone marrow progenitors, the mesenchymal stem cells (MSCs), inducing their differentiation and maturation. Despite the production of the receptor activator of nuclear factor kappa-Β ligand (RANKL) and the partial blockage of the production of osteoprotegerin (OPG) by activated osteoblasts, the prevalent effect of IL-33 on the skeleton is anti-osteoporotic. Arrows with a “forbidden” signal mean that il-33 has anti-osteoporotic activities.
Biomedicines 13 00197 g003
Table 1. Corticosteroid conversion table drawn up from the available literature with clinical studies of sufficiently homogeneous enrolled populations.
Table 1. Corticosteroid conversion table drawn up from the available literature with clinical studies of sufficiently homogeneous enrolled populations.
Oral Corticosteroids
(OCS)
Approximate Equivalent Dose
(mg)
Cortisone25 mg
Hydrocortisone20 mg
Deflazacort7.5 mg
Prednisolone5 mg
Prednisone5 mg
Methylprednisolone4 mg
Triamcinolone4 mg
Betamethasone0.75 mg
Dexamethasone0.75 mg
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Furci, F.; Scaramozzino, M.U.; Talesa, G.R.; Pelaia, C. Connections and Unmet Needs: Severe Asthma Biologics and Osteoporosis. Biomedicines 2025, 13, 197. https://doi.org/10.3390/biomedicines13010197

AMA Style

Furci F, Scaramozzino MU, Talesa GR, Pelaia C. Connections and Unmet Needs: Severe Asthma Biologics and Osteoporosis. Biomedicines. 2025; 13(1):197. https://doi.org/10.3390/biomedicines13010197

Chicago/Turabian Style

Furci, Fabiana, Marco Umberto Scaramozzino, Giuseppe Rocco Talesa, and Corrado Pelaia. 2025. "Connections and Unmet Needs: Severe Asthma Biologics and Osteoporosis" Biomedicines 13, no. 1: 197. https://doi.org/10.3390/biomedicines13010197

APA Style

Furci, F., Scaramozzino, M. U., Talesa, G. R., & Pelaia, C. (2025). Connections and Unmet Needs: Severe Asthma Biologics and Osteoporosis. Biomedicines, 13(1), 197. https://doi.org/10.3390/biomedicines13010197

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

Article Metrics

Back to TopTop