Autor : Saldarini, Fernando1, Litewka, Diego2, Pascansky, Daniel3, Sivori, Martin3
1Pulmonology Section, Hospital Gral. Agudos “Donación Francisco Santojanni”, Buenos Aires. 2Pulmonology Service, Hospital Gral. Agudos “Dr. Juan A. Fernández”, Buenos Aires. 3Pulmonology University Center, “Dr.J.M.Ramos Mejía”, Faculty of Medicine, University of Buenos Aires, Pulmonology and Tisiology Unit, Hospital Gral. Agudos “Dr.José María Ramos Mejía”, Buenos Aires.
https://doi.org/10.56538/ramr.UVKK6634
Correspondencia : Martín Sivori: Urquiza 609, CABA (Autonomous City of Buenos Aires). E-mail: sivorimartin@yahoo.com
ABSTRACT
Monoclonal antibodies for the
treatment of severe asthma with a T2-high phenotype have been a significant
therapeutic breakthrough, improving patients’ quality of life and reducing
severe exacerbations. However, certain aspects remain controversial. The following
were chosen to be addressed, in the form of questions: is airway remodeling
possible with biologics?; are anti-biologic antibodies
a concern?; is complete remission achievable?; when, how, and to which biologic
should a “switch” be made?; can biologics be discontinued?; what approach
should be taken with a patient who wishes to become pregnant or is already
pregnant? and what about lactation? how
is adherence to biologic treatments like? Each question was answered based on
the review of the scientific evidence of each one.
There is not enough evidence to
confirm if biologics prevent long-term airway remodeling, or if they are safe
during pregnancy or lactation (with the exception of omalizumab). Currently,
there are no established criteria for the concept of complete remission with
biologics. Some proposed criteria include the absence of symptoms and
exacerbations, not using oral corticosteroids, normal spirometry, suppression
of T2 inflammation, and control of comorbidities. However, further research is
needed to validate these criteria. Once control is achieved, biologics should
not be discontinued. However, it is possible to consider de –escalating other
second– or third-line medications, while maintaining regular treatment with
inhaled corticosteroids + LABAs (long-acting beta-2 adrenergic bronchodilators)
at the lowest possible dose.
Key words: Severe asthma, T2-high phenotype, Biologics, Remission, Pregnancy,
Adherence
RESUMEN
Los anticuerpos monoclonales para el tratamiento del asma
grave fenotipo T2 alto, han sido un gran avance terapéutico, mejorando
la calidad de la vida del paciente y reduciendo las exacerbaciones severas.
Sin embargo, persisten aspectos de controversia. Sobre ellos, en formato de
preguntas, se eligieron: ¿es posible el remodelamiento de la vía
aérea con los biológicos?; ¿son un problema los anticuerpos
anti-biológicos?; ¿ae puede alcanzar la remisión completa?;
¿cuándo, cómo y a cuál biológico hacer el “switch”?;
¿se pueden discontinuar?;¿qué conducta se debe
tomar en una paciente que desea embarazarse, o lo está? ¿y con respecto a la lactancia?; ¿cómo es la
adherencia con los biológicos?. Se respondieron las preguntas en base a
la revisión de la evidencia científica de cada cada pregunta.
No existe suficiente evidencia para asegurar que se evita
el remodelamiento de la vía aérea a largo plazo con los
biológicos, así como su seguridad en el embarazo o lactancia (a
excepción del omalizumab). A la fecha, no hay criterios establecidos
para el concepto de remisión completa con biológicos. Se han
propuesto algunos criterios como ausencia de síntomas y exacerbaciones y
no uso de corticoides orales, espirometría normal, supresión de
la inflamación T2 y control de las comorbilidades. Pero para validarlos
aún se requieren más investigaciones. Una vez alcanzado el
control no se debe suspender el biológico, pero si se puede evaluar
desescalar los otros medicamentos de 2° o 3° línea, pero manteniendo
siempre el tratamiento regular con corticoides inhalados + LABA, a la menor
dosis posible.
Palabras Claves: Asma grave, Fenotipo T2 alto, Biológicos, Remisión, Embarazo,
Adherencia
Received: 03/28/2024
Accepted: 9/12/2024
Asthma is a heterogeneous,
inflammatory airway disease characterized by recurrent episodes of
bronchospasm, bronchial hyperreactivity, and increased bronchial secretions.1 It affects approximately 340 million people worldwide, with
a significant heterogeneity in terms of prevalence across Latin America,
ranging from 5% to 24%.2
In Argentina, it is estimated
that 9.36% of the population (approximately 2.5 million people) has asthma,
based on patient records from medical diagnoses.3
Severe asthma accounts for 3% to 5% of the asthma population,
though it has different epidemiological characteristics in Latin America.1,4 Severe asthma
is characterized by persistent symptoms, increased emergency visits, unplanned
outpatient appointments, higher rates of hospitalization, and greater use of
rescue medications, systemic corticosteroids, antibiotics, and high doses of
controller medications (inhaled corticosteroids, long-acting beta-agonists,
long-acting anticholinergics, and in some cases, leukotriene inhibitors). This
leads to a significant impact on the use of healthcare resources and an
increase in mortality rates.1,5-7 The evaluation carried out in this group of
patients for the purpose of classifying them as severe poses a challenge that
requires considering some variables, such as ensuring good adherence, proper
use of inhalers, and the presence or absence of comorbidities. Patients with
severe asthma bear a heavy disease burden, including impacts on their
well-being, social life, mental health, and adverse effects.1,5-7 The concept
of difficult-to-control asthma (DCA) focuses on ruling out these variables to
clearly define severe asthma. 1,5-8
Severe asthma represents a
heterogeneous syndrome with multiple clinical variants. Over the past two
decades, it has been intensely studied, and different phenotypes have been
defined.9-13 Establishing
the asthma phenotype in patients with severe uncontrolled asthma is part of the
diagnosis and evaluation of these individuals, since it can lead to
differential treatment and have prognostic implications.6
Two inflammatory phenotypic patterns have been defined: T2-high
(present in allergic and eosinophilic asthma) and non-T2, also called T2-low.5-7 Both T2-high
phenotypes often show some degree of overlapping. The fraction of exhaled
nitric oxide (FeNO), eosinophilia, and IgE are good biomarkers for the T2-high
phenotype.5-7 The
T2-allergic phenotype represents 40-50% of severe asthma and has an atopic
basis orchestrated by the activation of T helper type 2 cells (Th2), the
production of interleukins IL-4, IL-5, and IL-13, and isotype switching in B
lymphocytes towards IgE production.5-7 The
T2-eosinophilic phenotype represents more than 25% of severe asthma and is
characterized by the presence of eosinophils in bronchial biopsies and sputum.
It may be associated with chronic rhinosinusitis and nasal polyps.5-7 Severe T2-low
asthma is characterized by low levels of peripheral blood and sputum
eosinophils, a paucigranulocytic or neutrophilic profile, low FeNO levels, and
a poor response to glucocorticoids. In some cases, it is associated with chronic
airflow limitation, air trapping, and a history of smoking.5-7
Over the past twenty years,
biologics have been developed which now play a central role in the management
of severe T2-high asthma, offering a very acceptable efficacy/safety profile.
The first monoclonal antibody developed for the allergic IgE-mediated phenotype
was omalizumab, which was approved by the FDA (Food and Drug Administration)
in 2003 and by the EMEA (European Medicines Agency) in 2005, and it is widely
used in our country.14 Subsequently,
other biologics targeting eosinophilic responses in patients with severe asthma
were developed, including inhibitors of IL-4, IL-5, and IL-13.15-18 Mepolizumab and reslizumab are IL-5
inhibitors. Benralizumab inhibits the IL-5 receptor α. Dupilumab inhibits the IL-4 receptor α subunit, interfering with the actions of both IL-4
and IL-13.15-18 Mepolizumab,
benralizumab, and dupilumab are commercially available in our country.
Dupilumab has been developed for the T2-high phenotype and is the only new
biologic that does not include eosinophilia. Tezepelumab is the first antibody
to inhibit the thymic stromal lymphopoietin (TSLP), an alarmin cytokine
originally identified as a lymphocyte growth factor. It binds to the TSLP
receptor complex (including the IL-7 receptor α) and activates T-helper 2 lymphocytes, along with a
wide variety of immune and non-immune cells.19
It is being marketed in many developed countries, but not yet in
our country.20-22 While various
molecules are under clinical development to treat patients with the T2-low
phenotype, none of them has yet progressed to phase III clinical development
stages, so none of them is commercially available.
Despite the growing use of
biologics in severe asthma for the T2-high phenotype, many critical and
challenging aspects persist, where scientific evidence is still limited, and
questions remain unanswered. The objective of this review is to critically
analyze the most important aspects of the biologics available in our country in
the form of questions, based on a review of the published scientific evidence.
Basing on their daily practice managing patients with severe asthma with the
T2-high phenotype, the authors have selected the most relevant questions to ask
on a daily basis:
1. Is airway remodeling possible
with the treatment with biologics in severe asthma?
2. Are anti-biologic antibodies a
concern?
3. Is complete remission
achievable with biologics?
4. When, how, and to which
biologic should a “switch” be made? Should biologics be discontinued?
5. What approach should be taken
with a patient who wishes to become pregnant or is already pregnant? And what about lactation?
6. How is adherence to treatment
and biologics like?
Methodology
Literature search was conducted
in the MEDLINE, EMBASE, Cochrane, SciELO, and Lilacs databases until January
31, 2024, using search terms relevant to the respective questions.
1. Is airway remodeling possible with the treatment with biologics in
severe asthma?
The consequence of persistent
airway inflammation in asthma is the response to diminished innate immunity
and to environmental factors. The common final pathway is the development of
chronic airflow obstruction due to airway wall remodeling and alterations in
biomechanical properties, accompanied by mucus plugs and closure of small
airways.23-25 Structural
cells, such as epithelial cells and bronchial smooth muscle cells contribute to
the inflammatory response, mediated by chemokines, cytokines, and growth
factors.23-25 Epithelial
damage or stress (represented by the increase in epidermal growth factor
receptor) triggers cellular activation and releases pro-angiogenic factors
(which promote neovascularization) and growth factors such as transforming growth
factor beta (TGF-β) and vascular
endothelial growth factor (VEGF). These factors activate subepithelial
mesenchymal cells, leading to the proliferation of the extracellular matrix and
fibroblasts.23-25 However, epithelial cells also release chemokines such as
IL-8, which is a chemoattractant for neutrophils. The bronchial wall is thicker
in severe asthma compared to mild asthma, as is the mass of the bronchial
smooth muscle.23-25 The activation of the bronchial smooth muscle releases
chemotactic factors that recruit mast cells and myofibroblasts, as well as
extracellular matrix proteins and additional angiogenic factors. Mast cells
express Th2 cytokines such as IL-4 and IL-13.23-25 Fibrocytes also infiltrate the bronchial wall
in the most severe forms of asthma. Neutrophils and mast cells located in the
glands are associated with mucus plugs in fatal asthma. The thickening of the
reticular basement membrane is caused by the extracellular matrix deposition
and is a marker of remodeling. Finally, fibrosis contributes to fixed airflow
obstruction in severe asthma.23-25
Both airway remodeling and
inflammation can begin in childhood (ages 2 to 4) and persist through school
age, eventually differentiating in adulthood where inflammation continues, and
remodeling remains stable. The implications of this persistence over time in
asymptomatic patients, and whether this temporal evolution is
independent or not, remain unknown.23-25
Interestingly, biologics have
been shown to reduce some characteristics of airway remodeling. The first
study evaluated the effect of mepolizumab in 24 atopic patients, showing a
significant reduction in extracellular matrix proteins, thus suggesting an
effect of eosinophils on TGF-β1.26 At the
European Respiratory Society Congress 2023, preliminary results were presented
from the MESILICO study, where after one year, patients with late-onset severe
asthma experienced a reduction in the thickness of the basement membrane.27 Benralizumab
has been shown to reduce the smooth muscle mass of the bronchial wall.28 Similarly, 13
severe allergic patients treated with omalizumab achieved a 5% reduction in the
bronchial wall area.29 Finally, with
tezepelumab, no effect has been determined on basement membrane thickness or
epithelial integrity.30
2. Are anti-biologic antibodies a concern?
The incidence of immunogenicity
from anti-biologic antibodies (ABAs) and their potential deleterious impact on
the efficacy and safety of biologics has been poorly studied.31
In theory, biologics could be more immunogenic than the small
molecules of drugs, as they may directly block the receptors (acting as
neutralizing antibodies) and/or increase their clearance via the
reticuloendothelial system, or even form autoimmune complexes.31
Most of the available information on ABAs comes from studies on
tumor necrosis factor inhibitors in patients with rheumatoid arthritis, where
they have been shown to decrease circulating drug levels and worsen clinical
outcomes. However, there is limited data on the use of biologics in asthma.
Chen et al conducted a systematic review and meta-analysis of 46 clinical
studies, reporting an overall incidence of 2.91% (95% CI [confidence interval],
1.60–4.55). The incidence of ABAs in patients treated with benralizumab was
8.35%; dupilumab, 7.61%; reslizumab, 4.39%; mepolizumab, 3.63%; tezepelumab,
1.12%; and omalizumab, 0%.31 It has been
determined that subcutaneous administration, lower doses, and longer intervals
between doses are associated with a higher incidence of ABAs.31
In conclusion, since ABAs are not
currently available in routine practice, future studies will need to be carried
out in order to determine their clinical impact on the management of patients
with severe asthma.
3. Is complete remission achievable with biologics?
Asthma remission is defined as
the reduction or disappearance of asthma symptoms and a decreased need for
medications to control it.1,8,32,33 In simple terms, it means that asthma is in an
inactive or well-controlled state, a concept that has gained renewed interest
with the advent of biologic treatments. Remission can occur for several
reasons. In some cases, it may result from lifestyle changes, such as avoiding
asthma triggers like allergens or irritants, maintaining a clean and smoke-free
environment, and following an appropriate treatment plan. A good treatment plan
should include the regular administration of asthma medications (commonly
inhaled glucocorticoids, with or without LABAs, and biologic therapies in cases
of severe asthma). This state is referred to as “clinical remission” and must
be sustained for at least twelve months (without symptoms, exacerbations, or
the use of systemic steroids, apart from having lung function tests with normal
results). On the other hand, the term “complete remission” is used in cases
where there is no evidence of bronchial hyperreactivity or bronchial
inflammation.1,6,32,33
Scientific evidence on asthma
remission in both adults and children comes from clinical studies and long-term
observations. These studies have shown that a combination of the appropriate
medical treatment, the management of triggering factors, and adherence to a
personalized care plan can lead to asthma remission in many individuals.
It is important to note that
remission does not mean a definitive cure for asthma. In some cases, symptoms
may reappear in the future, especially if preventive measures and proper asthma
management are not sustained. Therefore, even during remission, it is
essential to continue with a regular medical follow-up and maintain a healthy
lifestyle to prevent relapses.2
Patients with severe asthma bear
a heavy disease burden, including impacts on their well-being, social life,
mental health, and adverse effects.34 Cohort
studies indicate that approximately 50% of patients with severe asthma have
been treated with systemic corticosteroids2
over extended periods, despite the well-documented adverse
reactions.35
In 2020, an expert consensus
defined asthma remission as the cases of patients without any symptoms, with no
exacerbations, no use of corticosteroids, and improved forced expiratory
volume in one second (FEV1),
agreed upon by both the patient and physician, for at least twelve months. This
was termed clinical remission without treatment.33
Baseline factors may predict which patients are more likely to
achieve remission, as for example shorter disease duration, better lung
function, greater asthma control, and earlier age of disease onset.34 A
“super-response” has been observed in approximately one-third of severe asthma
patients treated with biologics, which is likely a precursor to achieving
remission.36
Remission has become the primary
treatment goal for other conditions like rheumatological diseases and
inflammatory bowel disease. However, in asthma, the concept of remission is
still under development (TABLE 1).33 Recently, asthma remission has been evaluated in a post-hoc
analysis of both clinical trials and clinical cohorts, aiming to identify its
prevalence in patients receiving biologics (TABLE 2). 37
Between 15% and 37% of the patients achieved remission, though
definitions of remission were made independently across studies, leading to
heterogeneity.37 Elevated
eosinophil levels and nitric oxide levels were identified as important
predictors of remission.37 Notably, a significant proportion of patients in the placebo
arm achieved remission with medium to high doses of inhaled corticosteroids.
This shows the anti-inflammatory role of inhaled corticosteroids and their
impact on disease remission. It is important to say that there are studies
where 20% of children with asthma experience spontaneous remission.37
In theory, failing to achieve
early clinical remission may reduce the likelihood of achieving long-term
remission.38 A post-hoc
analysis of the QUEST and TRAVERSE clinical trials demonstrated that at week 52
of the QUEST, 31.7% of patients treated with dupilumab versus 17.7% of those on
placebo achieved clinical remission, whereas at week 48 of the TRAVERSE
(patients were included in this study once they had completed the QUEST
study), clinical remission rates were 36.4% for patients treated with
dupilumab/dupilumab and 29.6% for those in the placebo/dupilumab group (placebo
in the first trial and dupilumab in the second).38
This could reflect a “missed opportunity” in the second study
arm, as the magnitude of the remission was lower.38
There are significant challenges
in the clinical assessment of complete asthma remission. Measuring bronchial
hyperreactivity is laborious and contraindicated in patients with severe
bronchial obstruction. Direct evaluation of airway inflammation through
procedures such as bronchoscopy or induced sputum analysis is complex and not
widely available.39 Some patients
may have fixed airway obstruction that does not improve with treatment, and
their symptoms can be heavily influenced by comorbidities associated with
asthma.39 We are still
in the early stages of developing this concept, and more evidence is needed to
support it.39 In the
meantime, adopting a therapeutic strategy aimed at modifying the natural course
of asthma seems a reasonable approach.
Is remission a realistic goal for the future care of asthma, or are we
setting the bar too high?
Achieving remission seems to be a
reasonable expectation in asthma because spontaneous remission, commonly
referred to as “outgrown” asthma, is considered a frequent occurrence in
children with asthma. Studies show that between 5% and 69% of children with
asthma experience spontaneous remission during adolescence or adulthood.40 The wide range of remission rates among these study
populations reflects differences in the criteria used to define asthma and
remission, including asthma severity, the age of disease onset, and the
duration of the follow-up period. When applying the strict criteria for
remission mentioned earlier, the proportion of patients achieving complete remission
is small.33 Only 20% of
children with well-documented mild to moderate asthma achieved complete
remission by the age of 23.40 Adults with
asthma, (with childhood- or adulthood- onset disease) who have been followed
over time may show even lower rates of spontaneous remission or complete
disease remission. Spontaneous remission does exist, but it is collectively
thought that complete remission without relapse is uncommon.
It is unlikely that individuals
with moderate to severe asthma, frequent exacerbations, poor lung function, and
heightened inflammation driving their disease will experience spontaneous remission.
This more symptomatic population is the one that most urgently needs long-term
disease modification.
Biologic drugs targeting type 2
pathways reduce exacerbations, hospitalizations, and corticosteroid use while
improving lung function and the patient’s quality of life.17
A future goal would be that reducing
specific inflammatory pathways could enhance the average airway caliber and
modify the course of the disease. Real-world studies show that after the
withdrawal of omalizumab or mepolizumab in long-term treated subjects, more
than half experienced exacerbations within the following year, similar to
control subjects.41
Overall, studies to date suggest
that discontinuing biologics –even after long-term use– in a large proportion
of patients with severe asthma does not reverse the inflammatory mechanisms
driving the disease.
How will we know if the disease has subsided or entered remission?
If we continued administering
biologics and inhaled preventive medications on a standard schedule, we could
not measure the success or identify the characteristics associated with a
positive response. Only through careful tapering of preventive medications
could we identify this small subset of responders. We should advocate for
adopting a personalized tapering approach. Our goal should be good control with
the least amount of medication. As we adjust biologic therapies, we may see
that some patients can reduce their use of associated preventive medications.
Clinical trials on treatment de-escalation will need to be conducted to test
these hypotheses. New biologics in clinical development, such as those
targeting epithelial inflammatory signaling, may more permanently reduce the
inflammatory signals that drive asthma, and their clinical impact will need to
be determined.
As a physician, how does the definition of asthma remission affect the
care you provide to each patient?
The short answer is: it
shouldn’t. This definition is intended solely for clinical research purposes
and should not be applied to individual patients, nor should it be used to
authorize or withdraw particular therapies. In fact, although asthma guidelines
continue to evolve, they still do not include remission as a treatment outcome.35 Therefore,
the goal of the treatment should remain focused on reducing the risk and
achieving sustained control, rather than pursuing remission. From a clinical
perspective, the relationship between control and remission is not known yet.
It is worth noting that the discussed definition of “remission” is a consensus
statement from a group of experts.5 There are no clinical studies demonstrating if this
definition leads to better patient outcomes. This definition serves as a
starting point to compare therapies, but it will also refine the definition of
remission in future studies.42 Someday, as
with other chronic diseases, we may refer to patients whose asthma has entered
remission as a real clinical outcome, one that may not represent a cure but can
be controlled or even be in a state of dormancy with appropriate treatments.42 In fact,
until more studies are conducted, we will not even know the proportion of adequately
treated asthma patients who could meet the criteria for this definition of
remission. Therefore, as a physician, it is important to be aware of these
discussions and the potential utility of a remission definition, but it is too
early to apply this concept to clinical practice.
4. When, how, and to which biologic should a “switch” be made?
Should biologics be discontinued?
Discontinuation of omalizumab therapy
The discontinuation of omalizumab
has been evaluated using real-world data.43
Molimard et al described data from 61 responder patients who
discontinued omalizumab after a mean treatment
duration of 22.7 ± 13.1 months. They found that 55% of patients lost control of
the disease after a median interval of 13.0 months (mean 20.4 ± 2.6 months).44 A recent
study using the French health system database followed 19,203 patients over
more than 10 years. It showed that among patients with controlled asthma who
discontinued omalizumab for at least 16 weeks, 70%, 39%, and 24% remained
controlled without restarting omalizumab 1, 2, and 3 years after
discontinuation.45 Currently, further studies are needed to define the criteria
for discontinuing omalizumab therapy.
Switching from omalizumab to other biologics
With the development of other
biologics, several real-world studies have addressed this issue.46-48 In one study,
145 patients on omalizumab who experienced more than two asthma exacerbations
per year were switched to mepolizumab for 32 weeks.46
The study demonstrated improvements in asthma control,
exacerbation rates, and lung function. Similarly, another study examined
patients with uncontrolled eosinophilic asthma who were receiving omalizumab
and were switched to benralizumab.48 These
patients showed improvements in their evolution in terms of exacerbation rates,
lung function, IgE levels, FeNO, and eosinophil counts.47-48 In another study, the most frequent switch was
from omalizumab to an anti-IL-5 therapy (49.6%), primarily due to low efficacy
and/ or adverse effects, in a real-world study involving 3,531 asthma patients.49
To conclude, studies related to
omalizumab switching have predominantly been conducted in patients with
eosinophilic asthma, where anti-IL-5 therapy offers clear advantages. Consequently,
prospective studies with structured switching criteria are needed in order to
identify patients who could benefit from a broader spectrum of options.
Anti-IL-5 therapy
Switching from omalizumab to anti-IL-5 therapies
Several studies evaluated the
switch to mepolizumab from omalizumab. The OSMO study assessed the change in
asthma control in patients with blood eosinophil counts greater than 150 cells/mL who did not achieve optimal control with
omalizumab.50 Exacerbations
decreased from 3.26 to 1.18 events/year, after 32 weeks of treatment. The ACQ-5
(Asthma Control Questionnaire) and SGRQ (St. George’s Respiratory
Questionnaire) scores improved significantly, regardless of baseline
eosinophil levels, comorbidities, exacerbation history, or oral corticosteroid
use. Another retrospective study found that switching to mepolizumab reduced
the exacerbation rate, the proportion of patients dependent on oral
corticosteroids, and the number of workdays lost.51
A real-world study investigated
the switch from omalizumab to benralizumab.48
After one year of benralizumab
treatment, significant improvements were noted in exacerbation rates, ACT
(Asthma Control Test) scores, FEV1,
and SNOT-22 (Sino-Nasal Outcome Test-22) scores.48
Patients with severe eosinophilic
asthma who do not respond to omalizumab would benefit from switching to an
anti-IL-5 therapy. The primary endotype in these patients is thought to involve
eosinophilic inflammation not mediated by the IgE pathway.52
Switching to benralizumab from mepolizumab
In cases of insufficient
response, switching from an anti-IL-5 monoclonal antibody (mAb) to an
anti-IL-5Rα mAb has also been
studied.53 Out of 665
patients treated with anti-IL-5 mAb therapy, 60 switched to benralizumab. The
FEV1 improved
from 61% to 68%, and the ACT from 16 to 19 points. Additionally, two
retrospective studies demonstrated improvements in exacerbation rates, ACT
scores, FEV1,
quality of life (QoL), and oral corticosteroid doses after switching from
mepolizumab to benralizumab.54-55 Even though
benralizumab appears more effective than mepolizumab at reducing blood and
lung tissue eosinophils, no consistent differences in clinical efficacy have
been demonstrated between the two.55-58 But, if the
efficacy of mepolizumab were insufficient, switching to benralizumab would be
a validated option.53,59
This is based on the fact that
several differences have been demonstrated in the mechanism of action of these
molecules. Given that basophils express IL-5Rα, their depletion could be induced via
benralizumab-mediated cytotoxic activity, as suggested in in vitro studies
and supported by reductions in circulating basophils.59-60 Benralizumab also promotes macrophage-mediated
phagocytosis of eosinophils.61 In addition
to Th2 and ILC2 as major source of IL-13, eosinophils and basophils expressing
IL-5Rα also produce
functional IL-13.62-
64 By
depleting these cells that aren’t a significant source of IL-13 in patients
with elevated FeNO, benralizumab would show another difference between it and
mepolizumab.
Discontinuation of anti-IL-5 therapy
With regard to the
discontinuation of the anti- IL-5 therapy, the COMET study evaluated 155
patients who discontinued mepolizumab after at least three years of use, and
compared them to 144 patients who continued treatment. Patients who
discontinued anti IL-5 therapy showed a shorter time to first exacerbation,
loss of asthma control and increased eosinophil levels. However, cases of
severe exacerbations (requiring emergency care or hospitalization) were rare.41
Anti-IL-4/IL-13 therapy
Switching to dupilumab
There are many cases of switching
to dupilumab from other biologics, since it is the most recently approved
biologic drug.
In real-world studies from
France, the majority of patients who switched biologics (97%) moved from
mepolizumab or omalizumab to dupilumab.65
A study in Japan found that 60% of patients who switched to
dupilumab experienced significant reductions in exacerbations and decreased
need for oral corticosteroids.66 In Germany, a
study of 38 patients with severe asthma previously treated with anti-IL-5/IL-5Rα mAb or anti-IgE therapies without satisfactory
results were switched to dupilumab.67
76% of those patients achieved better symptom control, an
improved lung function, and reductions in FeNO, IgE levels, use of corticosteroids
and exacerbations 3 to 6 months after the switch. Patients with elevated FeNO
(>25 ppb) on prior biologic therapies showed greater responses compared to
those with non-elevated FeNO levels (<25 ppb). Blood eosinophil levels were
reported to increase in these studies switching to dupilumab, but this was not
associated with an increase in adverse effects.
To sum up, this therapeutic
approach is supported by published data regarding the switching of biologics
in approximately 1,527 patients with severe asthma. The data came from various
sources, as for example the information gathered from different clinical
trials (OSMO, post-hoc analyses of MENSA and SIRIUS, ANANKE), various
real-world studies and case reports.
Should other controller drugs be discontinued?
The current goal of asthma
treatment includes the concept of clinical remission, which could ideally lead
to discontinuing treatment. Recent consensus guidelines have proposed several
definitions of clinical remission, complete remission, and super-responders in
severe asthma patients.33 These definitions encompass several aspects of the disease,
such as symptoms, exacerbations, lung function, and inflammation. Several
studies have demonstrated that biological treatments attain many of these goals.68 For this
reason, it might be feasible to consider discontinuation of a biological
treatment. Several studies have been published on the discontinuation of
biologics in severe asthma, some of which have been discussed earlier.41,43-45,69 Most of these
studies report that after discontinuation, symptoms worsen, and the frequency
of exacerbations increases. However, in selected patients, this discontinuation
strategy might be feasible. For that purpose, it is important to identify the
characteristics of these patients. First, patients shouldn’t have any
exacerbations, and they should have adequate symptom control and stable lung
function with the biological treatment at the time treatment discontinuation is
considered. According to multiple reports, suppression of T2 inflammation is
necessary to achieve clinical remission.70
Similarly, comorbidities should also be
controlled. As mentioned earlier (TABLE 1), currently there are no established
criteria for the concept of complete remission free of biologics. However,
further research is still needed to validate these criteria. Regarding the
de-escalation of the rest of the baseline controller treatment once asthma
control (or remission) is achieved, the SHAMAL study shows that patients who
met clinical remission criteria during biological therapy face a risk of
accelerated lung function decline due to uncontrolled inflammation when
inhaled corticosteroids are discontinued as part of maintenance therapy.71 For this
reason, if treatment de-escalation is considered for these patients, it is
recommended that they discontinue only second- or third-line medications
(leukotriene receptor antagonists, LAMAs [long-acting muscarinic
antagonists]), while always maintaining regular baseline treatment with
inhaled corticosteroids + LABAs at the lowest possible dose.
5. What approach should be taken with a patient who wishes to become
pregnant or is already pregnant? And what about lactation?
Pregnancy and lactation are
generally significant concerns within the medical community, particularly due
to the need for medication and the potential teratogenic effects on the fetus.
It is now well established that
good asthma control has a very positive impact on pregnancy development, on the
fetus, and on childbirth.
However, managing severe asthma
remains a challenge for physicians, undoubtedly, both pregnancy and lactation
add complex issues that must be addressed through clear guidelines, approved
medications, and, in many cases, decisions should be jointly made and defined
by a multidisciplinary team, since there are limited studies of level A
evidence regarding the use of monoclonal antibodies (biologics) during
pregnancy. There are no specific clinical trials involving pregnant women;
human teratogens are usually identified through case reports. In the United
States, the frequency of congenital defects is approximately 3% of live births,
the stillbirth rate is 0.625%, preterm births account for 10.1%, and low birth
weight occurs in 8.24% of cases. However, it is unclear how much of this can be
solely attributed to medications.72
In the future, we will likely
have more concrete safety data, as emerging publications begin to explore the
treatment of asthma during pregnancy based on treatable traits.73
To establish practical
recommendations for pregnant women and women who are breastfeeding, general
observational studies are typically used. These include pregnancy registry
studies, cohort studies, case-control studies, and database analyses (including
data from adverse event reports).74 One of the
primary registries used for biological products is MotherToBaby (last updated
in November 2022 for biologics), which has ongoing studies limited to the U.S.
and Canada but currently lacks sufficient data for the scenarios previously
described.75 The Food and
Drug Administration (FDA) also maintains a list of open registries for various
medications, which is updated as new data and outcomes become available.76
Studies conducted in animals have
shown that most of the current biologics cross the placenta and are
concentrated in breast milk, but do not have adverse effects; however, these
findings remain experimental. Other published studies rely primarily on case
reports.
Based on the most recently
published data regarding safety during pregnancy from the first to the third
trimester, the following recommendations are proposed (Table 3).77
Omalizumab
The safety profile of omalizumab
is well-established, with data from more than 9,500 patients in clinical
trials and over 400,000 patient-years post-marketing. This has provided safety
data since its approval by the FDA in 2003 and the European Medicines Agency
(EMA) in 2005. Additionally, like other IgG molecules, omalizumab crosses the
placenta during the second and third trimesters of pregnancy.78
The observational study of the
use and safety of Xolair (omalizumab) during pregnancy (EXPECT), conducted in
the United States from 2006 to 2018, is the largest prospective study on this
topic. It reported results from 230 pregnant women with asthma exposed to
omalizumab either eight weeks before or at any time during pregnancy.79-80 The study
identified a congenital anomaly rate of 8.1%, a live birth rate of 99.1%, a
stillbirth rate of 0.9%, and a preterm birth rate of 15.0% in this cohort.79-80 The findings
from the registry were compared with a cohort of pregnant women with asthma who
had not been exposed to omalizumab. This comparison group, called the Quebec
External Comparator Cohort (QECC), included 1,153 women, matched by age and
disease severity.80 This
comparison showed that the live birth rate and the prevalence of major
congenital malformations were similar to those of the EXPECT sub-cohort.80 A higher rate
of low birth weight was found in babies born to patients who had been treated
with omalizumab (13.7% in EXPECT vs. 9.8% in QECC).80
However, 64.9% of women in the EXPECT sub-cohort had severe
asthma, compared to 21.2% in the QECC group, which makes it difficult to
determine whether the low birth weight was caused by the exposure to the
medication or the severity of the disease.80
There are other studies, primarily case reports, that didn’t find
any difference with regard to the data reported above.
Mepolizumab
The Mepolizumab Pregnancy
Exposure Study is a prospective, observational exposure cohort study that
investigates pregnancy outcomes in women exposed to mepolizumab during
pregnancy, in comparison with the results of women who didn’t use mepolizumab
during pregnancy but did use other asthma medications (the “treated disease”
comparison group), and the results of women exposed to non-teratogenic agents
(the “non-asthmatic” comparison group).82-83 This study was initiated after the approval
and marketing authorization of the European Medicines Agency (EMA). The
objective of the study is to monitor planned and unplanned pregnancies exposed
to mepolizumab and assess its potential teratogenic effects, focusing on major
congenital defects as the primary outcome, and the secondary outcomes,
including preterm birth, newborns who are small for the gestational age,
spontaneous abortion, and stillbirth.83
The study is conducted by the Research Center of the Organization
of Teratology Information Specialists (OTIS), located at the University of
California, San Diego.83 The target
sample size is: 200 women in the mepolizumab-exposed cohort, 300 women in the
treated disease cohort, and 300 women in the non-asthmatic cohort.83 The study is
expected to run for 6.5 years from the start of recruitment, which began in
2016-2017.76,82-83
Benralizumab
Like its predecessor, the
anti-IL-5 agent, benralizumab, which has been approved for use in 2017, is not
approved for use in pregnant women. There is only one ongoing study regarding
its safety in this group (ClinicalTrials.gov Identifier: NCT03794999)84. This
study began on March 20, 2019, and its estimated primary completion date is
February 27, 2026. Its primary objective is to monitor both planned and unplanned
pregnancies in order to assess potential teratogenic effects (congenital
defects) associated with exposure to benralizumab.84 This exposed group will be compared to two unexposed
reference groups. It is estimated that 800 pregnant women with asthma who have
been exposed to benralizumab at any time during their pregnancy or within 8
weeks prior to their last menstrual period will participate.84 As
control group, the study will include pregnant women with asthma who haven’t
been exposed to benralizumab during pregnancy or within the 8 weeks prior to
their last menstrual period, along with a third group of pregnant women without
an asthma diagnosis, who have not been exposed to any known human teratogen and
have not taken benralizumab during pregnancy. This study aims to provide
information about the potential risks associated with the use of benralizumab
during pregnancy and contribute to scientific knowledge regarding its safety in
this population.
Dupilumab
Currently, there are no specific
studies addressing the relationship between asthma and pregnancy-lactation.
However, research is underway in North America (USA-CANADA) focusing on
evaluating the safety of using dupilumab during pregnancy and its impact on
newborns. A notable example of these studies is the “Post-Authorization Safety
Study in North America” (ClinicalTrials.gov NCT04173442), which has been
ongoing since October 24, 2018, and is scheduled to conclude on July 9, 2026.85
This study is organized into three distinct cohorts: 1. Women who have used
dupilumab for an indication other than asthma or atopic dermatitis; 2. Those
who have been exposed to dupilumab within 10 weeks following the last menstrual
period or at any time during the current pregnancy; 3. Comparison cohort
without disease: women exposed to dupilumab within 10 weeks before the first
day of the last menstrual period, women diagnosed with any approved indication
for dupilumab, and those who have had the first contact with the study after
receiving a prenatal diagnosis of any major structural defect. The primary
purpose of this study is to provide valuable information about the safety of
dupilumab during pregnancy, with special attention to possible effects on
maternal health and child development.85
Tezepelumab-reslizumab
There are no safety studies
regarding pregnancy and lactation.86
Conclusions on the use of biologics during pregnancy and lactation
In the first place, it is
essential to strengthen the doctor-patient relationship to understand and plan
future pregnancies in patients with severe asthma who are receiving biologics.
It is important to understand the
benefit of having controlled asthma compared to uncontrolled asthma during
pregnancy and the risks associated with this situation.
While it is often difficult to
differentiate the side effects that occur in pregnant women and in the fetus in
this group of patients with severe asthma receiving various medications,
exacerbations may also occur alongside other comorbidities.
At present, given the practical
and ethical challenges of intervention studies in this patient group, we
emphasize the importance of basing the use of biologics on the limited
publications available. Currently, the only biologic considered safe for use
during pregnancy is omalizumab, and future results from ongoing studies are
still awaited for other biologics.
6. How is adherence to treatment and biologics like?
The WHO defined adherence in 2003
as “the degree to which a patient’s behavior, in terms of medicine-taking,
following a diet, or making lifestyle changes, corresponds with the recommendations
of the healthcare provider.”87
It is well known that
non-adherence is very common among asthma patients, and it is due to several
factors: psychosocial aspects of the patient, factors inherent to the disease
itself, doctor-patient relationship, and even access to medications, all of
which have been widely determined in several studies.88
Non-adherence has been reported in 30- 70% of patients. Busby et al determined
in patients with severe asthma that the factors associated with poor adherence
were the fact of belonging to ethnic minorities (OR 3.10, 95% CI 1.68–5.73) and
having two or more changes in preventive medication (OR 2.77, 95% CI 1.51–5.10).89
In the SAPPHIRE study, it was estimated that 75% adherence to inhaled
corticosteroids resulted in a 49% reduction in exacerbations.90 In
the different definitions of “poorly controlled asthma”, assessing adherence
problems and addressing them before labeling a patient as having severe asthma
is a mandatory step in the recommendations of different international and
national guidelines. Almost a quarter of the exacerbations and two-thirds of
asthma-related hospitalizations are linked to poor adherence to the preventive
treatment.90,91 It is assumed that patients enrolled in clinical
studies also adhered to the treatment. As per protocol, in clinical studies, it
is necessary to ensure treatment adherence more than 80% of the time, and this
is achieved not only through patient self-reporting of medication intake but
also by counting the doses of the drugs under investigation and using
electronic dosing devices during each visit or through telemedicine. Therefore,
probably better asthma control can be achieved in these patients solely by
improving adherence. As a common factor in almost all clinical development
studies of new biologics, a “clinical study effect” has been observed in the
placebo arms, in the reduction of severe exacerbations (between 30-50%),
improvement in FEV1 (less than 200 ml), asthma control, and quality of life
(both with improvements greater than the clinically significant minimal
differences).92 It is striking to see how the three studies investigating
biologics in corticosteroid-dependent patients (SIRIUS, ZONDA, and VENTURE)
achieved significant reductions in corticosteroid use (35-65% reduced the dose
by half), and even complete discontinuation (8-25%).92 The hypothetical
reasons for this result could be based on the idea that the patients enrolled
in the studies were probably undertreated or poorly adherent, with poor asthma
control; and it was observed in the study that by improving treatment adherence,
standardizing the treatment regimen, and ensuring proper follow-up, asthma
control was achieved, leading to the previously detailed results.92
In the placebo arms of phase III studies, the real impact of adherence has
been demonstrated.92 Ensuring good adherence is always the first
step for a patient with poorly controlled asthma, and improving it has a
beneficial impact on quality of life, asthma control, reduction of
exacerbations, and systemic corticosteroid use, and also saves healthcare
system resources.93
Pre-filled autoinjector devices (AIDs) and adherence
AIDs are simple to use,
practical, and enable effective home management with a low risk of critical
errors.94 Potential candidates include children, adolescents, and
adults of working age.94 Elderly patients with neurological,
psychiatric, or rheumatological conditions affecting the upper limbs may not
be suitable. Proper education of the patient is critical. The first dose should
be administered in a specialized center for severe asthma or in a physician’s
office. Instructions on timing, storage, and cleaning of the AID are essential
for successful use. A suitable doctor-patient relationship is essential.88,94 It is important to note that not all dosing
regimens for the biologics available in our country are the same. All are
administered subcutaneously. Omalizumab and mepolizumab are administered every
four weeks. Benralizumab is administered every four weeks for the first three
doses, and starting from the fourth dose, every eight weeks. Dupilumab is
administered every two weeks.
In Argentina, reslizumab is not
commercially available. Among the other four biologics, there are pre-filled
autoinjector devices that have improved treatment adherence. Their correct
administration and adherence have been confirmed by measuring the concentration
of the biologic in the blood in optimal levels.94-97
Adherence and FeNO suppression test
The FeNO Suppression Test (FST)
was described more than ten years ago to evaluate non-adherence to asthma
treatment.98 Using a device adapted to a
dry powder inhaler, data are recorded for one week simultaneously with the
inhalation of baseline preventive medication. A 42% decrease in FeNO between
the average values of days 0/1 and days 4/5 is considered positive.98,99
More recently, Couillard et al
have used the FST to identify adherent patients with T2 corticosteroid-resistant
phenotypes, determining that FeNO and eosinophils provide complementary and
different information: FeNO reflects airway inflammation, while eosinophilia
reflects systemic inflammation.100
Butler et al determined in a
study of 135 patients that those with a negative FST were more likely to be
treated with biologics (39 out of 54 patients, 72%) compared to those with a
positive FST (35 out of 81 patients, 43%, p=0.001). High maintenance doses of
corticosteroids (CS) and previous admission to the ICU were identified as
predictors. An increase in FeNO correlates with better asthma control and
adherence to high doses of ICS/LABA.101
CONCLUSIONS
Treatment with biologics for
patients with severe, uncontrolled T2-high phenotype asthma has been a major
advance in the last twenty years, with proven efficacy and safety, improving
the patient’s quality of life by reducing severe exacerbations and
hospitalizations. However, controversial aspects still exist. There is not
enough evidence to confirm if biologics prevent long-term airway remodeling, or
if they are safe during pregnancy or lactation (with the exception of
omalizumab). Currently, there are no established criteria for the concept of
complete remission with biologics. Some proposed criteria include the absence
of symptoms and exacerbations, not using oral corticosteroids, normal
spirometry, suppression of T2 inflammation, and control of comorbidities.
However, further research is needed to validate these criteria. Once control
is achieved, biologics should not be discontinued. However, it is possible to
consider de-escalating other second- or third-line medications (leukotriene
receptor antagonists, LAMAs), always maintaining regular base treatment with
inhaled corticosteroids + LABAs at the lowest possible dose.
Conflict of interest
Dr. Fernando Saldarini has
participated in medical conferences on asthma for AstraZeneca, GlaxoSmithKline,
ELEA, TEVA Chile, Janssen, and SANOFI for continuing medical education
programs.
Dr. Diego Litewka has
participated in conferences and continuing medical education programs on asthma
for Novartis, Sanofi, and GlaxoSmithKline.
Dr. Martin Sívori has
participated in conferences and continuing medical education programs on asthma
for AstraZeneca, ELEA, and GlaxoSmithKline.
Dr. Daniel Pascansky has
participated in conferences, consultations on asthma for AstraZeneca,
GlaxoSmithKline, Boehringer Ingelheim, Elea, Casasco, as well as continuing
medical education programs on asthma for Novartis.
REFERENCES
1. Reddel HK, Yorgancioglu A.
Global Strategy for Asthma Management and Prevention. GINA Update 2023. Acceso el 19 de febrero de 2024 en
https://ginasthma.org/wp-content/uploads/2023/07/GINA-2023-Full-report-23_07_06-WMS.pdf
2. Forno E, Gogna M, Cepeda A, et al. Asthma in Latin America. Thorax 2015;70:898-905.
https://doi.org/10.1136/thoraxjnl-2015-207199
3. Arias S, Neffen H, Bossio JC,
et al. Prevalence and features of asthma in Young adults in urban areas of Argentina.
Arch Bronconeumol 2018;54:134-9.
https://doi.org/10.1016/j.arbres.2017.08.021
4. Neffen H, Moares F, Viana K,
et al. Asthma severity in four countries of Latin America. BMC Pulm Med 2019;19:123. https://doi.org/10.1186/s12890-019-0871-1
5. Holguin F, Cardet JC, Chung
KF, et al. Management of severe asthma: an European
Respiratory Society/ American Thoracic Socienty Guideline. Eur Respir J 2020;55:1900588. https://doi.org/10.1183/13993003.00588-2019
6. Alobid I, Álvarez Rodríguez C, Ferreira
J, et al. GEMA 5.3. Guía Española para el manejo del asma. Acceso
el 19 de febrero de 2024 en www.gemasma.com
7. Global Initiative for Asthma. Difficult to treat & severe qsthma in adolescent and adult patients:
Diagnosis and Treatment Version 4. August 2023. Acceso el
19 de febrero de 2024 en
https://ginasthma.org/wp-content/uploads/2023/09/GINA-Severe-Asthma-Guide-2023-WEB-WMS.pdf
8. Moore WC, Meyers DA, Wenzel
SE, et al. National Heart, Lung, and Blood Institute’s Severe Asthma Research
Program. Identification of asthma phenotypes using a clustering
analysis in the Severe Asthma Research Program. Am J Respir Crit Care
Med 2010;181:315-23. https://doi.org/10.1164/rccm.200906-0896OC
9. Wenzel S. Asthma phenotypes:
the evolution from clinical to molecular approaches. Nat Med 2012;18:716-25. https://doi.org/10.1038/nm.2678
10. Agache IO. From
phenotypes to endotypes to asthma treatment. Curr Opin Allergy Clin
Immunol 2013;13:249-56.
https://doi.org/10.1097/ACI.0b013e32836093dd
11. Fitzpatrick AM, Moore WC.
Severe asthma phenotypes - how should they guide evaluation and treatment? J
Allergy Clin Immunol Pract 2017;5:901-8.
https://doi.org/10.1016/j.jaip.2017.05.015
12. Shaw DE, Sousa AR, Fowler SJ,
Fleming LJ, Roberts G, Corfield J, et al; U-BIOPRED Study Group. Clinical and inflammatory characteristics of the European
U-BIOPRED adult severe asthma cohort. Eur Respir J 2015;46:1308-21. https://doi.org/10.1183/13993003.00779-2015
13. Kuo CS, Pavlidis S, Loza M,
Baribaud F, Rowe A, Pandis I, et al; U-BIOPRED Study Group. T-helper cell type
2 (Th2) and non-Th2 molecular phenotypes of asthma using sputum
transcriptomics in U-BIOPRED. Eur Respir J 2017;49:1602135.
https://doi.org/10.1183/13993003.02135-2016
14. Xolair [omalizumab] full
prescribing information. Acceso el 19 de febrero
de 2024 en www.xolair.com/prescribing_information.html
15. Manka LA, Weschler ME. Selecting the right biologic for your patients with severe asthma.
Ann Allergy Asthma Immunol 2018;121:406-13.
https://doi.org/10.1016/j.anai.2018.07.033
16. Walsh GM. Recent
developments in the use of biologics targeting IL-5, IL-4or IL-13 in severe
refractory asthma. Expert Rev Respir Med 2018;12:957-63.
https://doi.org/10.1080/17476348.2018.1520095
17. Busse WW. Biological
treatments for severe asthma: a major advance in asthma care. Aller Inter 2019;68:154-66. https://doi.org/10.1016/j.alit.2019.01.004
18. Bel EH, Wenzel S, Thompson
PJ, et al. Oral glucocorticoid-sparing effect of mepolizumab in eosinophilic
asthma. N Engl J Med 2014;371:1189-97.
https://doi.org/10.1056/NEJMoa1403291
19. Marfone G, Spadaro G, Braile
M, et al. Tezepelumab: a novel biological therapy for the treatment of severe
uncontrolled asthma. Exp Op Invest Drugs 2019;28:931-40.
https://doi.org/10.1080/13543784.2019.1672657
20. Menzies-Gow A, Colice G,
Griffiths JM, et al. NAVIGATOR: a phase 3 multicentre, randomized,
double-blind, placebo controlled, parallel-group trial to evaluate the efficacy
and safety of tezepelumab in adults and adolescents with severe, uncontrolled
asthma. Respir Research 2020;21:266.
https://doi.org/10.1186/s12931-020-01526-6
21. MenziesGow A, Ponnarambil S, Downie J, et al. DESTINATION: a phase 3, multicentre,
randomized, doubleblind, placebocontrolled, parallelgroup trial to evaluate the
longterm safety and tolerability of tezepelumab in adults and adolescents with
severe, uncontrolled asthma. Respir Research 2020;21:279.
https://doi.org/10.1186/s12931-020-01541-7
22. Wechsler ME, Colice G,
Griffiths JM, 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 Research 2020;21:264.
https://doi.org/10.1186/s12931-020-01503-z
23. Brightling CE, Gupta S, Gonem
S, Siddiqui. Lung damage and airway remodeling in severe
asthma. Clin Exper Allergy 2011:1-12. https://doi.org/10.1111/j.1365-2222.2011.03917.x
24. Domvri K, Porpodis K.
Targeting inflammation or remodeling in asthma? Is there a right way? Front
Med 2023;10:20231.
https://doi.org/10.3389/fmed.2023.1241920
25. Kardas G, Kuna P, Panek M.
Biological therapies of severe asthma and their possible effects on airway
remodeling. Front Immunol 2020;11:1134.
https://doi.org/10.3389/fimmu.2020.01134
26. Flood-Page P, Menzies-Gow A,
Phipps S, Ying S, Wangoo A, Ludwig MS, et al. Anti-IL-5 treatment reduces
deposition of ECM proteins in the bronchial subepithelial basement membrane
of mild atopic asthmatics. J Clin Invest 2003;112:1029-36.
https://doi.org/10.1172/JCI17974
27. Domvri K, Tsiouprou I,
Bakakos P, Rovina N, Stiropoulos P, Voulgaris A. Effect of Mepolizumab on
airways remodeling in patients with late-onset severe eosinophilic asthma and
fixed obstruction (preliminary data of the MESILICO study). Eur Res J 2023;
62:OA3152. https://doi.org/10.1183/13993003.congress-2023.OA3152
28. Laviolette M, Gossage DL, Gauvreau
G, et al. Effects of benralizumab on airway eosinophils in asthmatic patients
with sputum eosinophilia. J Allergy Clin Immunol
2013;132:1086-96.
https://doi.org/10.1016/j.jaci.2013.05.020
29. Zastrzezynska W, Przybyszowski M, Bazan-Socha S, et
al. Omalizumab may decrease the thickness of the reticular
basement membrane and fibronectin deposit in the bronchial mucosa of severe
allergic asthmatics. J Asthma 2020;57:468-77.
https://doi.org/10.1080/02770903.2019.1585872
30. Diver S, Khalfaoui L, Emson
C, et al. Effect of tezepelumab on airway inflammatory cells, remodelling, and
hyperresponsiveness in patients with moderate-to-severe uncontrolled asthma
(CASCADE): a double-blind, randomised, placebo-controlled, phase 2 trial.
Lancet Respir Med 2021;9:1299-312.
https://doi.org/10.1016/S2213-2600(21)00226-5
31. Chen ML, Nopsopon T, Akenroye
A. Incidence of anti-drug antibodies to monoclonal antibodies in asthma: a
systematic review and meta-analysis. J Allergy Clin Immunol Pract 2023;10:1-10. https://doi.org/10.1016/j.jaip.2022.12.046
32. Thomas D, McDonald VM, Pavord ID, Gibson PG. Asthma remission: what is it and how
can it be achieved? Eur Respir J 2022;60:2102583.
https://doi.org/10.1183/13993003.02583-2021
33. Menzies-Gow A, Bafadhel M,
Busse WW, et al. An expert consensus framework for asthma
remission as a treatment goal. J Allergy Clin Immunol 2020;145:757-65. https://doi.org/10.1016/j.jaci.2019.12.006
34. Jackson DJ, Busby J, Pfeffer
PE, et al. Characterisation of patients with severe asthma in the UK Severe
Asthma Registry in the biologic era. Thorax 2021;76:220-7.
https://doi.org/10.1136/thoraxjnl-2020-215168
35. Bleecker ER, Menzies-Gow AN,
Price DB, et al. Systematic literature review of systemic corticosteroid use
for asthma management. Am J Respir Crit Care Med 2020;201:276-93.
https://doi.org/10.1164/rccm.201904-0903SO
36. Upham JW, Le Lievre C,
Jackson DJ, et al. Defining a severe asthma
super-responder: findings from a Delphi process. J Allergy Clin Immunol Pract
2021;9:3997-4004.
https://doi.org/10.1016/j.jaip.2021.06.041
37. Pérez de Llano L, Cisneros C,
Domínguez-Ortega J, et al. Response to
monoclonal antibodies in asthma: definitions, potential reasons for failure and
therapeutic options for suboptimal response. J Investig Allergol Clin Immunol
2023;33:1-13. https://doi.org/10.18176/jiaci.0857
38. Douglass JA, Pavord I,
Brusselle G, et al. Dupilumab leads to clinical asthma remission indicative of
comprehensive improvement in patients with asthma: results from the LIBERTY
ASTHMA QUEST and TRAVERSE STUDIES. Intern Med J 2022;52
Suppl. 5:5-32. https://doi.org/10.1111/imj.52_15894
39. Pérez de
Llano L, Veiga-Teijeiro I, Dacal-Riva D. Contribution of the Remission Concept
to the Treatment of Asthma. Arch
Bronconeumol 2023;59:550-1.
https://doi.org/10.1016/j.arbres.2023.03.009
40. Carpaij OA,
Burgess JK, Kerstjens HAM, Nawijn MC, van den Berge M. A review on the pathophysiology of asthma remission.
Pharmacol Ther 2019;201:8-24.
https://doi.org/10.1016/j.pharmthera.2019.05.002
41. Moore WC, Kornmann O, Humbert
M, et al. Stop- ping versus continuing long-term mepolizumab treatment in
severe eosinophilic asthma (COMET study). Eur Respir J 2022;59:2100356.
https://doi.org/10.1183/13993003.00396-2021
42. Bacharier LB. Asthma
guidelines: where to next? Ann Allergy Asthma Immunol 2022;128:346-7.
https://doi.org/10.1016/j.anai.2021.12.017
43. Nagase H, Suzukawa M, Oishi
K, Matsunaga K. Biologics for severe asthma: the real-world evidence,
effectiveness of switching and prediction factors for the efficacy. Allergol
Int 2023;72:11-23.
https://doi.org/10.1016/j.alit.2022.11.008
44. Molimard M, Mala L, Bourdeix
I, Le Gros V. Observational study in severe asthmatic patients after discontinuation
of omalizumab for good asthma control. Respir Med 2014;108:571-6.
https://doi.org/10.1016/j.rmed.2014.02.003
45. Humbert M, Bourdin A, Taille
C, et al. Real-life omalizumab exposure and discontinuation in a large
nationwide population-based study of paediatric and adult asthma patients. Eur Respir J 2022;60: 2103130. https://doi.org/10.1183/13993003.03130-2021
46. Chapman KR, Albers FC, Chipps
B, et al. The clinical benefit of mepolizumab replacing
omalizumab in uncontrolled severe eosinophilic asthma. Allergy 2019;74:1716-26.
https://doi.org/10.1111/all.13850
47. Bagnasco D, Menzella F, Caminati M, et al. Eficacy of mepolizumab in patients with previous omalizumab treatment
failure: real-life observation. Allergy 2019;74:2539-41. https://doi.org/10.1111/all.13937
48. Pelaia C, Crimi C, Nolasco S, et al. Switch from omalizumab to Benralizumab in allergic patients with severe
eosinophilic asthma: a real-life experience from Southern Italy. Biomedicines
2021;9:1822.
https://doi.org/10.3390/biomedicines9121822
49. Menzies-Gow AN, McBrien C,
Unni B, et al. Real world biologic use and switch patterns in severe asthma:
data from the international severe asthma registry and the US CHRONICLE study.
J Asthma Allergy 2022;15:63-78.
https://doi.org/10.2147/JAA.S328653
50. Chapman KR, Albers FC, Chipps
B, et al. The clinical benefit of mepolizumab replacing
omalizumab in uncontrolled severe eosinophilic asthma. Allergy 2019;74:1716-26.
https://doi.org/10.1111/all.13850
51. Carpagnano GE, Resta E, Povero M, et al. Clinical and economic consequences of switching from
omalizumab to mepolizumab in uncontrolled severe eosinophilic asthma. Sci Rep 2021;11:5453.
https://doi.org/10.1038/s41598-021-84895-2
52. Scioscia G, Nolasco S,
Campisi R, et al. Switching Biological Therapies in Severe Asthma. Int J Mol
Sci 2023;24:9563. https://doi.org/10.3390/ijms24119563
53. Drick N, Milger K, Seeliger
B, et al. Switch from IL-5 to IL-5-Receptor alpha antibody treatment in severe
eosinophilic asthma. J Asthma Allergy 2020;13:605-14.
https://doi.org/10.2147/JAA.S270298
54. Kavanagh JE, Hearn AP,
d’Ancona G, et al. Benralizumab after sub-optimal response to mepolizumab in
severe eosinophilic asthma. Allergy 2021;76:1890-3.
https://doi.org/10.1111/all.14693
55. Martínez-Moragon E,
García-Moguel I, Nuevo J, Resler G. ORBE study investigators. Real-world
study in severe eosinophilic asthma patients
re-fractory to anti-IL5 biological agents treated with Benralizumab in Spain
(ORBE study). BMC Pulm Med 2021;21:417.
https://doi.org/10.1186/s12890-021-01785-z
56. Laviolette M, Gossage DL,
Gauvreau G, et al. Effects of Benralizumab on airway eosinophils in asthmatic
patients with sputum eosinophilia. J Allergy Clin Immunol 2013;132:1086-96.e5. https://doi.org/10.1016/j.jaci.2013.05.020
57. Busse W, Chupp G, Nagase H,
et al. Anti-IL-5 treatments in patients with severe asthma by blood eosinophil
thresholds: indirect treatment comparison. J Allergy Clin Immunol 2019;143:190-200.e20. https://doi.org/10.1016/j.jaci.2018.08.031
58. Bourdin A, Husereau D, Molinari
N, et al. Matching-adjusted indirect comparison of Benralizumab versus interleukin-5
inhibitors for the treatment of severe asthma: a systematic review. Eur Respir
J 2018;52:1801393. https://
doi.org/10.1183/13993003.01393-2018
59. Sridhar S, Liu H, Pham TH,
Damera G, Newbold P. Modulation of blood inflammatory markers by Benralizumab
in patients with eosinophilic airway diseases. Respir Res 2019;20:14. https://doi.org/10.1186/s12931-018-0968-8
60. Lommatzsch M, Marchewski H,
Schwefel G, Stoll P, Virchow JC, Bratke K.
Benralizumab strongly reduces blood basophils in severe eosinophilic asthma.
Clin Exp Allergy 2020;50:1267-9.
https://doi.org/10.1111/cea.13720
61. Dagher R, Kumar V, Copenhaver
AM, et al. Novel mechanisms of action contributing to Benralizumab’s potent
anti-eosinophilic activity. Eur Respir J 2022;59:2004306.
https://doi.org/10.1183/13993003.04306-2020
62. Lei A, He Y, Yang Q, Li X, Li
R. Role of myeloid cells in the regulation of group 2 innate lymphoid
cell-mediated allergic inflammation. Immunology 2020;161:18-24.
https://doi.org/10.1111/imm.13232
63. Schmid-Grendelmeier P,
Altznauer F, Fischer B, et al. Eosinophils express functional IL-13 in
eosinophilic inflammatory dis-eases. J Immunol 2002;169:1021-7.
https://doi.org/10.4049/jimmunol.169.2.1021
64. Lambrecht BN, Hammad H, Fahy
JV. The cytokines of asthma. Immunity 2019;50:975-91. https://doi.org/10.1016/j.immuni.2019.03.018
65. Dupin C, Belhadi D,
Guilleminault L, et al. Effectiveness and safety of dupilumab for the treatment
of severe asthma in a real-life French multi-centre adult cohort. Clin Exp
Allergy 2020;50:789-98.
https://doi.org/10.1111/cea.13614
66. Numata T, Araya J, Miyagawa
H, et al. Real-world effectiveness of dupilumab for patients with severe
asthma: a retrospective study. J Asthma Allergy 2022;15:395-405.
https://doi.org/10.2147/JAA.S357548
67. Mümmler C,
Dünzelmann K, Kneidinger N, et al. Real-life effectiveness of biological
therapies on symptoms in severe asthma with comorbid CRSwNP. Clin Transl Allergy 2021;11:e12049. https://doi.org/10.1002/clt2.12049
68. Menzies-Gow A, Hoyte FL,
Price DB, et al. Clinical remission in severe asthma: a pooled post hoc
analysis of the patient journey with Benralizumab. Adv Ther 2022;39:2065- 84. https://doi.org/10.1007/s12325-022-02098-1
69. Hamada K, Oishi K, Murata Y,
Hirano T, Matsunaga K. Feasibility of dis-continuing biologics in severe
asthma: an algorithmic approach. J Asthma Allergy 2021;14:1463-71.
https://doi.org/10.2147/JAA.S340684
70. Cohn L. Can asthma biologics
change the course of disease and induce drug-free remission? J Allergy Clin
Immunol 2022;150:59-61.
https://doi.org/10.1016/j.jaci.2022.04.005
71. Jackson DJ, Heaney LG,
Humbert M, et al. Reduction of daily maintenance inhaled corticosteroids in
patients with severe eosinophilic asthma treated with benralizumab (SHAMAL): a
randomised, multicentre, open-label, phase 4 study. Lancet
2023; published online Dec 7. https://doi.org/10.1016/S0140-6736(23)02284-5. https://doi.org/10.1183/13993003.congress-2023.RCT798
72. Center of Disease Control of
United State of North America. Acceso el 19 de febrero
de 2024 en https://www.cdc.gov/pregnancy/spanish/meds/treatingfortwo/index
73. Joshi E, Gibson PG, McDonald VM, et al. Treatable traits in asthma during pregnancy: a call for a shift towards
a precision-based management approach. Eur Respir Rev 2023; 32: 230105.
https://doi.org/10.1183/16000617.0105-2023
74. Chambers C. Chapter: Safety
of Asthma and Allergy medications during pregnancy, In. Asthma, Allergic and
Immunologic Diseases During Pregnancy. Editors Namazy,
J., Schatz, M. Springer, 2019, pp 15-27. https://
doi.org/10.1007/978-3-030-03395-8_2
75. MotherToBaby. Acceso el 1 de enero de 2024 en
https:// mothertobaby.org/ongoing-study/asthma
76. List of Pregnancy Exposure Registries. Acceso el 19
de febrero de 2024 en
https://www.fda.gov/science-research/womens-health-research/list-pregnancy-exposure-registries
77. Ruth P. Cusack, Christiane E.
Whetstone, Gail M. Gauvreau, Professorc.Immunol Allergy Clin N Am
2023;43:169-85. https://doi.org/10.1016/j.iac.2022.07.007
78. Middleton PG, Gade EJ, Aguilera C, et al. ERS/TSANZ Task Force Statement on the management of reproduction and
pregnancy in women with airways diseases. Eur Respir J 2020;55:1901208.
https://doi.org/10.1183/13993003.01208-2019
79. Saito J, Yakuwa N, Sandaiji
N, et al. Omalizumab concentrations in pregnancy and lactation: A case study.
J Allergy Clin Immunol Pract 2020;8:3603-4.
https://doi.org/10.1016/j.jaip.2020.05.054
80. Namazy J, Cabana MD, Scheuerle AE, et al. The Xolair Pregnancy Registry (EXPECT): the safety of omalizumab use
during pregnancy. J Allergy Clin Immunol 2015;135:407-12.
https://doi.org/10.1016/j.jaci.2014.08.025
81. Namazy JA, Blais L, Andrews
EB, et al. Pregnancy outcomes in the omalizumab pregnancy registry and a
disease-matched comparator cohort. J Allergy Clin Immunol 2020;145:528-36. https://doi.org/10.1016/j.jaci.2019.05.019
82. Food and Drug Adminsitration
(FDA). Mepolizumab (Nucala) [prospecto]. Acceso el 19 de febrero
de 2024 en
https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/125526Orig1s021,761122Orig1s011Corrected_lbl.pdf
83. Study 200870. GSK Study Register. Study entry at:
https://www.gsk-studyregister.com/en/trialdetails/id=200870
84. AstraZeneca Clinical Trials. Benralizumab pregnancy exposure study. D3250R00026.
(ClinicalTrials.gov: NCT03794999). Acceso el 19 de febrero
de 2024 en https://www.astrazenecaclinicaltrials.com/study/D3250R00026/
85. Regeneron Pharmaceuticals.
Post-authorization Safety Study in North America to Monitor Pregnancy and
Infant Outcomes Following Administration of Dupilumab During Planned or
Unexpected Pregnancy (Clinical Trials.gov: NCT04173442). Acceso el 19 de febrero de 2024 en https://classic.clinicaltrials.gov/ct2/show/NCT04173442
86. AstraZeneca. Tezepelumab (Tezspire) [prospecto].
Sitio web de la Administración de Medicamentos y Alimentos de EE. UU.
Disponible en:https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/761224s000lbl.pdf
87. World Health Organization.
Adherence to long term therapies: evidence for action. Geneva. 2003. Acceso el 2 de febrero de 2024 en
https://www.paho.org/en/documents/who-adherence-long-term-therapies-evidence-action-2003
https://doi.org/10.1016/S1474-5151(03)00091-4
88. Braidos F. Failure in asthma
control: reasons and consequence. Scientifca 2013:549252:
1-15. https://doi.org/10.1155/2013/549252
89. Busby J, Matthews JG,
Chaudhuri R et al. Factors affecting adherence with treatment advice in a
clinical trial of patients with severe asthma. Eur Respir J 2022;5:2100768. https://doi.org/10.1183/13993003.00768-2021
90. Williams LK, Peterson EL,
Wells K, et al. Quantifying the proportion of severe
asthma exacerbations attributable to inhaled corticosteroid non-adherence. J
Allergy Clin Immunol 2011;12:1185-91.
https://doi.org/10.1016/j.jaci.2011.09.011
91. Barnes CB, Ulrik CS. Asthma
and adherence to inhaled corticosteroids: current status and future
perspectives. Respir Care 2015;60:455-68. https://doi.org/10.4187/respcare.03200
92. Sivori M, Pascansky D. Asma grave T2 alto:
Análisis del diseño de los estudios clínicos de los nuevos
biológicos. Rev Amer Med Resp 2022;1:98-115.
93. Costello RW, Cushen B.
Looking back to go forward: adherence to inhaled therapy before biologic
therapy in severe asthma Eur Respir J 2020;55:20000954.
https://doi.org/10.1183/13993003.00954-2020
94. Menzella F, Ferrari E, Ferrucci SM, et al. Self-administration of omalizuamb: why not? A
literature review and expert opinion. Exp Opin Biol Ther 2021;21:499-507. https://doi.org/10.1080/14712598.2021.1882990
95. Bernstein D, Pavord ID,
Chapman KR et al. Usability of mepolizumab single-use prefilled autoinjector
for patient self-administration. J Asthma 2020;57:987-98.
https://doi.org/10.1080/02770903.2019.1630641
96. Martin UJ, Fuhr R, Forte P,
et al. Comparison of autoinjector with access prefilled syringe for
benralizumab pharmacokinetic exposure: AMES trial. J Asthma 2019;58:93-101.
https://doi.org/10.1080/02770903.2019.1663428
97. Cohen YZ, Zhang X, Xia B, et al. Pharmacokinetics of subcutaneous dupilumab injection
with and autoinjector device or prefille siringe. Clin Pharmac Drug Dev 2022;11:675-81.
https://doi.org/10.1002/cpdd.1073
98. McNicholl DM, Stevenson M,
McGarvey LP, et al. The utility of fractional exhaled nitric
oxide suppression in the identification of nonadherence in difficult asthma.
Am J Respir Crit Care Med 2012;186:1102-8. https://doi.org/10.1164/rccm.201204-0587OC
99. Heaney LG, Busby J, Bradding
P, et al. Remotely monitored therapy and nitric oxide
suppression identifies nonadherence in severe asthma. Am
J Respir Crit Care Med 2019; 199:454-64. https://doi.org/10.1164/rccm.201806-1182OC
100. Couillard S,Shrimaner R,
Chaudhuri R, et al. Fractional exhaled nitric Oxide Nonsuppression indentifies
corticosteroid-resistant type 2 signaling in severe asthma. Am J Respir Crit
Care Med 2021;731-3.
https://doi.org/10.1164/rccm.202104-1040LE
101. Butler CA, McMicahel AJ,
Honeyford K, et al. Utility of fractional exhaled nitric oxide suppression as
prediction tool for progression to biologic therapy. ERJ Open Res 2021;7:00273-2021. https://doi.org/10.1183/23120541.00273-2021