Skip to main content

Allergic diseases in the elderly: biological characteristics and main immunological and non-immunological mechanisms

Abstract

Life expectancy and the number of elderly people are progressively increasing around the world. Together with other pathologies, allergic diseases also show an increasing incidence in geriatric age. This is partly due to the growing emphasis on a more accurate and careful diagnosis of the molecular mechanisms that do not allow to ignore the real pathogenesis of many symptoms until now unknown, and partly to the fact that the allergic people from 20 years ago represent the elderly population now. Moreover, environmental pollution predisposes to the onset of allergic asthma and dermatitis which are the result of internal pathologies more than the expression of allergic manifestations. At the same time the food contamination permits the onset of allergic diseases related to food allergy. In this review we provide the state of the art on the physiological changes in the elderly responsible for allergic diseases, their biological characteristics and the major immunological and extra immunological mechanisms. Much emphasis is given to the management of several diseases in the elderly, including anaphylactic reactions. Moreover, some new features are discussed, such as management of asthma with the support of physical activity and the use of the AIT as prevention of respiratory diseases and for the purpose of a real and long lasting benefit. The mechanisms of adverse reactions to drugs are also discussed, due to their frequency in this age, especially in polytherapy regimens. Study of the modifications of the immune system is also of great importance, as regards to the distribution of the lymphocytes and also the presence of a chronic inflammatory disease related to the production of cytokines, especially in prevision of all the possible therapies to be adopted to allow an active and healthy aging.

Background

The aging population in Europe and all over the world implicates increasing social problems and a specific scientific support is by now as required in order to ensure what is called a “successful aging”. In fact, epidemiological data attest always more the need for a healthy aging to contain the increased economic burden for assistance of a growing number of people over 65 in condition of “fragility”. Aging is an unavoidable, universal, complex process characterized by progressive loss of functional reserves and reduction of the capability to adapt to the environment. This biological phenomenon derives from an interaction between genetic and environmental factors. Healthy lifestyle and favorable environment allow genetically predisposed subjects to reach extreme longevity and maintain acceptable health status and auto-sufficiency [1].

Disfunctions of the immune system are the basis of many disabling diseases that occur during aging, including neurodegenerative and cardiovascular diseases, whose appearance is due to the state of proinflammatory, also called “inflamm-aging”, that is typical of the elderly [2]. The mechanisms that enable a “successful” aging are related to certain physical and immunologic conditions. This is due to multiple factors, including the increased pollution; moreover the allergic pediatric population that was born 50–60 years ago has reached the age of senescence. Therefore, diseases such as food allergy, urticaria, allergic reactions to drugs, and anaphylaxis are becoming more frequent in the elderly population [3].

The purpose of this work is to provide the state of the art on the physiological changes in the elderly responsible for allergic diseases, their biological characteristics and the major immunological and extra immunological mechanisms.

Lymphocyte subpopulations in the elderly

Most information on lymphocyte subpopulations and their changes occurring with age derive from the cytofluorimetric analysis using monoclonal antibodies directed to surface antigens of gated lymphoid cells collected from peripheral venous blood. It is well known that the total number of lymphocytes—the main circulating subset of leukocytes in the first years of life declines since young adulthood and then reaches very low numbers from age 60 onwards. And it is also known that usually about 2% of all lymphocytes appear to recirculate in the blood, while the remaining are resident in gut and spleen or circulate through all the lymphoid system (lymph nodes, mucosal aggregates, thoracic duct, tonsils etc.). Therefore our assumption is that the blood circulating 2% should reflect the proportion of lymphocyte subpopulations really present in the entire body. This can be true for some (e.g. γ/δ T cells, NK T cells, CD8 αα, and many others), but not all subsets, because some are strictly compartmentalized mainly to the mucosa. The main lymphocyte subpopulations discussed briefly here will be T subsets, B cells and innate immunity.

At birth high numbers of B cells are present, and low NK cells, and they will change in opposite directions during the individual’s lifetime. T cells are generated through thymic selection and the entire repertoire is established by the 10th year of life, while the thymus will undergo progressive involution from the age of 30. T cells represent the major subset (55–70% of all lymphocytes) in young people and so remain until late age. However the two main T cell subsets, identified by the coreceptor molecules CD4 and CD8, show different changes with ageing, with a progressive increase of CD8+ cells with advancing age.

CD4+ and CD8+ peripheral blood T lymphocytes show mutually exclusive expression of CD45RA or CD45R0, two isoforms of the common leukocyte antigen that help recognize respectively virgin/unprimed and memory/activated T cells [4]. Changes in the subsets have been observed with age: at birth 95–99% of the CD3+ T lymphocytes expressed the CD45RA isoform, while an increase of CD45R0+ cells was observed until 30 years. Nonetheless, in very old age a proportion of CD45RA+ among CD4+ cells was still present (about 20%) and even more in CD8+ lymphocytes (up to 40–50%). These data originate from very selected subjects without chronic common pathologies or taking drugs able to affect the blood subpopulations. The reverse “frail” or disabled condition can lead to different observations, with many older subjects showing many more CD8+ and “memory” phenotypes. These considerations lead to analyze the presence and role of thyme in older age. The generation of new T cells is indeed crucial to maintain a functional immune system, and lymphocytes defined “recent thymic emigrants”, as well as the detection of the so called “T cell receptor rearrangement excision circles (TREC)”, have been studied [5], leading to the conclusion that new T cells are produced even in old age, although in very low numbers. Molecules able to stimulate thymic production include cytokines like interleukin (IL)-7, IL-15 and others. The level of expression of the interleukin 7 receptor (IL-7R) gene in blood has recently been found to be associated with familial longevity and healthy ageing [6]. IL-7R is crucial for T cell development and important for immune competence.

The increased CD4:CD8 ratios in the elderly were significantly lower in Cytomegalovirus (CMV)-seropositive individuals, that in our experience represent >98% of aged subjects. This study also showed a lower naïve and a larger late-differentiated compartment of CD8+ αβ T-cells. Longitudinal studies suggest that accumulation of these cells is a dynamic process as an age-associated change [7].

A coincevable hypothesis is that these events are at least partly due to the effects of the maintenance of essential immune surveillance against persistent viral infections, notably CMV, which may exhaust the immune system over time, as shown in the NONA immune longitudinal study [4]. From this and other analysis an immune risk profile (IRP) was identified, defined by an inverted CD4/CD8 ratio and associated with persistent cytomegalovirus infection and increased numbers of CD8+ CD28− cells, which led to a relative 6-year survival in NONA individuals.

As regards the functional characteristics of these T cell compartments in the elderly, exhausted or non-functional CD8+ cells have been described in several studies. T cell clones (TCC) of two healthy centenarians were studied and compared them with TCC obtained by three young normal subjects: totally 180TCC were analyzed. In young donors, 35TCC were CD4+, 56 CD8+ and two were alpha beta +CD4−CD8− (double negative). In centenarians, 46CD4+TCC, 38CD8+, 2CD4+CD8+ (double positive) and 1 gamma delta+ double negative were obtained [8]. The cytokine analysis showed that the Th profiles of CD8+TCC are nearly overlapping in two groups, whereas a major shift from a Th1 to a Th0 pattern is shown by CD4+TCC.

It is well known that T cells from aged subjects have a reduced ability to produce IL-2, but other functions seem to be upregulated, since IL-1, IL-4, IL-6 and TNFα production are increased. These cytokines are known to control B cell differentiation, through isotype switch and Ig production. A significant age-related increase of the serum level of immunoglobulin classes (IgG and IgA but not IgM) and IgG subclasses (IgG1, 2 and 3, but not IgG4) is detected [9]. An age-related decrease of circulating B cells occurs in parallel. The impairment of primary responses to immunization, and other aspects of humoral immunity, including mucosal responses, autoantibody production led to the hypothesis of a complex derangement of B cell function and/or compartmentalization with age. Unusual IgD(−)CD27(−) double-negative B cell population was described in the elderly [10]. Most of these were less functional memory B cells, so it is speculated these were also late memory or exhausted cells that have down-modulated the expression of CD27 and filled the immunologic space in the elderly.

Finally, also adaptive immunity undergoes severe deterioration with age and represents the main problem in the elderly. Evidence accumulated over the last decade supports the hypothesis that aging also has a profound impact on innate immunity, including reduced or dysfunctional NK lymphocytes, which in turn markedly affects the health and longevity of older people [11].

The role of cytokines in the elderly

Aging is a dynamic process characterized by a continuous remodeling sustained by DNA repair, apoptosis, immune response, oxidative stress and inflammation. To reach extreme longevity, it is necessary for all these mechanisms to interact in an efficient way. The theory of inflammaging focuses on the activation of a subclinical, chronic low-grade inflammation that occurs with aging. It is a manifestation of immunosenescence, which is the sum of changes affecting the functionality of the immune system in elderly.

A major contributor of inflammaging is the antigenic load of persistent infection which starts from intrauterine life and continues for an entire lifespan. The pro-inflammatory phenotype of senescent cells protects against infectious diseases but, at the same time, contributes to inflammaging, as large quantities of pro-inflammatory cytokines are produced/released in order to repair tissue damage [12]. Many age-related diseases share an inflammatory pathogenesis; moreover, the activation of inflammatory pathways appears to be involved in the pathophysiology of sarcopenia and frailty. Long-lived people, especially centenarians, seem to cope with chronic subclinical inflammation through an anti-inflammatory response, called therefore “anti-inflammaging” [13]. We believe that if inflammaging is a key to understand aging, anti-inflammaging may be one of the secrets of longevity.

Circulating cytokines are involved both in inflammation and anti-inflammation, and are the expression of a system that involves genes and environment.

Proinflammatory cytokines in aging

The most relevant proinflammatory cytokines involved in aging processes are interleukin (IL)-1, IL-6, tumor necrosis factor (TNF) and interferon (IFN)-γ. High levels of these cytokines are associated with increased risk of morbidity and mortality in the older subject. In particular, IL-1 is involved in the alteration of nutritional status, in cognitive decline, and development of Alzheimer’s disease [14] and as a risk factor for depressive symptoms in aged people [15]. IL-6 and TNF serum levels influence the onset of frailty, poor physical performance, loss of muscle strength, cognitive decline, cerebrovascular and cardiovascular events [14, 16]. These cytokines are also closely linked to the genesis of cancers (IL-6), to cardiac remodeling in heart failure [14] and to the risk of community-acquired pneumonia requiring hospitalization (both IL-6 and TNF) [17].

An increase of intracellular IFN-γ, together with other type 1 cytokines, within three CD8+ T subsets (naïve, effector/cytotoxic and memory) was observed in aged subjects [18]. Conversely, the % age of IFN-γ positive cells significantly decreased in a virgin CD4+ subset as well as in activated/memory T cells from old subjects [19]. However, in a study on nonagenarians, IFN-γ plasma levels are unmodified compared to young controls, suggesting a substantial maintenance of relevant T cell functions [20].

Another potent proinflammatory cytokine that plays a relevant role in aging is IL-18. A recent study showed that IL-18 is a predictor of mortality for heart failure in a group of octogenarians. A study of follow-up (of mean 2.4 years) underlighted that subjects who died for heart failure had higher serum values of IL-18 compared with those still alive [21]. IL-18 serum levels increase with ageing [22], but at the same time longevity is characterized by a reduced incidence of ischemic events.

Our group evaluated serum levels of IL-18 and IL-18 binding protein (IL-18BP) in healthy centenarians, younger healthy controls and patients with chronic ischemic cardiac disease. Centenarians showed significantly higher levels of IL-18 and IL-18BP compared to each control group. Elevated IL-18 levels were also present in patients with chronic ischemic syndrome; centenarians exhibited a lower level of free IL-18 than chronic ischemic patients. These results indicate that inactivation of IL-18 by IL-18BP may explain the apparent paradox of high serum levels of IL-18 in centenarians, without signs of vascular diseases [23].

Significant increased IL-12 serum levels were found in nonagenarians compared to young controls; this might be related to the increase of NK cell functions characterizing aging processes [20]. Different data were shown by other authors [24] who found that age was not correlated with Toll-like receptor (TLR)-mediated IL-12 and IL-23 production after stimulation of whole blood cells. In contrast, poor nutritional status and frailty and not age by itself were associated with decreased IL-12 and IL-23 production. These results in frail, elderly patients could account for their susceptibility to many pathogens [24].

In a study of our group centenarians subjects showed significantly higher IL-15 levels compared to both young and old controls [25]. These findings may explain, at least in part, the characteristic increase of memory cells in elderly subjects and may lead to a better understanding of the capacity of adaptation to the environment of centenarians, defending themselves from infections through immune-inflammatory responses.

We also found increased IL-22 serum levels in older patients with chronic heart failure [26] and in healthy centenarians [27]. In particular, only the II and III NYHA class had IL-22 values significantly higher than the controls, whereas there was no difference between the IL-22 levels of NYHA class IV and the controls [26]. The reason may be the decline of immune function in patients with chronic heart failure. In fact we can hypothesize that the reduction of IL-22 levels with the progression of NYHA class can be responsible of the impaired ability of these patients to respond to infections, as IL-22 has anti-microbial properties [26]. Therefore, it is likely that the pro-inflammatory condition sustained by IL-22 is protective against infection, promoting the longevity of these subjects [27]. It might be speculated that under certain circumstances the activity of this cytokine against infection may be more relevant to survival than its potential negative impact on inflammation.

The data on IL-2 in long-lived people are controversial. An age-related decrease in IL-2 production by stimulated PBMC or lymphocytes and IL-2 serum levels has been reported [2833]. Other authors found that intracellular production of IL-2 showed a progressive age-dependent increase in CD8+ T cells [18]. However, in other studies, serum IL-2 levels were unmodified in healthy aged people [34] and centenarians [20].

Anti-inflammatory cytokines in aging

Among anti-inflammatory cytokines IL-10 plays a central role. High levels of this cytokine in whole blood samples were found to be associated with successful aging and, particularly, with a markedly reduced risk of death from a cardiovascular event in aged people [16], but diminished resistance to infectious diseases [35]. High levels of IL-10 have been found in centenarians; this condition may result in protection from cancer [36].

IL-10 production is tightly regulated and several single nucleotide polymorphisms controlling production have been described. IL-10 low-producer genotypes seem to play a particular role in susceptibility to inflammatory diseases, together with age-related ones, whereas IL-10 high producer genotypes are involved in longevity [37].

Studies on IL-10 polymorphisms showed different results between males and females [reviewed in 38]. For this reason it seems that gender is a major variable in the genetics of longevity and that men and women probably follow different strategies to reach it. Discordant results on the association between IL-10 polymorphisms and longevity have been obtained in other populations. Therefore, cytokine/longevity associations have a population-specific component, being affected by the population-specific gene pool as well as by gene-environment interactions, behaving as survival rather than longevity genes [39].

IL1-Ra can be considered a significant predictor of mortality in elderly. A prospective population-based study, including 285 nonagenarians, demonstrated that plasma levels of IL-1Ra were higher in subjects who died during a 4-year follow-up than in those who survived [40]. However, another study on 1131 elderly Italians showed a significant age-related increase of IL-1Ra plasma levels. In long-lived individuals, this seems to be a safeguard mechanism to cope with the age-associated increased inflammatory state [41].

Contrasting data emerge from studies about the role of IL-4 on aged and long-lived people. Some authors [20] found a reduction of IL-4 serum levels and an increase of IL-13 in a group of nonagenarians, mirroring the maintenance of some effector mechanisms of the immune response in advanced ages. On the contrary, other authors [18], found that type 2 intracellular cytokines, including IL-4, increased within virgin, memory and effector/cytotoxic CD8+ T cells in aged people.

Another study determined IL-4 production in three CD4+ T-subsets—virgin, activated/memory, and effector/memory—at different ages. IL-4 positive cells appeared to increase, with age. In particular, IL-4 positive cells significantly increased in activated/memory T cells from nonagenarian subjects. The authors also found a statistically significant decreased ratio of IFN-γ/IL-4 within activated/memory T cells in old people in comparison with young people. These data suggest a dynamic shift towards an increased role of type 2 cytokines and a diminished role of type 1 cytokines in human ageing. However, it is important to remember that this phenomenon, which could be interpreted as a compensatory mechanism to the increased pro-inflammatory status, occurs within a background where type 1 cytokines are quantitatively dominant [19].

Also data reported on correlation between TGF-β levels and age are contrasting. A study reported serum TGF-β1 levels inversely associated with age [42]. However, high levels of TGF-β have been found in a group of octogenarians and nonagenarians [43] and in two different groups of centenarians [36, 44]. This condition suggests that TGF-β could both counteract and counterbalance the harmful effects of inflammaging and may result in protection from cancer. A recent study did not detect any differences in serum levels of TGF-β and TGF-β mRNA levels from PBMCs between young and old women [45].

Longevity is characterized by a balance between pro-inflammatory and anti-inflammatory agents, which act as key players. A pro-inflammatory tendency can confer high resistance against infectious diseases but, on the other hand, may increase susceptibility to inflammation-based diseases throughout life. An anti-inflammatory trend, instead, may cause an increased susceptibility to infections in pre-reproducing life and might not allow attainment of old age. The currently available knowledge is of great importance, but not sufficient to explain the secret of longevity. Data reviewed in this work are sometimes conflicting. The reason for the discrepancy is unknown, however, several factors may be involved, such as ethnic, lifestyle, cultural and genetic differences among the populations analyzed.

The studied populations are very heterogenic, the cohorts of patients present discrepancies and are not comparable. Some works were performed on healthy aged people, other works enrolled people only considering age, without considering health status and without selecting people for pathologies. Furthermore, it is known that low-grade inflammation is also associated with conditions such as obesity, diet, smoking, and physical inactivity. Therefore the balance between life style and physiological changes during aging on one hand and risk factors for age-associated diseases on the other should be taken into consideration.

However, despite all these controversial data regarding the clinical relevance of different cytokines, several studies have assigned a pivotal role in inflammatory or anti-inflammatory processes to some mediators.

According to a perspective suggested in recent years, the best candidates to become centenarians are not the strongest and most robust subjects among their age cohort, but subjects that better adapt to the environment, showing more biological plasticity. Some characteristics emerged among this group of exceptional individuals: better control of oxidative stress and remodeling of the immune system with intense anti-inflammatory activity.

Low-grade chronic inflammation is characteristic of aged people and centenarians, but long-lived are also able to avoid the main age-related diseases, thanks to the contrasting action of anti-inflammatory agents [46].

Climate change and influence on elderly allergic diseases

There is a strong correlation between global warming and greenhouse gases and the worst estimates of the past about it were completely overcome.

The principal determinant of global warming is increasing concentration of air pollutants, as underlined by a report of World Health Organization (WHO) [47]. Last report of European Environment Agency stated that up to 96% of the European Union’s urban population is currently exposed to fine particulate matter (PM) at concentrations higher than those recommended in WHO guidelines [47]. Furthermore, increased temperatures are reported as extreme events for seasonal allergy: rising temperatures recorded in recent years cause elongation and anticipation of the pollen season, increased pollen production, spread of invasive species. Increase of temperature and solar irradiation, associated with ozone gas and particulates, determines the formation of PM, including coarse (PM10) and fine (PM2.5) [48]. In normal ambient conditions PMs are a mixture of coarse (2.5–10 μm), fine (<2.5 μm) and ultrafine particles (<0.1 μm), derived from different processes, with variable chemical matters. It should also be noted that, although ultrafine fraction accounts for less than 1% of PM mass, it represents the greatest part in terms of number of particles (typically >80%) [49]. This mixture of PM causes inflammatory effects on bronchial mucosa, increasing the risk of asthma and the number of exacerbations in subjects with bronchial hyper reactivity [50]. PMs are directly involved also in cardiovascular diseases, including vasomotor and cardiac autonomic dysfunction. Moreover, hemostatic unbalance, oxidative stress and inflammatory responses have been shown to contribute to the short-term and long-term adverse effects of pollution exposure [51]. The exposure to ozone and to fine PM (PM2.5) can cause adverse health effects, including premature mortality due to cardiopulmonary diseases and/or lung cancer; recent studies highlighted that mortality rates due to air pollution are different according to geographical area, suggesting region-specific air pollution control strategies [52].

Mechanism responsible of health effects

Pathogenetic mechanisms of PM on health effects are yet matter of controversy [53], in fact atmosphere is the medium of transit for a wide variety of biogenic particles. Among biogenic particles, the bio aerosol consists of very different types of particles, such as viruses, bacteria, mold, plant fibers or pollen, with a wide distribution in size (from nanometers to a few 100 μm). These phenomena are able to activate several inflammatory mediators in respiratory tract directly acting on respiratory mucosa and indirectly promoting increase in pollens and air pollutants concentration, with consequent effects on allergic and respiratory diseases [54].

The PM pollutants from sources such as hydrocarbons, volatile organic compounds and heavy metals can generate oxidants products. Organic compounds generate an oxidative stress through redoxing cycle of quinone-based radicals, complexing of metal resulting in electron transport and depletion of antioxidants, secondary to chemical reactions between quinones and thiol-containing compounds. Metals directly support electron transport to generate oxidants, with simultaneously decreasing of antioxidants. In addition, to direct generation of oxidants by organic and metal components, cellular responses contribute to oxidative stress: reactive oxygen species (ROS) production occurs in mitochondria, cell membranes, phagosomes and endoplasmic reticulum. Oxidative stress following PM exposure determines a series of cellular reactions that includes activation of kinase cascades and transcription factors and release of inflammatory mediators, which eventually lead to cell injury or apoptosis. Consequently, oxidative stress is a central mechanism by which PM exposure leads to injury, disease and mortality [55]. Furthermore, exposition to diesel exhaust particles and ozone in human controlled trials illustrated the important role of pollutants in immunopathogenesis of both allergic mechanisms and immune responses in airway diseases, such as asthma [56].

Influence of environment in elderly allergic diseases

Further findings show that some populations, especially the elderly, are particularly sensitive to short-term ozone exposure [57], with an higher risk of PM–associated hospitalization death for this population [58].

As regards the impact of pollutants on the respiratory pathologies affecting older people, some data suggest a possible role of a changed immune response as it has been demonstrated in individuals during senescence [53]. Although, atmospheric pollution plays a major role in all age groups’ health [59, 60]. For example, it is known that, in older asthmatic patients, traffic pollution exposure is the strongest predictor of poorer asthma-related QoL [61] and high daily exposure, together with obesity and not-atopic status, is associated with poorer asthma control [62]. It is also documented that mortality and risk of hospitalization in this population is increased during more intense and longer-lasting heat wave periods. Peaks of mortality and hospitalization not only coincide with the day with highest temperature of the heat wave, but also could persist for the 5 subsequent days [63]. Furthermore, in another study, cardiovascular and respiratory diseases were analyzed in an elderly population of Moscow, as they are also related to air pollution and, in generally, are considered as major causes of hospital emergency admissions [64].

Recently, many studies focus on the potential mechanisms of air pollution in inducing cardiopulmonary diseases. In fact, air pollution seems to be associated with increased plasma viscosity, abnormality of autonomic function of the heart, including increased heart rate, decreased heart variability and increased cardiac arrhythmias [65]. These findings provide possible pathways in which air pollution affects cardiovascular system. Pollutants can indeed induce oxidative processes in mitochondria, apoptosis or necrosis of macrophages and respiratory epithelial cells, resulting in decrease of host defense to respiratory infections and in increase of respiratory tract reactivity [66]. Moreover, patients with respiratory diseases, such as chronic obstructive pulmonary disease, often have a systemic defect in their antioxidant defenses and air pollution could cause significant additional oxidative stress as response to lung inflammations [67].

Nowadays health impact derived from climate change in Europe do not regard vector-borne infectious diseases. In fact, geographical areas of several vector-borne diseases and/or of their vectors are already changing in altitude, as result of global warming. Mosquito-borne parasitic and viral diseases are among those diseases most sensitive to climate. In addition, more intense weather events create advantageous conditions for infectious diseases. For example, heavy rains permit a better survival of insect breeding sites, drive rodents out from burrows and can contaminate clean water systems [6871].

Climate changes have an impact on health due to increases in temperature, which is amplified in central parts of the cities (heat island), with a significant impact on the health of the resident population, especially for elderly and people with cardio-respiratory disease (in particular asthma and COPD). Elevated temperature reduced attendance of public areas and meeting points, such as parks or gardens. Some conditions can worsen this situation, i.e. elderly and/or subjects with chronic diseases, those with lower income, lonely subjects, immigrants and people with poor housing conditions. The reduction in hospital emergency admissions after the implementation of interventional programs supported the hypothesis of causal link between air pollution and diseases [70].

Recently, the Task Force of Air pollution and Climate Change of Italian Society of Allergy, Asthma and Clinical Immunology proposed a National Health Program to support the public administration of cities. In fact, it is possible to create gardens and parks with not allergic pollen plants, as suggested in different studies [71]. This aspect is important because the public gardens in our cities are visited by many children and elderly every day. In sunny and wind days this population is exposed to numerous pollens and the presence of allergen pollens.

In conclusion, the effect of global warming on health is documented in many documents, where the major warnings come from increased concentration of air pollution. In particular, urban population is currently exposed to fine PM concentrations, representing a daily problem for fragile population as elderly. Association among age, air pollution and climate changes were observed, confirming the particular interest in this population. Although, further studies are necessary to define better how air pollution and climate change are mainly responsible in elderly allergic diseases.

Allergic conjunctivitis in the elderly

Although the eye was reported to be the first organ involved in the allergic reaction of the first described case of hay fever almost 200 years ago, ocular allergy has never received the same attention that has been given to respiratory and skin allergy [72]. On the other hand, in view of the peculiar anatomy and the prominent representation of the immune system in the ocular tissues, the eye has always represented an extremely useful model to study the immune and allergic response to environmental and endogenous stimuli [73]. There are several reasons for ocular allergy being considered the “Cinderella” of allergic diseases, despite the fact that eye involvement represents one of the major causes of poor quality of life and may often be serious enough to affect vision. At first, it is the attitude of allergists to focus mainly on respiratory and skin manifestations, considering the eye symptoms only a complication of rhinitis, under the arguable definition of rhinoconjunctivitis. Secondly, the involvement of ocular tissues in several autoimmune and systemic diseases has often been thought to be prominent competence of other medical disciplines, such as rheumatology and internal medicine. At last, the prevalent surgical commitment of many ophthalmologists has so far made difficult a proper collaboration with allergists to focus on diseases commonly considered of minor priority in both pharmacological and clinical research and practice.

The spectrum of atopic eye diseases encompasses seasonal allergic conjunctivitis (SAC), perennial allergic conjunctivitis (PAC), vernal keratoconjunctivitis (VKC), atopic keratoconjunctivitis (AKC), atopic blepharoconjunctivitis (ABC), and giant papillary conjunctivitis (GPC). Common to these diseases are a papillary conjunctivitis and, with the exception of GPC, evidence of a type I, IgE-mediated, hypersensitivity response [74]. Although each entity has unique immunopathogenic pathways, these conditions have all been described as chronic allergic conjunctivitis (CAC).

Life expectancy around the world is growing and it has been reported that by 2030 people over 65 will reach 20% of the total population [75]. In recent years, increasing attention has been dedicated to the evaluation of allergic diseases in the elderly, often underdiagnosed and confused with signs and symptoms of the physiological physical and functional deterioration with age, as well as of their impact on well-being and quality of life [3]. However, very few data are available in literature regarding the prevalence, the role and the management of allergic conjunctivitis in the aged population [76]. Most of the reported evidence, in fact, relates to contact conjunctivitis, often associated with drug use and due to impaired lacrimation in this particular population subset. However, although allergic conjunctivitis affects mainly children and young adults, an increasingly number of cases is being diagnosed in the elderly. Atopic keratoconjunctivitis seems to be the most commonly observed clinical picture and is therefore addressed in details below.

AKC was first described by Hogan in a group of highly atopic patients who developed chronic conjunctivitis, progressive corneal vascularization and scarring [77].

Currently, there is no agreement about the diagnostic criteria for AKC [78]. One proposed definition states that AKC is a chronic ocular surface non-infectious inflammatory condition, always associated with other atopic diseases (typically atopic dermatitis, but also periocular eczema, asthma, or rhinitis), occurring at any time in the course of the associated atopic disease independently from its degree of severity and with evidence of corneal involvement.

The pathogenesis of AKC involves the production of various cytokines by effector cells which are still under investigation, as their immunopathogenic roles are not fully understood. The main effector cells in AKC are mast-cells, lymphocyte T cells, eosinophils and conjunctival epithelial cells. Once activated through antigen presentation or via pro-inflammatory stimuli, they release different preformed mediators and cytokines activating a predominantly Th1 cell response (i.e. IL-2, IFNγ IL-12 and IL-8). A Th2 response is also present in these patients, but to a lesser degree. Furthermore, a peculiar feature of AKC is the increased susceptibility to infection resulting from the compromised innate immunity as demonstrated by patients’ deficiencies in keratinocyte-derived antimicrobial peptides and in both sweat and tears IgA. This results in eye infections with Staphylococcus aureus, as well as Herpes simplex virus.

The symptoms of AKC are characterized by itching, watering and a stringy mucoid discharge. Patients with AKC may have prolonged exacerbations of disease that are difficult to control and which are associated with redness, severe photophobia, extreme discomfort/pain and blurring of vision. The disease is bilateral and symmetrical. The tarsal and fornix conjunctiva are affected in all forms of the disease and are chronically inflamed. Papillary hypertrophy of both the upper and lower tarsal conjunctiva is common in the early years of the disease and giant upper tarsal papillae may occur in some patients. The conjunctiva is often so thickened by infiltrate that the tarsal vessels are obscured. Later in the course of the disease the papillae may be largely replaced by sheet scarring. Trantas dots may occur transiently at the limbus, and on the tarsal conjunctiva, during exacerbations of the disease and are usually associated with limbal swelling. Severe inflammation results in punctate corneal erosions and filamentary keratitis, which can progress to frank corneal erosions and a shield ulcer with a mucous plaque within 24 h. Plaques may result in corneal thinning, vascularization and perforation. Patients often have severe eczema of the eyelids and increased pigmentation of periorbital skin (panda eyes). There may be, at last, an associated atopic blepharoconjunctivitis.

The clinical features together with a personal or family history of atopic dermatitis are usually sufficient for the diagnosis. Laboratory investigations are needed where the diagnosis is uncertain. Diagnostic dilemmas may occur in patients who do not respond to topical therapy and in the presence of conjunctival scarring to differentiate AKC from other causes of conjunctival scarring associated with chronic conjunctivitis such as primary Sjogren’s syndrome, ocular rosacea, ocular mucous membrane pemphigoid and surface neoplasia. Relevant ophthalmic investigations include: ocular cytology, upper tarsal conjunctival punch biopsy and tear IgE assessment. Confocal microscopy may represent an additional tool for helping to refine the diagnosis when uncertain. Specific serum IgE and skin prick tests have poor value in the diagnosis.

The management of AKC is often a therapeutic challenge because of the chronicity of the disease punctuated by exacerbations of keratoconjunctivitis and complicated, especially in the elderly, by secondary infectious and herpetic keratitis, glaucoma, cataract and keratoconus [79]. The goals of treatment are to achieve symptomatic control, reduce the frequency and morbidity of corneal complications and minimize the side effects. Atopic keratoconjunctivitis therapy may be divided into conservative measures and higher risk second-line therapeutic agents [80]. Conservative measures include cold compresses, lubrication with preservative free drops, and the use of topical cromones. The addition of topical or oral antihistamines may be helpful in relieving the symptom of itch in some patients. Patients with more severe disease are refractory to these treatments and require the use of second-line topical therapeutic agents, starting with prolonged courses of topical or oral steroids to control the inflammation. The disadvantage of these agents is that they often achieve symptomatic and inflammatory control, but at the expense of steroid side-effects including raised intraocular pressure, cataracts, arterial hypertension and osteoporosis. The addition of immunosuppressive agents such as cyclosporin A and tacrolimus has been described in the treatment of AKC. Although these agents are effective and may be useful as steroid-sparing agents, they need to be prescribed carefully in view of an increased risk of local and systemic infections, particularly in aged people whose immune response may be already impaired. The use of newer biological therapies is currently being explored [81]. Agents such as infliximab (anti-TNFα), alefacept (T-cell inhibition), and rituximab (anti-CD-20) have proven to be effective in the treatment of atopic dermatitis and may have a role in patients with AKC unresponsive to conventional systemic immunosuppressive therapy. Topical or systemic antibiotics are, at last, useful for managing ulcerative blepharitis.

Allergic asthma in the elderly

Asthma in the older adults has become a hot topic in respiratory medicine because it poses intriguing questions in terms of pathophysiological mechanisms, and challenges the traditional diagnostic approach and therapeutic algorithm that are commonly applied to younger populations. One of the main complicating factors lies in the fact that the respiratory system is particularly susceptible to time-related deterioration in structure and function [82], approaching a condition that is often confused with that of chronic obstructive bronchitis or emphysema. The prevalence of asthma in older populations is not different from that of younger age groups; however, geriatric asthma presents with clinical and functional features that make its management a potential complex and complicated task; among them, the atopic condition, or more precisely, the lack of it. The question is not whether asthma in the elderly is a rare disease, but rather whether allergic asthma is a rare entity in the elderly. The immune system undergoes an involution process with increasing age; as a consequence the immunoglobulin production, including IgE, declines leading to the misconception that asthma in the elderly has a non-allergic pathogenesis. The clinical implications of this phenomenon are that atopy is relegated to a corner in the assessment of the geriatric respiratory patient. This has greatly contributed to underdiagnosis and misdiagnosis of asthma in the most advanced ages.

Atopy refers to the predisposition to develop allergic diseases, such as allergic asthma, and describes the ability to mount IgE responses to common allergens. The initial observations on general population samples of a reduction of atopy with age reinforced the theory that allergic asthma is a rare nosologic entity in the elderly. With the aim of providing a definite answer to this doubts, Scichilone and colleagues [83] conducted a systematic revision of published articles on the topic, and found that, overall, the prevalence of allergic sensitization is lower in the most advanced ages. These findings were confirmed by a survey conducted on large European population [84], which showed that positive skin prick test for all allergens tended to be less common in older adults than in younger ones. It must be stated, however, that the published literature in the field suffers from the lack of longitudinal studies to confirm the reduction in the atopic condition with ages. In other words, a lower prevalence of atopy in the elderly as demonstrated by cross-sectional observational studies could simply reflect a lower prevalence of atopy in the same individuals at younger ages. Another factor to be kept in mind is that, when defining atopy, increased total IgE levels and positive skin prick test to allergens are used interchangeably, whilst the use of specific IgE towards aeroallergens could be more appropriate. A distinction should be made between asthma occurring for the first time in older ages (late-onset asthma), and asthma that begins in younger ages (early onset asthma). The former is historically believed to have a lower likelihood of allergic features [85]. The assessment of the features of late onset asthma is complicated by the fact that some investigations included as “late” asthma cases presented in adulthood. In addition, the so-called “recall bias” could have affected the definition of asthma in the elderly as late-onset rather than long standing.

Persistent antigen exposure characterizes the natural course of allergy: the continuous stimulation with the same antigenic proteins leads to the expression of new surface molecules together with the expansion of effector and memory cells. This translates into a decrease in the number of naive cells and an increase in the number of sensitized cells. Therefore, in allergic asthma the naive T cells tend to decrease whereas the T lymphocytes are constantly stimulated by the aeroallergens. Of note, age also affects mast cell and basophil mediator releaser and the responsiveness of other inflammatory cells to the chemoattractive and activating signals of mast cell and basophil mediators, which influence the inflammatory cascade that follows the IgE receptor bridging.

The haematopoietic compartment of bone marrow is replaced by fatty adipose tissue with increasing age [86], with a functional decline in stem cell precursors which, however, seems to occur only under stressing conditions; in basal conditions the absolute numbers of eosinophils and basophils in young versus older subjects does not differ [87]. An old investigation carried out in elderly subjects showed that paracortical and medullary zones of lymph nodes were strongly reduced, with lower germinal centres [88]. Furthermore, aging is associated with changes in the function of lymphocytes, such as a lower antibody response, a decreased ability to produce high affinity antibodies and reduced IgG isotype class switching [89]. Increasing age is also associated with an attenuation of dendritic cell function. Agrawal et al. [90] reported that peripheral blood dendritic cells are similar in young and aged subjects, but migration and phagocytosis are impaired in older adults. Aging is characterized by macrophage dysfunctions consisting in the reduction of expression of TLRs, secretion of cytokines following activation, and phagocytic ability [91].

Scichilone and colleagues [83] point out that, despite lower prevalence of atopy in the elderly, the prevalence of the allergic component remains high in older individuals suffering from asthma, and this may have clinical consequences. For example, the age-related reduction in the production of IgE does not decrease the efficacy of a pharmacological approach with anti-IgE in the severe forms of asthma occuring in older ages. Although less frequent, the allergic reactions in older adults can be even worse than in younger people, because of the inability of other organs and systems to compensate. For example, a systemic anaphylactic reaction may have dramatic consequences in elderly subjects in whom the cardiovascular system is not able to effectively compensate. In addition, atopy can interact with other conditions (primarily viral infections), eliciting more severe exacerbations in the elderly. Regardless of age, the assessment of atopy is crucial in the comprehensive evaluation of the chronic respiratory patient, in that, it provides the unique opportunity to act on the environment (removal of allergens) and to modify the natural history of the allergic condition with allergen immunotherapy. Indeed, age per se does not preclude the use of allergen desensitisation.

The relationship between the presence of the allergic component and the geriatric age may be complicated by the incorrect interpretation of skin testing. This may be due to the age-associated reduction in skin reactivity of the elderly to histamine and allergens. In addition, the area of the skin where the test is performed could be atrophic with decreased cellularity, thus influencing the response to allergens. The reduction in blood vessels and mast cells reduces the binding sites for allergen and the release of histamine to produce wheal and flare. Finally, mast cells function can be impaired by prolonged exposure to sun, as often occurs in the elderly. A negative skin prick test does not rule out completely the presence of an allergic condition. The concept of localized mucosal allergy with the production of specific IgE antibodies in the absence of atopy has been put forward, and is currently an object of investigation. Taken together, these observations allow to speculate that the immunological and inflammatory responses to stimuli may be different in the older ages, implying a different history of the disease. It is logical then to hypothesize that asthma in the elderly will have to be treated accordingly.

Allergen immunotherapy in the elderly

General aspects of AIT

Allergen (specific) immunotherapy (AIT) was introduced in clinical practice more than one century ago, with the supposed aim of “vaccinating” against hypothetical “aerogenic toxins”. Despite the rationale was wrong, the procedure resulted to be clinically effective and rapidly spread. Subcutaneous injections (SCIT) remained the only mode of administration for more than 70 years, when new modalities were proposed, with the aim of improving the safety and convenience. Among the various routes proposed, the sublingual one (SLIT) rapidly gained credibility, so that it was accepted as a viable alternative to SCIT in all official documents and guidelines [92]. In general, the clinical efficacy of SLIT and SCIT are equivalent, although SLIT has a more favourable safety profile [93].

To date, the practice of SCIT is sufficiently standardized, as testified by position papers and practice parameters [9497]. On the other hand, SLIT can be administered as drops, monodose vials or tablets, with variable timings and doses depending on the manufacturer. In the last decade, highly standardized products in tablets (grass, mite, ragweed) have been approved as drugs by EMA and FDA. The aim of AIT is of interfering with the immune response to the offending allergen, thus inducing a tolerance that results in a reduction of symptoms and medication intake upon natural exposure to the allergen itself. SCIT usually consists of an updosing phase (with gradually increasing doses of the allergen) followed by a maintenance phase, were the maximum dose is given at regular intervals (usually monthly) for 3–5 years. With SLIT, due to the favorable safety profile, the updosing phase is absent or very short, and the maintenance is given on daily basis.

Mechanisms of AIT

Differently from traditional medications, AIT is not a mediator or receptor inhibitor but rather a biological response modifier. Its mechanisms of action are multiple and complex, and exerted at various levels of the allergen-induced inflammatory reaction. The complexity of the immune-mediated actions of AIT have been extensively described in many reviews [98100].

Briefly, the sensitizing molecules contained in the allergen extract (independent of the mode of administration) are recognized by antigen presenting cells, processed and presented to Th lymphocytes. This results in the activation of T regulatory (Treg) cells which, through the production of numerous cytokines (IFN-γ, IL-10, IL-12) induce:

  • A relative shift from the Th2 towards the Th1 phenotype (allergic subjects have an imbalance in the Th2/Th1 lymphocyte in favour of Th2). This reduces the production of IL-5, IL-4, IL-13 that specifically favour the allergic inflammation.

  • A decrease in allergen-specific IgE synthesis.

  • An increased production of allergen-specific IgG4, which block the IgE-facilitated antigen presentation.

All these modification are not permanent, but they are long-lasting, as testified by the fact that the clinical benefit obtained in a successful course of AIT may persist for years after discontinuation. In addition, the disease-modifying effect can affect the natural history of allergic diseases (for instance reducing the risk of asthma onset) [101].

The “problem” of AIT in the elderly

As a matter of fact, the use of AIT in elderly people has been until the last decade a neglected aspect. Looking at the available literature, and particularly at randomized trials, it appears that patients aged 65 years or older are never included in the study population [9496]. Guidelines are usually elusive. Some of them indicate older ages as a relative contraindication to AIT [102], some others do not mention this aspect [97], and in others caution is recommended, especially concerning the problem of safety and the treatment of severe reactions [103]. This fact is the consequence of two consolidate beliefs. First, allergic diseases are expected to be relatively rare in aged patients and, second, it is thought that with ageing the immune system loses its ability to efficiently respond with specific IgE. On this background, AIT has never been considered a therapeutic option for older people. The context is further complicated by the frequent presence of comorbidities, and consequent poly-pharmacotherapy, that can interfere with AIT (e.g. ACE inhibitors, betablockers or major antidepressants).

Indeed, in the recent years it has become clear that the allergic respiratory diseases are not a rare situation in elderly people [104]. In fact, the prevalence of respiratory allergy (asthma and/or rhinitis) have been shown to range between 5 and 10% in various studies [105, 106]. Lombardi et al. [107] have recently shown that the prevalence of allergic sensitizations with clinical manifestations in older subjects approximates 50%. In another cross-sectional study, involving about 2000 elderly patients with a clinical suspicion of allergy, the prevalence of asthma, seasonal rhinitis and perennial rhinitis were found to be 5, 13 and 17%, respectively [108]. Thus, despite the problem is considered of scarce relevance, the available data testify for a not negligible social impact. Concerning the immune system, the phenomenon of “immune-senescence” is well known since many years. It is characterized by an overall reduction in: bone marrow proliferation, antigen-presenting function, capacity of mounting an antigen-specific humoral response [89, 109, 110]. This would result in a decreased capacity of synthesizing IgE. It is true that in several studies, the acquired immunity seem to decline with age [for review see 84]. On the other hand, several studies have demonstrated that the Th2-driven response in the elderly does not differ from that of younger subjects [111, 112] and that the production of IgE does not always decline [113]. In addition, the nasal cytology aspect in elderly subjects with allergic rhinitis is identical to that of young patients [114], and a local nasal specific IgE response is detectable also in aged patients [115].

The evidence

Due to the above mentioned limits, it is easy to understand that there are very few clinical trials exploring the effects of AIT in elderly subjects. In a nonrandomized open controlled study of SCIT for birch and ragweed allergy involving a total of more than 100 patients, the clinical benefit was identical in patients aged less or more than 55 years, and greater than in controls not receiving SCIT [116]. Subsequently, other Authors [117] performed a retrospective study where the effects of SLIT (symptom/medication scores, bronchial hyperreactivity, pulmonary function) were compared between young adults (18–55 years, n = 49) and older patients (55–65 years, n = 40). They found no difference between the two age groups in the magnitude of the achieved clinical efficacy. More recently, Bozek et al. [118] explored the effects of SLIT in 108 patients aged 60–75 years and receiving dust mite SLIT or placebo for 3 years. Main findings were that total nasal scores decreased by 44% VS baseline in the SLIT and by 6% in the placebo group and medication score decreased vs baseline 35% in SLIT group, although no change was observed between groups at the specific nasal challenge. The same Authors assessed the efficacy of pre-seasonal grass pollen AIT in subjects aged 65 years or more and suffering from seasonal rhinitis [119]. Sixty patients were randomized to receive AIT or placebo for 3 years, and the area under the curve for symptoms + medications score was evaluated at the 3rd year. The reduction of this parameter versus baseline was significantly greater in the active group, and the symptom score, medication score and combined decreased by 50% on average.

Polytherapy may represent a problem in selected cases and with selected drugs that in principle can interfere with the course and treatment of severe reaction if any occur. For instance, beta blockers may counteract and monoamine oxidase inhibitors may potentiate the effects of epinephrine, when it is given for anaphylactic reactions (one of the possible although rare side effects of AIT). Indeed, the new selective betablockers do not carry this risk, and in fact have been indicated only as relative contraindication in the more recent literature reviews, whereas monoamine oxidase inhibitors do not represent a contraindication. The same holds true for ACE inhibitors, for which the risk of worsening hypotension in anaphylaxis has not been substantiated [97, 120]. Cardiovascular diseases are not per se a contraindication to AIT as well. Venom AIT represent a special case, since the risk of anaphylaxis after sting in sensitized subjects (especially the elderly) is always greater than any possible risk due to cardiovascular diseases or drugs interfering with epinephrine.

In conclusion, there is increasing evidence that allergic disorders in the elderly are not rare, and an efficient IgE response can be expected also in this age group. On the other hand, the few evidence so far available for AIT, consistently confirm that the treatment is effective also in older subjects. Thus, provided that the IgE-mediated nature of the disease is clearly ascertained and that the offending allergen is unequivocally identified, AIT can be prescribed also in the elderly, with the same indications and modalities applied in young adults. In the elderly, additional care should be paid in assessing the presence of comorbidities, the use of multiple drugs, and the expected compliance.

Chronic urticaria in the elderly

As all organs, the skin ages with structural and functional consequences that may lead to clinical conditions. The “skin aging” hallmarks are: (1) atrophy of the epidermis and dermis due to loss of hydration; (2) progressive loss of function and structural integrity resulting in impaired immune response and skin barrier function; (3) skin vascular impairment; (4) metabolic imbalance of reactive oxygen species, and components of the extracellular matrix [121]. Aged skin is characterized by atrophy, wrinkling, fragility, alterations in pigmentation, a higher frequency of benign and malignant tumours, and a greater tendency to xerosis [122]. These factors contribute to greater susceptibility to dermatologic diseases in individuals over the age of 65 years. In this context, one of the most common skin diseases is the allergic contact dermatitis (ACD), which is mainly due to the direct exposition to nickel sulphate (11–12%) and fragrance/balsam of Peru (7–9%). Atopic dermatitis (AD) is much less common in the elderly in comparison with children and young adults. AD is associated with seasonal mucosal allergies, asthma and positive prick tests to various allergens [123]. Late onset AD, without the usual history of atopy, may explain eczema of unknown origin and negative patch tests. Scabies should enter the differential diagnosis in generalized dermatitis, as institutional acquisition of scabies is common in the elderly.

Chronic urticaria (CU) is a common nosological entity in older individuals. This is especially true for the spontaneous form of the disease, although there are few data regarding epidemiology and clinical features. Urticaria is defined as itchy wheals with or without angioedema that usually persists for less than 24 h. According to current EAACI/GA2LEN/EDF/WAO guidelines, CU can be classified as spontaneous, physical, or other [124]. Chronic spontaneous urticaria (CSU) is the most common subtype of all forms of non-acute urticaria, and is characterized by wheals that develop independently of external stimuli and last for a minimum of 6 weeks. In general, the underlying causes of CSU are difficult to identify in most patients. It is estimated that about 0.5–1% of the population suffers from CSU, and that about one quarter of the population has experienced urticaria at some point during their lives. Both sexes can be affected, but in general, females appear to suffer from urticaria nearly twice as frequently as males.

The pathogenic mechanisms of CSU are unclear. Evidence of an autoimmune etiology is reported in about 45% of CSU patients, remaining unknown in the remaining subjects. Circulating autoantibodies specific to high-affinity immunoglobulin E (IgE) receptors or dermal mast cell bound IgE activate mast cells and induce degranulation with cytokine release [125]. The autologous serum skin test (ASST) is a screening test for autoreactivity; if positive, it suggests the presence of circulating histamine-releasing factors of any type, and not only of functional autoantibodies. Atopy is proposed to play some role in the pathogenesis of CSU, especially in the aspirin-intolerant CU phenotype. Several hypotheses exist regarding the relationship of atopy and aspirin intolerance; however, the exact association remains unclear.

Systemic diseases that may induce elderly urticaria should be carefully evaluated. Findings derived from the Mayo Clinic’s electronic database revealed that patients presenting a new diagnosis of CU at older ages were more likely to have underlying monoclonal gammopathy of undetermined significance (MGUS). Some authors have also emphasized the role of drug-induced urticaria in the geriatric population [126].

Angioedema in the absence of urticaria can be due to overproduction of bradykinin [127]. It is exceptional that hereditary deficiency in the C1 inhibitor (HAE-C1-INH) has its onset in the elderly. It is more frequent that acquired C1 inhibitor deficiency (AAE-C1-INH) presents in the older age, and it is characterized by activation of the classical complement pathway and accelerated catabolism of C1-INH due to lymphatic tissue neoplasms or autoimmune diseases. The prevalence of angiotensin-converting enzyme inhibitor angioedema (AE-ACEi) is relatively high, ranging 0.1–2.2%, and it should be suspected in all patients with AE who are receiving ACEi. Normal levels of complement factors help to reinforce the clinical suspicion and to rule out the possibility of AE with C1-INH deficiency.

In a retrospective investigation conducted on a large cohort of patients with CU from the National Health Insurance Research Database of Taiwan, Chen and colleagues found that a quarter was in the age range of 60–79 years and 3.4% were 80 years and older [128]. A study performed by Magen and collaborators on 1598 adults suffering from CSU found that 9.4% were elderly [129]. Interestingly, the latter showed lower rates of angioedema and dermographism, as well as fewer wheals and ASST positivity. At variance with the younger populations, no sex differences were demonstrated in the elderly, whereas comorbid conditions, such as diabetes mellitus, chronic renal failure, hypertension, Hashimoto’s thyroiditis, and malignancies were more prevalent in the elderly [129].

Ventura et al. [130] found that CSU due to infections was more common in the elderly, especially the CSU forms related to Helicobacter pylori and those related to the sensitization to the parasitic nematode Anisakis simplex. Ban et al. [131] reported that in the elderly with CU there is a high prevalence of AD compared with younger groups. The prevalence of AD was equal to 37.8 versus 21.7%, respectively (p = 0.022). Disorders of the skin barrier are found in subjects suffering from AD and this leads to the colonization of Staphylococcus aureus, the degraded ceramide of which penetrates the skin barrier and induces IgE sensitization.

Elderly CSU diagnosis

As a general concept, the diagnostic approach to CSU is not influenced by age. Indeed, the international guidelines do not recommend a specific work-out for elderly populations, although a careful assessment of the drug history should be encouraged (especially for aspirin and ACE inhibitors), and the presence of autoimmune or neoplastic diseases ruled out [132]. An extended laboratory and instrumental tests should be reserved to specific conditions as judged by the physician.

Elderly CSU treatment

The avoidance of causal and/or triggering factors is the first choice in the management of CSU. This is however a rather complicated task, given that in the majority of cases the recognition of these factors is unsuccessful. The pharmacological treatment of CSU raises a safety issues mainly due to concomitant diseases and polypharmacotherapy, which are common in older subjects and may cause drug-to-disease and drug-to-drug interactions. Indeed, the potential for drug interaction increases with age and with the number of drugs prescribed [133]. According to the international guidelines, the management of CSU is based on a step-wise approach, with sequential steps to be implemented according to the clinical response. The mainstay treatment for CSU in the elderly consists of new-generation non-sedating antihistamines. First generation H1 receptor antagonists are lipophilic and therefore may cross the blood–brain barrier. For this reason, elderly individuals may be at increased risk of adverse effects involving the CNS (confusion, sedation, dizziness, sleepiness, and impaired cognitive function). Agostini et al. [134] showed that the administration of diphenhydramine in elderly hospitalized patients was associated with increased risk of cognitive decline compared with non-exposed patients. Because of the lack of specificity for the H1 receptor, due to activation of dopaminergic, serotoninergic, muscarinic and cholinergic receptors, the first-generation antihistamines have also additional adverse effects with an higher risk of urinary retention, constipation, as well as arrhythmias, peripheral vasodilatation, and postural hypotension. First-generation antihistamines should therefore be prescribed with extreme caution in elderly patients. Second-generation oral H1 anti-histamines have a safety profile with reduced capacity to induce CNS-related adverse effects as they have a low potential to cross the blood–brain barrier, and provide selective H1 receptor blockade without anticholinergic activity. It is instead important to remember that second-generation oral H1 antihistamines potentially requiring a dose reduction in patients with hepatic diseases or dysfunction include cetirizine, ebastine, levocetirizine, and loratadine. Those potentially requiring a dose reduction in patients with renal dysfunction include cetirizine, ebastine, fexofenadine and levocetirizine. Conversely, no dosage adjustment for desloratadine is required in the elderly healthy subjects [135]. Pharmacokinetic/pharmacodynamic studies in special populations indicate that bilastine’s dose adjustment is not necessary in elderly patients, or in hepatic or renal insufficiency [136]. The safety of second-generation oral H1 receptor antagonists up-dosing has not been systematically evaluated in the older population.

With regard to the third-line therapy for CSU, very few studies explored the effect of montelukast in the elderly. Available data reassure on the safety profile of montelukast in the elderly patients, and dose adjustments are not necessary even in individuals affected by renal or hepatic failure. It must be stated, however, that montelukast can interact with drugs that interfer with the CYP3A4, CYP2C8, or CYP2C9 systems.

Alternative treatment options have not been systematically evaluated in elderly patients with CSU. Corticosteroids and cyclosporine could be used with caution in cases resistant to therapy. Old age may be a risk factor for particular corticosteroid-associated side effects and adverse reactions (diabetes, cataract, and osteoporosis).

The anti-IgE humanized monoclonal antibody (omalizumab) has been successfully tested in subjects with CSU up to 75 years of age, namely 4–9% in the study of Maurer and colleagues [137], and 3.7–5.2% in that of Saini and colleagues [138]. Kaplan and coauthors showed that anti-IgE treatment was effective irrespective of age [139]. No omalizumab dose adjustment is recommended for elderly patients or those with impaired renal or hepatic function, and there is no evidence of clinically significant drug interactions [140]. The literature contains sporadic reports of older patients with difficult-to-treat CSU and/or chronic inducible urticaria treated with omalizumab; in one case the highest age was 82 years. Nowadays, specific data are not available on omalizumab treatment in elderly patients with CSU.

In conclusion, CSU in the elderly induce severe disability and decrease in quality of life and CSU can be associated with other diseases (autoimmune, infectious, neoplastic, etc.). Polypharmacological treatment is very frequent in the elderly patients, and should be taken into account in the treatment of CSU, as it can induce drug interaction due to the various comorbidities and changes in pharmacokinetics and pharmacodynamics. Like adults and children, the mainstay treatment for CSU in the elderly consists of new-generation non-sedating antihistamines.

Food allergy in the elderly

The incidence of IgE-mediated allergic reactions is rising worldwide. The changes of the demographic parameters let foresee such a meaningful increase in the proportion of allergic patients in the next few years, so that the possibility of missed recognition or under diagnosis should be avoided [141]. In particular, a recent paper on food allergy carried out in a geriatric nursing home on subjects with a mean age of 77 years showed that 40% of the patients had specific IgE to respiratory allergens and 24.8% to food allergens, with a direct correlation to the positivity of skin prick tests (SPT). According to the authors the problem of food allergy (FA) in the elderly is underestimated, because epidemiological studies are focused mainly on children and on adults, as if the elderly could not be affected by these allergies [142].

An incidence of 5% FA in people over 65 was diagnosed in outpatients of the Allergy Section at Hospital Vall d’Hebron, in comparison to the 36% diagnosed in people aged between 40 and 65 and to the 6 9% in the youngest [75]. The profile-sensitization was not much different among the groups, except for the Rosaceae fruits and fish, which were found to be much more frequent in younger people [75]. In addition, data from the literature show that with regard to the clinical manifestations anaphylaxis from food allergens is less frequent in the elderly in comparison with young subjects [143]. Another recent work has shown that in a geriatric Immunoallergology Unit one-third of the patients over 65 who were afferent to the structure and had been visited and subjected to specific allergy tests for suspected food allergy were positive to SPT to food; it confirmed the diagnosis of FA [76].

To understand the reasons of FA occurrence in the elderly it is appropriate to reevaluate the physiological changes that occur in the mucosal immune system. In fact it should not be forgotten that the gastrointestinal tract is the largest immunologic system with an important number of lymphocytes; part of them are isolated and part of them are aggregated in lymphatic structures called Peyer’s patches. In addition, this system plays a key role in all reactions involving mucosal immunity, including FA. It takes particular importance in the elderly due to the physiological involution of the thymic gland and its related functions [144].

As a matter of fact it is in the intestinal mucosa and in the immune related tissue, the so called gut associated lymphoid tissue, that occur the modifications which are responsible for the change in the oral tolerance. Generally the oral tolerance that is established at a young age is maintained even over 65, unless new antigens which are unknown to the immune system [145] are introduced. As a consequence, the induction of tolerance to new antigens is reduced in elderly mice. These data show clearly that the answers of the Secretory Antigen-specific IgA (S-IgA) and oral tolerance is modified in aged mice; even with regard to the protection from infectious diseases, some alterations that should be better investigated can occur especially in humans [146]. Another important factor responsible for changes in oral tolerance is the alterations of the gastrointestinal permeability. The intestinal barrier integrity interruptions may arise after the appearance of alterations in the gastroenteric mucosa [147]. They occur mainly at the level of tight junction (Tj) for alterations of both canonical and non-canonical activation of NK-κB pathways in intestinal epithelial cells [148]. These structures, that generally provide a “barrier” effect, become more permeable both for the alteration of mucosal epithelium due to inflammatory diseases of the gastrointestinal tract and for the effect of some cytokines whose secretion is increased in the elderly, such as IL-6, TNFα and IL-β, responsible for a “pro-inflammatory” status, the so called “inflamm-aging” [13, 149]. In particular the epithelial cells, which are responsible for the production of large amounts of cytokines, including the IL-β, increase the permeability of the mucous membrane through the reduction of the proteins of the Tj and occludens zonula [150]. This weakening of the “barrier effect” results in a modification of the mechanisms of oral tolerance and thus predisposes to FA. Another key factor is the presence of inflammatory cytokines at this level. A recent study confirmed it proving that in the biopsies of the intestinal mucosa of old baboons there is an up-regulation of micro RNA, miR-29a and of inflammatory cytokines IFN-γ, IL-6 and IL-1β[1511]. The role of S-IgA should not be forgotten too. These immunoglobulins play a critical function both for the maintenance of the intestinal microbiota and consequently for the modulation of local immune responses [151].

With regard to the IgA there are conflicting studies about the maintenance of an optimal level of IgA in the gastroenteric tract in the elderly. Although the IgA reduced levels can rather reflect an alteration of the migration of plasma cells secreting IgA from effector to inductor sites, rather than their actual numerical reduction [152]. However it is reported that Antigen-specific responses of IgA type are weaker in elderly animals. In addition, young mice have less somatic mutations of IgA if compared to older mice, and the analysis of more than a million VH sequences showed that the IgA repertoire of the young differs from that of the elderly [153]. It is not yet fully clarified how these differences can result in the decreased efficacy of the security role exerted by the IgA in humans.

Moreover, the habit of eating raw fish, common in Italy and in other countries, is connected to the possibility of developing allergic sensitization to Anisakis simplex, whose incidence increases over the years because of the increase of exposure [154]. This sensitization is expressed with typical clinical manifestations of FA including especially hives, but also more severe reactions such as asthma and anaphylactic shock [155]. In particular it has been shown that even in this case a situation of impaired gastric permeability, which can promote massive penetration of allergens and induce important clinical manifestations, can also coexist [156].

According to Diesner et al. [142] during senescence the changes that occur in the innate immune system, as well as in adaptive, justify the appearance of FA. At the same time the lack of some micronutrients, including zinc and iron or vitamin D may be a risk factor. In particular the reduced level of zinc, frequent in elderly subjects, is responsible for the reduction of Th1 cytokines, while it does not interfere with the production of Th2 cytokines, that promote the increase of allergic diseases in the elderly [157]. The reduction of iron, which occurs often in the elderly, is another risk factor. In fact, it is responsible for a diminished antibody response, especially with regard to the IgG4 subclass that can prevent activation of effector cells through the capture of allergens before they can act the IgE-mediated binding [158].

Even vitamin D could play a role through T cells and antigen presenting cells by promoting tolerance via inhibition of inflammatory responses and the induction of regulatory T cells [159]. Another contributing factor is represented by atrophic gastritis, which is a frequent disease among the elderly, or from the excessive use of alcohol, proton pump inhibitors or antacids. As a consequence undigested proteins that take on allergenic properties would remain in the stomach triggering FA [160].

In conclusion, FA represents an emerging problem in the elderly, complicated by gastrointestinal mucosa permeability and impaired function of the local immune system.

Drug allergy in the elderly

We define “adverse drug reactions” (ADRs) any untoward reactions to a medication. ADRs may be broadly divided into two types: A (reactions occurring in most normal patients, given sufficient dose and duration of therapy: common and predictable) and B (drug hypersensitivity reactions restricted to a small subset of the general population: rare and unpredictable). A drug allergy (DA), i.e. an allergic drug reaction, is an adverse drug reaction that results from a specific immunologic response to a medication, therefore a Type B reaction (10–15% of ADRs). It is a reaction which cannot be usually predicted and occurs in a susceptible kind of patients. There are exceptions: predictable hypersensitivity reactions to abacavir, dapsone, carbamazepine, allopurinol end flucloxacillina in patients with certain leucocyte antigen (HLA). Signs and symptoms are different from the pharmacologic actions or the collateral effects of the drug. DA account for about 6–10% of all ADRs, but for up to 10% of the fatal reactions [161]. The term “immunologic drug reaction” is also used to describe these reactions. Immunologic drug reactions can be mediated either by humoral immunity (IgE, IgG), or by lymphocytes. There are different systems for classifying DA. One is based upon the immunologic mechanism and identifies four categories (Gell and Coombs): Type I, immediate in onset and mediated by IgE and mast cells and/or basophils, Type II, delayed in onset and caused by antibody (usually IgG)-mediated cell destruction, Type III, delayed in onset and caused by IgG-drug immune complex deposition and complement activation and Type IV, delayed in onset and T-cell mediated (Type IV reactions may be further subdivided into IVa, IVb, IVc, and IVd). Types I and IV reactions are far more common than types II and III, the latter usually following prolonged, higher dose therapy. Most medications cause just one type, although certain drugs, such as penicillin, or glucocorticoids can induce all types [162]. A second classification is based upon the timing of symptom onset. The WAO has recommended that, based upon the timing of the appearance of symptoms, immunologic drug reactions be divided into immediate reactions (onset within 1 h after exposure) and delayed reactions (onset after 1 h) [163]. However some IgE-mediated reactions may appear after 1 h, particularly if the drug is assumed per os and especially if together with food. Usually, delayed reactions begin after weeks of continuous treatment. The reaction, also called “drug-induced hypersensitivity syndrome” (DiHS), is characterized by fever, rash, and multi organ involvement, and may or may not be associated with eosinophilia and lymphocytosis. It can persist for weeks to months, even after the medication is stopped. One such disorder is “drug rash with eosinophilia and systemic symptoms” (DRESS), a systemic drug reaction that begins 1–12 weeks into continuous treatment [164]. The liver (hepatitis) and heart (hypersensitivity myocarditis) may be affected.

While the prevalence and the risk factors associated with the ADRs in the general adult population have been well documented, much less is known about the ADRs, and in particular about the DAs, in the elderly population.

A number of factors in older individuals contribute to their increased risk for developing a drug-related problem. These include female sex, frailty, coexisting medical problems, memory issues, and use of multiple, interacting, prescribed and non-prescribed medications [165].

A study of Alhawassi et al. [166] review the Literature to estimate the prevalence of the ADRs in the elderly in the acute care setting and to identify the factors associated with the increased risk in the elderly. The mean prevalence of the ADRs in the elderly in the studies included in this review was 11.0% (95% confidence interval [CI] 5.1–16.8%). The median prevalence of the ADRs leading to hospitalization was 10.0% (95% CI 7.2–12.8%), while the prevalence of the ADRs occurring during hospitalization was 11.5% (95% CI 0–27.7%). There was wide variation in the overall ADRs prevalence, from 5.8 to 46.3%. The Authors conclude that the ADRs constitute a significant health issue for the elderly in the acute care setting. While there was wide variation in the prevalence of ADRs in the elderly, according to this study about one every ten admissions of elderly patients is due to ADRs and 10% elderly patients will suffer from an ADR during the hospital stay. Older female patients and those with multiple comorbidities and medications appear to be at the highest risk of ADR in the acute care setting.

Drug-related hospitalizations account for 2.4–6.5% of all medical admissions in the general population; the proportion is much higher for older patients [161, 167]. In the United States, it is estimated that annually from 2007 to 2009 there were 99,628 emergency hospitalizations for adverse drug events in individuals aged 65 years or more, and two-thirds of these were due to unintentional overdoses [168]. A meta-analysis found a fourfold increase of the rate of hospitalizations related to adverse drug events in older adults compared with younger adults (16.6 versus 4.1%); it was estimated that 88% of the ADR hospitalizations among older adults, and 24% among younger adults, were preventable [169].

A meta-analysis on UK hospitals found that the frequency of drug-related hospitalizations was between 2.4 and 6.2% [170] and many of these were considered preventable [171]. A recent study in the Netherlands [172] that about 41,000 hospital admissions per year were related to ADRs and about the half were potentially preventable. The risk was doubled if patients were 65 or older. The elderly are particularly at risk for drug-related problems [173]. About 30% of hospital admissions of patients aged 65 and over are due to ADRs, most of which are preventable [174]. It is also of concern that the likelihood of severe long-term outcomes such as disability and death following ADRs is significantly higher in the elderly population [175]. Epidemiological studies have found that the classes of drugs most commonly associated with ADRs in the elderly include diuretics, warfarin, non-steroidal anti-inflammatory drugs (NSAIDs), selective serotonin reuptake inhibitors, beta-blockers and angiotensin converting enzyme (ACE)-inhibitors, chemotherapy for cancer, glucocorticoids [176180].

The data of the Italian Group of Pharmacovigilance in the Elderly (GIFA) have shown that age is not an independent risk factor but it is associated with significant changes in pharmacokinetics and pharmacodynamics that predispose the elderly patients to higher risks. Polypharmacy, changes in pharmacokinetics/pharmacodynamics and drug interactions are the main factors predisposing the elederly patients to ADRs [181].

Previous data demonstrates that most of the ADRs in the elderly are “preventable”, therefore they are not Type B reactions. Ventura et al. [182] investigated the prevalence of unpredictable drug adverse reactions among patients admitted to the Emergency Departments (EDs) of three large Italian hospitals in the period 2005–2008. Clinical characteristics and demographics were carefully recorded in a dedicated database. The assessment of the drug reactions was carried out by an allergist after the first emergency evaluation. Over the considered period, 56,031 patients were admitted at the ED, 2644 (21.2%) of which for ADR. Out of those patients, 55 (2.1%) were identified as unpredictable ADRs. In 96% of the cases the clinical presentation was cutaneous and antibiotics were the most frequently responsible drugs. Patients over 65 years accounted for 37% of the reactions. In those patients the multiple drug regimens were significantly more frequent, as well as the presence of comorbidities.

In 2007 the Finnish National Agency for Medicines made available on its website (http://www.nam.fi/publications/tabu) the summary of reports received between 1973 and 2006 about ADRs (including vaccines) in patients ≥75 years. Approximately one-third of the 2013 cases concerned anti-infective drugs. The reactions described had the characteristics of type B reactions.

Therefore it can be concluded that: (I) today there are no adequate studies that define the epidemiology of DA in the elderly and (II) most of the described ADRs are not characteristic DAs. By using the evidence from the studies it is only possible to estimate that the DAs in the elderly have a prevalence of 0.6–2.1% but constitute as much as 10% the fatal adverse reactions.

There are three certainties about the DAs in the elderly: (a) giving the rising age of the population (a risk condition) it can be concluded that at the present time is absolutely essential to proceed with adequate epidemiological studies; (b) in the elderly the diagnosis must be made according to current guidelines [183185] and there should therefore be a “systems” approach, which has a historic equivalent in the geriatric “multi-dimensional” approach. Evaluation involves a meticulous history of past and present ADRs, the acquisition of additional information from medical records and an analysis of the temporal patterns between drug administration and onset of symptoms. Once this data has been assembled, it must be combined with our knowledge about the specific types of allergic reactions caused by the various classes of drugs, in order to identify the potential culprit agents. Advances in drug allergy include identification of HLA associations for penicillin allergy and a micro RNA biomarker/mechanism for toxic epidermal necrolysis [186]. Finally (c) the correct choice of therapy in the elderly population may be the administration of an unrelated medication meeting the criteria of a “lean prescription” and Beers’s criteria [187] a careful administration of a related medication or the desensitization to culprit drug.

Anaphylaxis in the elderly

Anaphylaxis is a severe and potentially fatal hypersensitivity reaction that can affect patients of all ages [188]. The elderly differ from other age groups in terms of risk factors because significant comorbid conditions, such as cardiovascular or cerebrovascular disease, hypertension and cardiac arrhythmias may occur. Moreover, in this population of patients compensatory mechanisms can be physiologically reduced or decreased due to the intake of antihypertensive drugs like β-blockers and ACE inhibitors. Also, trigger agents are usually different than younger patients. All these factors influence the outcome and the response to treatment during anaphylaxis in the elderly. Therefore elderly patients must be considered particularly vulnerable to severe anaphylaxis [189].

Campbell et al. [143] studied presentation and management of patients with anaphylaxis who were 50 or 65 years or older in a consecutive cohort of patients presenting to an emergency department. Cardiovascular symptoms were more likely to occur in older patients and patients 50 or 65 years or older were less likely to be dismissed home directly from the emergency department. Moreover, older patients were less likely to be prescribed self-injectable adrenaline. Among possible triggers, food was the less common supposed cause of anaphylaxis in older patients.

A recent epidemiological study investigated trends in anaphylaxis admissions and fatalities by age, sex, and cause in England and Wales over a 20-year period [190]. In patients 60 years of age and older fatalities and hospital admissions were most common for iatrogenic cause and insect sting induced anaphylaxis, while fatal outcomes from food-induced anaphylaxis were most common in the second decade of life and very rare in adults older than 60 years.

In a population-based epidemiologic study conducted in USA using national databases, among various demographic factors, age was most significantly associated with anaphylaxis deaths, with a mortality rate that was highest in people aged between 75 and 84 years (2.04 per million population) and that was lowest in children 17 years or younger, and increased with advancing age [191].

The increased mortality and occurrence of cardiovascular involvement in older patients can be due to a bigger age-related susceptibility to mast cell-derived mediators on cardiovascular system [192], and to the underlying comorbidities, such as coronary diseases. Mast cell-released mediators are in fact able to induce vasospasm of large coronary arteries, a global reduction of myocardial blood flow by influencing the vasomotor tone of small intramural coronary arteries, and may exert direct dysrhythmogenic effects [193]. Data from two thousand allergists treating patients with anaphylaxis in three European countries showed that age was an important risk factor for the onset of circulatory symptoms with a high odds ratio in older patients [194].

Moreover concomitant cardiovascular medications, more often prescribed to patients with advanced age, could negatively influence the outcome. Antihypertensive medication use is associated with increased organ system involvement during anaphylaxis and increased odds of hospital admission, independent of age [195].

It must be also considered that elderly patients are more likely to receive many different medications (such as antibiotics, NSAIDs), particularly parentally administered medications that cause a consequent more rapid exposure of allergen. Brown and collaborators found that, in a large cohort of people admitted to Emergency Departments, older age and drug (both oral and injected) were significantly correlated with hypotensive reactions [196].

There are no absolute contraindications to the prescription of self-injectable adrenaline in older patients at risk of anaphylaxis, however some issues should be considered. The limited mobility or the presence of joint diseases, such as osteoarthritis of the hand, could reduce the ability to use the auto-injector. Also, a negative effect on therapeutically administered adrenaline is exerted by co-administration of cardiovascular medications such as β-blockers, however in patients suffering for heart diseases such as congestive heart failure, the use of β-blockers could improve the survival and the therapeutic benefit can be greater than the risk of aggravating anaphylaxis in some patients [197]. An evaluation of patient on case-by-case basis together with the cardiologist is therefore useful.

Benefits of physical activity in the elderly with respiratory diseases

In geriatric populations, physical activity represents part of a healthy lifestyle. It favours independence and increases quality of life, improving aerobic capacity, muscle strength, breathing pattern and cardiovascular function, with positive effects on cognitive and psychosocial aspects of daily living in the elderly. Furthermore, the findings from some studies (but not all), support the possibility that exercise may counteract immunosenescence, restoring immune function in older populations and reducing the increased incidence and severity of infectious diseases in senior subjects. It is well known, in fact, that exercise may influence several aspects of immune response, including T-cell phenotype and proliferation, response to vaccination, and cytokine production. However, the underlying mechanisms by which exercise can influence a number of cell types and immune responses still remains to be identified [198, 199].

Physical activity, therefore, seems to play a key role in avoiding the worsening of disabilities, including asthma and other respiratory diseases and may also represent an alternative therapy in patients for whom pharmacological treatment is unavailable, ineffective, or inappropriate. However, even if current data suggests that physical activity may be an effective and logistically easy and clever strategy for counteracting immunosenescence, it is currently underutilized in real clinical settings.

Exercise training and effects on immune and respiratory systems

Response to vaccines and novel antigens has been used as a model that may have clinical relevance to understand the role of exercise in modulating immune response. In a study by Smith et al. [200] comparing sedentary and physically active elderly, the physically active group had significantly higher anti-KLH (keyhole-limpet hemocyanin, a protein antigen) IgM, IgG, IgG1, and delayed-type hypersensitivity responses compared with the sedentary older group, showing that regular physical activity in a senior population is associated with a more efficient and robust immune response to novel antigenic challenge [201]. In general, long term exercise interventions seem to show the most promising results, in particular as regards Exercise-related improvements have been reported with respect to antibody titre, T cell function, macrophage response, alterations of the Th1/Th2 cytokine balance, the level of pro-inflammatory cytokines, and changes in naïve/memory cell ratio [200].

Pathophysiologically, in older patients with chronic airflow limitation as a result of asthma, COPD alone or in combination (ACOS), the normal age-related decline in lung function is amplified. Thus, mechanical ventilatory constraints and the perceived respiratory discomfort become the main factors limiting exercise [202].

Therefore, in clinical practice, middle-aged and older adults with moderate/severe persistent asthma are sometimes referred to pulmonary rehabilitation programs (PRP) that often include both breathing exercises and exercise training. Breathing exercises programs may increase muscle strength and are associated with a positive effect on patient health and quality of life [203]. The rationale underlying their participation in these programs is that the training protocol utilized and benefits achieved are similar to those observed in patients with COPD, who represent the majority of individuals referred to PRP [204].

To date, the majority of studies evaluating exercise training in asthma have been performed in children or young adults with mild-to-moderate persistent disease. A Cochrane meta-analysis of 13 randomized controlled trials of exercise training for asthma concluded that training improves cardiopulmonary fitness in the absence of any changes in lung function [205] while a more recent systematic review and meta-analysis of 17 studies and 599 patients [206] found that exercise training was shown to improve asthma symptoms, QoL, exercise capacity, BHR, EIB, and FEV1 in asthmatics. In this study, improvements in BHR explained part of the improvement in QoL and exercise capacity.

Another review showed that PRP involving exercise training was effective in improving exercise capacity, muscle force, quality of life and reducing symptoms in patients with COPD and asthma [207].

In patients with COPD and Asthma endurance training is recommended, in the form of continuous and interval training, having similar effects on endurance capacity, executed on either a bike or as walking [208]. However, this recommendation must take into consideration that endurance training may elicit an exercise-induced asthma attack and therefore patients must take the preventive treatments with Beta-2 agonists and a perform warm-up phase of at least 15 min prior to exercise. Strength training is important because of the frequently atrophied skeletal musculature, which triggers the increase of the exercise-induced ventilation by early lactate acidosis and consequently aggravates dyspnoea during exercise. Patients should continue the individualized training after discharge from hospital in the domestic environment, benefitting also of training facilities, patients’ groups and social circumstances. Because of the frequently present cardio-metabolic comorbidities the assessment of the exercise capacity as well as an evaluation of nutrition should be included into a holistic therapeutical approach. In patients with uncontrollable fear of exercise-induced attacks of asthma, additional psychological support should be given [208].

Functional exercise capacity is significantly improved after exercise training, as well as asthma symptoms and quality of life [203]. The efficacy of exercise training programs is demonstrated by a study in which, after a 3-year follow-up of asthmatic patients, that undergone a supervised 10-week exercise training program, a significant decrease in number of emergency room visits was observed. The high compliance rate after the 3 years (68%) showed that, when exercise programs are supervised by health professionals, patients choose to continue to be physically active [209].

To evaluate the outcomes of a PRP, the well-known six-minute walk test is often used as a measure.

As conclusion, a case report interestingly suggests that lifelong sports practice can be a great advantage for the asthmatic elderly [201]. A 86-year old athlete with a lifelong atopic asthma and COPD that competes in swimming and triathlon at an international level, with an advanced obstructive lung disease (FEV1/FVC = 34%), experienced only little perceived respiratory discomfort during exercise and had a peak aerobic capacity (VO2 peak) and cycle work rate in excess of 170% predicted. Therefore, while this case report does not diminish the relative importance of ventilator constraints in dyspnoea or exercise intolerance obstructive lung disease, it shows that the negative consequences of these ventilatory impairments can be attenuated by a constant, lifelong participation in high intensity exercise training with the impressive physiological and psychological adaptations associated.

Physical activity should be recommended as a supplementary therapy to medication [210]. Strategies to increase physical and sports activity participation among older people should include (i) raising awareness of the benefits and minimize the perceived risks of physical activity and (ii) improving the environmental and financial access to community-based exercise programs such as may exist for middle-aged and older individuals with other chronic condition [211]. Given this evidence, an increase in the use of physical activity programs by the healthcare community may result in improved health of geriatric populations [198]. If positive effects will also be confirmed in terms of counteraction to immunosenescence, exercise could be a highly cost-effective measure to improve human quality of life compared with other strategies currently being pursued.

Conclusions

The mechanisms that allow “successful” aging are related both to psychophysical conditions and to the immune system behavior. In this regard, in addition to the precise knowledge of the immunological mechanisms in geriatric age, of the lymphoproliferative responses and of the ROS production, the lifestyles, the diet and the physical activity should also be analysed.

Allergic diseases are often thought to be specific of childhood and youth, however, these conditions often persist into older age and can occasionally appear in the elderly. Several are the factors that make more complicated the diagnosis and the management of allergic diseases in aged people, such as frailty, comorbidities and multiple concomitant medications. At the same time, nutritional deficiencies such as vitamin D deficiency, genetic factors, hormonal imbalances, or inflammaging may affect the immune system, causing alterations of the responses of immunocompetent cells. All parameters along with the management approach proposed for diseases can act as support to improve the psychophysical conditions of people over 65 and ensure an active and healthy aging.

Abbreviations

NK:

natural killer

CD:

cluster of differentiation

TREC:

T cell receptor rearrangement excision circles

IL:

interleukin

IL-7R:

interleukin 7 receptor

CMV:

cytomegalovirus

IRP:

immune risk profile

TCC:

T cell clones

Ig:

immunoglobulin

TNF:

tumor necrosis factor

IFN:

interferon

IL-18BP:

IL-18 binding protein

TLR:

Toll-like receptor

NYHA:

New York Heart Association

PBMC:

peripheral blood mononuclear cell

IL-1Ra:

IL-1 receptor antagonist

TGF:

transforming growth factor

WHO:

World Health Organization

PM:

particulate matter

QoL:

quality of life

COPD:

chronic obstructive pulmonary disease

SAC:

seasonal allergic conjunctivitis

PAC:

perennial allergic conjunctivitis

VKC:

vernal keratoconjunctivitis

AKC:

atopic keratoconjunctivitis

ABC:

atopic blepharoconjunctivitis

GPC:

giant papillary conjunctivitis

CAC:

chronic allergic conjunctivitis

Th:

T helper

TNF:

tumor necrosis factor

AIT:

allergen (specific) immunotherapy

SCIT:

subcutaneous immunotherapy

SLIT:

sublingual immunotherapy

EMA:

European Medicines Agency

FDA:

Food and Drug Administration

Treg:

T regulatory

ACE:

angiotensin converting enzyme

ACD:

allergic contact dermatitis

AD:

atopic dermatitis

CU:

chronic urticaria

EAACI:

European Academy of Allergy and Clinical Immunology

GA2LEN:

Global Allergy and Asthma European Network

EDF:

European Dermatology Forum

WAO:

World Allergy Organization

CSU:

chronic spontaneous urticaria

ASST:

autologous serum skin test

MGUS:

monoclonal gammopathy of undetermined significance

HAE-C1-INH:

hereditary angioedemaC1 inhibitor

AAE-C1-INH:

acquired angioedemaC1 inhibitor

AE-ACEi:

angioedema–angiotensin converting enzyme inhibitor

CNS:

central nervous system

CYP:

cytochromes P

SPT:

skin prick tests

FA:

food allergy

S-IgA:

secretory antigen-specific IgA

TJ:

tight junction

ADRs:

adverse drug reactions

DA:

drug allergy

HLA:

human leucocyte antigen

DiHS:

drug-induced hypersensitivity syndrome

DRESS:

drug rash with eosinophilia and systemic symptoms

CI:

confidence interval

NSAIDs:

non-steroidal anti-inflammatory drugs

GIFA:

Gruppo Italiano di Farmacoepidemiologia nell’anziano

EDs:

Emergency Departments

KLH:

keyhole-limpet hemocyanin

ACOS:

asthma-COPD overlap syndrome

PRP:

pulmonary rehabilitation programs

BHR:

bronchial hyperresponsiveness

EIB:

exercise-induced bronchoconstriction

FEV:

forced expiratory volume

FVC:

forced vital capacity

ROS:

reactive oxygen species

References

  1. Nicita-Mauro V, Lo Balbo C, Mento A, Nicita-Mauro C, Maltese G, Basile G. Smoking, aging and the centenarians. Exp Gerontol. 2008;43:95–101.

    Article  CAS  PubMed  Google Scholar 

  2. Franceschi C, Bonafè M, Valensin S, Olivieri F, De Luca M, Ottaviani E, et al. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann N Y Acad Sci. 2000;908:244–54.

    Article  CAS  PubMed  Google Scholar 

  3. Ventura MT, Scichilone N, Gelardi M, Patella V, Ridolo E. Management of allergic disease in the elderly: key considerations, recommendations and emerging therapies. Expert Rev Clin Immunol. 2015;11:1219–28.

    Article  CAS  PubMed  Google Scholar 

  4. Cossarizza A, Ortolani C, Paganelli R, Barbieri D, Monti D, Sansoni P, et al. CD45 isoforms expression on CD4+ and CD8+ T cells throughout life, from newborns to centenarians: implications for T cell memory. Mech Ageing Dev. 1996;86:173–95.

    Article  CAS  PubMed  Google Scholar 

  5. Pinti M, Nasi M, Lugli E, Gibellini L, Bertoncelli L, Roat E, et al. T cell homeostasis in centenarians: from the thymus to the periphery. Curr Pharm Des. 2010;16:597–603.

    Article  CAS  PubMed  Google Scholar 

  6. Passtoors WM, van den Akker EB, Deelen J, Maier AB, van der Breggen R, Jansen R, et al. IL7R gene expression network associates with human healthy ageing. Immun Ageing. 2015;11(12):21.

    Article  CAS  Google Scholar 

  7. Strindhall J, Nilsson BO, Löfgren S, Ernerudh J, Pawelec G, Johansson B, et al. No immune risk profile among individuals who reach 100 years of age: findings from the Swedish NONA immune longitudinal study. Exp Gerontol. 2007;42:753–61.

    Article  CAS  PubMed  Google Scholar 

  8. Paganelli R, Scala E, Rosso R, Cossarizza A, Bertollo L, Barbieri D, et al. A shift to Th0 cytokine production by CD4+ cells in human longevity: studies on two healthy centenarians. Eur J Immunol. 1996;26:2030–4.

    Article  CAS  PubMed  Google Scholar 

  9. Paganelli R, Quinti I, Fagiolo U, Cossarizza A, Ortolani C, Guerra E, et al. Changes in circulating B cells and immunoglobulin classes and subclasses in a healthy aged population. Clin Exp Immunol. 1992;90:351–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Colonna-Romano G, Bulati M, Aquino A, Pellicanò M, Vitello S, Lio D, et al. A double-negative (IgD− CD27−) B cell population is increased in the peripheral blood of elderly people. Mech Ageing Dev. 2009;130:681–90.

    Article  CAS  PubMed  Google Scholar 

  11. Solana R, Pawelec G, Tarazona R. Immunity. Aging Innate Immun. 2006;24:491–4.

    CAS  Google Scholar 

  12. Larbi A, Franceschi C, Mazzatti D, Solana R, Wikby A, Pawelec G. Aging of the immune system as a prognostic factor for human longevity. Physiology. 2008;23:64–74.

    Article  CAS  PubMed  Google Scholar 

  13. Franceschi C, Capri M, Monti D, Giunta S, Olivieri F, Sevini F, et al. Inflammaging and anti-inflammaging: a systemic perspective on aging and longevity emerged from studies in humans. Mech Ageing Dev. 2007;128:92–105.

    Article  CAS  PubMed  Google Scholar 

  14. Michaud M, Balardy L, Moulis G, Gaudin C, Peyrot C, Vellas B, et al. Proinflammatory cytokines, aging, and age-related diseases. J Am Med Dir Assoc. 2013;14:877–82.

    Article  PubMed  Google Scholar 

  15. van den Biggelaar AH, Gussekloo J, de Craen AJ, Frölich M, Stek ML, van der Mast RC, et al. Inflammation and interleukin-1 signaling network contribute to depressive symptoms but not cognitive decline in old age. Exp Gerontol. 2007;42:693–701.

    Article  PubMed  CAS  Google Scholar 

  16. van den Biggelaar AH, De Craen AJ, Gussekloo J, Huizinga TW, Heijmans BT, Frölich M, et al. Inflammation underlying cardiovascular mortality is a late consequence of evolutionary programming. FASEB J. 2004;18:1022–4.

    Google Scholar 

  17. Yende S, Tuomanen EI, Wunderink R, Kanaya A, Newman AB, Harris T, et al. Preinfection systemic inflammatory markers and risk of hospitalization due to pneumonia. Am J Respir Crit Care Med. 2005;172:1440–6.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Zanni F, Vescovini R, Biasini C, Fagnoni F, Zanlari L, Telera A, et al. Marked increase with age of type 1 cytokines within memory and effector/cytotoxic CD8+ T cells in humans: a contribution to understand the relationship between inflammation and immunosenescence. Exp Gerontol. 2003;38:981–7.

    Article  CAS  PubMed  Google Scholar 

  19. Alberti S, Cevenini E, Ostan R, Capri M, Salvioli S, Bucci L, et al. Age-dependent modifications of type 1 and type 2 cytokines within virgin and memory CD4+ T cells in humans. Mech Ageing Dev. 2006;127:560–6.

    Article  CAS  PubMed  Google Scholar 

  20. Palmeri M, Misiano G, Malaguarnera M, Forte GI, Vaccarino L, Milano S, et al. Cytokine serum profile in a group of Sicilian nonagenarians. J Immunoass Immunochem. 2012;33:82–90.

    Article  CAS  Google Scholar 

  21. Sanchez I, Santana S, Escobar C, Santiago JL, Gonzalez A, Ribas N, et al. Clinical implications of different biomarkers in elderly patients with heart failure. Biomark Med. 2014;8:535–41.

    Article  CAS  PubMed  Google Scholar 

  22. Ferrucci L, Corsi A, Lauretani F, Bandinelli S, Bartali B, Taub DD, et al. The origins of age-related proinflammatory state. Blood. 2005;105:2294–9.

    Article  CAS  PubMed  Google Scholar 

  23. Gangemi S, Basile G, Merendino RA, Minciullo PL, Novick D, Rubinstein M, et al. Increased circulating interleukin-18 levels in centenarians with no signs of vascular disease: another paradox of longevity? Exp Gerontol. 2003;38:669–72.

    Article  CAS  PubMed  Google Scholar 

  24. Compté N, Zouaoui Boudjeltia K, Vanhaeverbeek M, De Breucker S, Tassignon J, Trelcat A, et al. Frailty in old age is associated with decreased interleukin-12/23 production in response to Toll-like receptor ligation. PLoS ONE. 2013;8:e65325.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Gangemi S, Basile G, Monti D, Merendino RA, Di Pasquale G, Bisignano U, et al. Age-related modifications in circulating IL-15 levels in humans. Mediators Inflamm. 2005;2005:245–7.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Gangemi S, Parisi P, Ricciardi L, Saitta S, Minciullo PL, Cristani MT, et al. Is interleukin-22 a possible indicator of chronic heart failure’s progression? Arch Gerontol Geriatr. 2010;50:311–4.

    Article  CAS  PubMed  Google Scholar 

  27. Basile G, Paffumi I, D’Angelo AG, Figliomeni P, Cucinotta MD, Pace E, et al. Healthy centenarians show high levels of circulating interleukin-22 (IL-22). Arch Gerontol Geriatr. 2012;54:459–61.

    Article  CAS  PubMed  Google Scholar 

  28. Candore G, Lorenzo G, Caruso C, Modica MA, Colucci AT, Crescimanno G, et al. The effect of age on mitogen responsive T cell precursors in human beings is completely restored by interleukin-2. Mech Ageing Dev. 1992;63:297–307.

    Article  CAS  PubMed  Google Scholar 

  29. Caruso C, Candore G, Cigna D, Di Lorenzo G, Sireci G, Dieli F, et al. Cytokine production pathway in the elderly. Immunol Res. 1996;15:84–90.

    Article  CAS  PubMed  Google Scholar 

  30. Gillis S, Kozak R, Durante M, Weksler ME. Immunological studies of aging. Decreased production of and response to T cell growth factor by lymphocytes from aged humans. J Clin Investig. 1981;67:937–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pawelec G, Barnett Y, Forsey R, Frasca D, Globerson A, McLeod J, et al. T cells and aging, January 2002 update. Front Biosci. 2002;7:d1056–183.

    Article  CAS  PubMed  Google Scholar 

  32. Rea IM, Stewart M, Campbell P, Alexander HD, Crockard AD, Morris TC. Changes in lymphocyte subsets, interleukin 2, and soluble interleukin 2 receptor in old and very old age. Gerontology. 1996;42:69–78.

    Article  CAS  PubMed  Google Scholar 

  33. Rink L, Cakman I, Kirchner H. Altered cytokine production in the elderly. Mech Ageing Dev. 1998;102:199–209.

    Article  CAS  PubMed  Google Scholar 

  34. Myśliwska J, Bryl E, Foerster J, Myśliwski A. Increase of interleukin 6 and decrease of interleukin 2 production during the ageing process are influenced by the health status. Mech Ageing Dev. 1998;100:313–28.

    Article  PubMed  Google Scholar 

  35. McElhaney JE, Zhou X, Talbot HK. The unmet need in the elderly: how immunosenescence, CMV infection, co-morbidities and frailty are a challenge for the development of more effective influenza vaccines. Vaccine. 2012;30:2060–7.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Salvioli S, Capri M, Bucci L, Lanni C, Racchi M, Uberti D, et al. Why do centenarians escape or postpone cancer? The role of IGF-1, inflammation and p53. Cancer Immunol Immunother. 2009;58:1909–17.

    Article  CAS  PubMed  Google Scholar 

  37. Caruso C, Balistreri CR, Crivello A, Forte GI, Grimaldi MP, Listì F, et al. The genetics of innate immunity and inflammation in ageing, age-related diseases and longevity. In: Pawelec G, editor. Immunosenescence. Austin: Landes Bioscience and Springer Science + Business Media; 2007. p. 1–20.

    Google Scholar 

  38. Minciullo PL, Catalano A, Mandraffino G, Casciaro M, Crucitti A, Maltese G, et al. Inflammaging and anti-inflammaging: the role of cytokines in extreme longevity. Arch Immunol Ther Exp. 2016;64:111–26.

    Article  CAS  Google Scholar 

  39. Pes GM, Lio D, Carru C, Deiana L, Baggio G, Franceschi C, et al. Association between longevity and cytokine gene polymorphisms. A study in Sardinian centenarians. Aging Clin Exp Res. 2004;16:244–8.

    Article  CAS  PubMed  Google Scholar 

  40. Jylhä M, Paavilainen P, Lehtimäki T, Goebeler S, Karhunen PJ, Hervonen A, et al. Interleukin-1 receptor antagonist, interleukin-6, and C-reactive protein as predictors of mortality in nonagenarians: the vitality 90+ study. J Gerontol A Biol Sci Med Sci. 2007;62:1016–21.

    Article  PubMed  Google Scholar 

  41. Cavallone L, Bonafè M, Olivieri F, Cardelli M, Marchegiani F, Giovagnetti S, et al. The role of IL-1 gene cluster in longevity: a study in Italian population. Mech Ageing Dev. 2003;124:533–8.

    Article  CAS  PubMed  Google Scholar 

  42. Lin Y, Nakachi K, Ito Y, Tamakoshi A, Yagyu K, Watanabe Y, et al. Variations in serum transforming growth factor-β1 levels with gender, age and lifestyle factors of healthy Japanese adults. Dis Markers. 2009;27:23–8.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Forsey RJ, Thompson JM, Ernerudh J, Hurst TL, Strindhall J, Johansson B, et al. Plasma cytokine profiles in elderly humans. Mech Ageing Dev. 2003;124:487–93.

    Article  CAS  PubMed  Google Scholar 

  44. Carrieri G, Marzi E, Olivieri F, Marchegiani F, Cavallone L, Cardelli M, et al. The G/C915 polymorphism of transforming growth factor β1 is associated with human longevity: a study in Italian centenarians. Aging Cell. 2004;3:443–8.

    Article  CAS  PubMed  Google Scholar 

  45. Halper B, Hofmann M, Oesen S, Franzke B, Stuparits P, Vidotto C, et al. Influence of age and physical fitness on miRNA-21, TGF-β and its receptors in leukocytes of healthy women. Exerc Immunol Rev. 2015;21:154–63.

    PubMed  Google Scholar 

  46. Salvioli S, Monti D, Lanzarini C, Conte M, Pirazzini C, Bacalini MG, et al. Immune system, cell senescence, aging and longevity-inflamm-aging reappraised. Curr Pharm Des. 2013;19:1675–9.

    CAS  PubMed  Google Scholar 

  47. World Health Organization: WHO. Air quality guidelines for particulate matter, ozone, nitrogen dioxide and sulphur dioxide. Global update 2005. In: Summary of risk assessment. Available at:  http://apps.who.int/iris/bitstream/10665/69477/1/WHO_SDE_PHE_OEH_06.02_eng.pdf. Accessed 14 Jan 2017.

  48. U.S. Environmental Protection Agency: National Ambient Air Quality Standards (NAAQS).  Available at: https://www.epa.gov/criteria-air-pollutants/naaqs-table. Accessed 14 Jan 2017.

  49. Pekkanen J, Kulmala M. Exposure assessment of ultrafine particles in epidemiologic time-series studies. Scand J Work Environ Health. 2004;30(Suppl 2):9–18.

    PubMed  Google Scholar 

  50. D’Amato G, Baena-Cagnani CE, Cecchi L, Annesi-Maesano I, Nunes C, Ansotegui I, et al. Climate change, air pollution and extreme events leading to increasing prevalence of allergic respiratory diseases. Multidiscip Respir Med. 2013;8:12.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Bai Y, Sun Q. Fine particulate matter air pollution and atherosclerosis: mechanistic insights. Biochim Biophys Acta. 2016. doi:10.1016/j.bbagen.2016.04.030.

    Google Scholar 

  52. Silva RA, Adelman Z, Fry MM, West JJ. The impact of individual anthropogenic emissions sectors on the global burden of human mortality due to ambient air pollution. Environ Health Perspect. 2016;124:1776–84.

  53. Brunekreef B, Holgate S. Air pollution and health. Lancet. 2002;360:1233–42.

    Article  CAS  PubMed  Google Scholar 

  54. Main CE. Aerobiological, ecological, and health linkages. Environ Int. 2003;29:347–9.

    Article  PubMed  Google Scholar 

  55. Ghio AJ, Carraway MS, Madden MC. Composition of air pollution particles and oxidative stress in cells, tissues, and living systems. J Toxicol Environ Health B. 2012;15:1–21.

    Article  CAS  Google Scholar 

  56. Alexis NE, Carlsten C. Interplay of air pollution and asthma immunopathogenesis: a focused review of diesel exhaust and ozone. Int Immunopharmacol. 2014;23:347–55.

    Article  CAS  PubMed  Google Scholar 

  57. Bell ML, Zanobetti A, Dominici F. Who is more affected by ozone pollution? A systematic review and meta-analysis. Am J Epidemiol. 2014;180:15–28.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Bell ML, Zanobetti A, Dominici F. Evidence on vulnerability and susceptibility to health risks associated with short-term exposure to particulate matter: a systematic review and meta-analysis. Am J Epidemiol. 2013;178:865–76.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Anderson HR, Favarato G, Atkinson RW. Long-term exposure to air pollution and the incidence of asthma: metaanalysis of cohort studies. Air Qual Atmos Health. 2013;6:47–56.

    Article  CAS  Google Scholar 

  60. Kannan JA, Bernstein DI, Bernstein CK, Ryan PH, Bernstein JA, Villareal MS, et al. Significant predictors of poor quality of life in older asthmatics. Ann Allergy Asthma Immunol. 2015;115:198–204.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Epstein TG, Ryan PH, LeMasters GK, Bernstein CK, Levin LS, Bernstein JA, et al. Poor asthma control and exposure to traffic pollutants and obesity in older adults. Ann Allergy Asthma Immunol. 2012;108:423–8.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Bobb JF, Obermeyer Z, Wang Y, Dominici F. Cause-specific risk of hospital admission related to extreme heat in older adults. JAMA. 2014;312:2659–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Shaposhnikov D, Revich B, Bellander T, Bedada GB, Bottai M, Kharkova T, et al. Mortality related to air pollution with the Moscow heat wave and wildfire of 2010. Epidemiology. 2014;25:359–64.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Brook RD, Franklin B, Cascio W, Hong Y, Howard G, Lipsett M, et al. Air pollution and cardiovascular disease: a statement for healthcare professionals from the expert panel on population and prevention science of the American Heart Association. Circulation. 2004;109:2655–71.

    Article  PubMed  Google Scholar 

  65. Yang M, Pan X. Time-series analysis of air pollution and cardiovascular mortality in Beijing, China. J Environ Health. 2008;25:294–7.

    CAS  Google Scholar 

  66. Van Eeden SF, Sin DD. Oxidative stress in chronic obstructive pulmonary disease: A lung and systemic process. Can Respir J. 2013;20:27–9.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Crimmins A, Balbus J, Gamble JL, Beard CB, Bell JE, Dodgen D, et al. Impacts of climate change on human health in the United States: a scientific assessment. U.S. Global Change Research Program, USGCRP, Washington, DC. 2016. p. 99–128.

  68. Bezirtzoglou C, Dekas K, Charvalos E. Climate changes, environment and infection: facts, scenarios and growing awareness from the public health community within Europe. Anaerobe. 2011;17:337–40.

    Article  PubMed  Google Scholar 

  69. Barata M, Ligeti E, De Simone G, Dickinson T, Jack D, Penney J, et al. Climate change and human health in cities. In: Rosenzweig C, Solecki WD, Hammer SA, Mehrotra S, editors. Climate change and cities: first assessment report of the urban climate change research network. Cambridge: Cambridge University Press; 2011. p. 179–213.

  70. Hurst JR, Vestbo J, Anzueto A, Locantore N, Mullerova H, Tal-Singer R, et al. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med. 2010;363:1128–38.

    Article  CAS  PubMed  Google Scholar 

  71. Frenguelli G, Passaleva A. La scelta delle piante destinate al verde ornamentale. Giorn It Allergol Immunol Clin. 2003;13:177–91.

    Google Scholar 

  72. Bonini S. Allergy and the eye. Chem Immunol Allergy. 2014;100:105–8.

    Article  PubMed  Google Scholar 

  73. Leonardi A, De Dominicis C, Motterle L. Immunopathogenesis of ocular allergy: a schematic approach to different clinical entities. Curr Opin Allergy Clin Immunol. 2007;7:429–35.

    Article  CAS  PubMed  Google Scholar 

  74. Bonini S, Gramiccioni C, Bonini M, Bresciani M. Practical approach to diagnosis and treatment of ocular allergy: a 1-year systematic review. Curr Opin Allergy Clin Immunol. 2007;7:446–9.

    Article  PubMed  Google Scholar 

  75. Cardona V, Guilarte M, Luengo O, Labrador-Horrillo M, Sala-Cunill A, Garriga T. Allergic diseases in the elderly. Clin Transl Allergy. 2011;1:11–20.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Ventura MT, D’Amato A, Giannini M, Carretta A, Tummolo RA, Buquicchio R. Incidence of allergic diseases in an elderly population. Immunopharmacol Immunotoxicol. 2010;32:165–70.

    Article  CAS  PubMed  Google Scholar 

  77. Hogan MJ. Atopic keratoconjunctivitis. Trans Am Ophthalmol Soc. 1952;50:265–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Guglielmetti S, Dart JK, Calder V. Atopic keratoconjunctivitis and atopic dermatitis. Curr Opin Allergy Clin Immunol. 2010;10:478–85.

    Article  CAS  PubMed  Google Scholar 

  79. Ridolo E, Montagni M, Caminati M. Emerging drugs for allergic conjunctivitis. Expert Opin Emerg Drugs. 2014;19:291–302.

    Article  CAS  PubMed  Google Scholar 

  80. O’Brien TP. Allergic conjunctivitis: an update on diagnosis and management. Curr Opin Allergy Clin Immunol. 2013;13:543–9.

    Article  PubMed  CAS  Google Scholar 

  81. Abelson MB, Shetty S, Korchak M, Butrus SI, Smith LM. Advances in pharmacotherapy for allergic conjunctivitis. Expert Opin Pharmacother. 2015;16:1219–31.

    Article  CAS  PubMed  Google Scholar 

  82. Verbeken EK, Cauberghs M, Mertens I, Clement J, Lauweryns JM, Van de Woestijne KP. The senile lung. Comparison with normal and emphysematous lungs. 2. Functional aspects. Chest. 1992;101:800–9.

    Article  CAS  PubMed  Google Scholar 

  83. Scichilone N, Callari A, Augugliaro G, Marchese M, Togias A, Bellia V. The impact of age on prevalence of positive skin prick tests and specific IgE tests. Respir Med. 2011;105:651–8.

    Article  PubMed  Google Scholar 

  84. Newson RB, van Ree R, Forsberg B, Janson C, Lötvall J, Dahlén SE, et al. Geographical variation in the prevalence of sensitization to common aeroallergens in adults: the GA(2) LEN survey. Allergy. 2014;69:643–51.

    Article  CAS  PubMed  Google Scholar 

  85. Braman SS, Kaemmerlen JT, Davis SM. Asthma in the elderly. A comparison between patients with recently acquired and long-standing disease. Am Rev Respir Dis. 1991;143:336–40.

    Article  CAS  PubMed  Google Scholar 

  86. Compston JE. Bone marrow and bone: a functional unit. J Endocrinol. 2002;173:387–94.

    Article  CAS  PubMed  Google Scholar 

  87. Gazit R, Weissman IL, Rossi DJ. Hematopoietic stem cells and the aging hematopoietic system. Semin Hematol. 2008;45:218–24.

    Article  PubMed  Google Scholar 

  88. Luscieti P, Hubschmid T, Cottier H, Hess MW, Sobin LH. Human lymph node morphology as a function of age and site. J Clin Pathol. 1980;33:454–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Weksler ME, Szabo P. The effect of age on the B-cell repertoire. J of Clin Immunol. 2000;20:240–9.

    Article  CAS  Google Scholar 

  90. Agrawal A, Agrawal S, Gupta S. Dendritic cells in human aging. Exp Gerontol. 2007;42:421–6.

    Article  CAS  PubMed  Google Scholar 

  91. Swift ME, Burns AL, Gray KL, DiPietro LA. Age-related alterations in the inflammatory response to dermal injury. J Investig Dermatol. 2001;117:1027–35.

    Article  CAS  PubMed  Google Scholar 

  92. Passalacqua G, Canonica GW. Allergen immunotherapy: history and future developments. Immunol Allergy Clin North Am. 2016;36:1–12.

    Article  PubMed  Google Scholar 

  93. Durham SR, Penagos M. Sublingual or subcutanous immunotherapy for allergic rhinitis? J Allergy Clin Immunol. 2016;137:339–49.

    Article  PubMed  Google Scholar 

  94. Bousquet J, Lockey R, Malling HJ. World Health Organization Position Paper. Allergen immunotherapy: therapeutical vaccines for allergic diseases. Edts Allergy. 1998;53:3–15.

    Article  CAS  Google Scholar 

  95. Bousquet J, Khaltaev N, Cruz AA, Denburg J, Fokkens WJ, Togias A, et al. Allergic rhinitis and its impact on asthma (ARIA) 2008 update (in collaboration with the World Health Organization, GA2LEN and AllerGen). Allergy. 2008;63:8–160.

    Article  PubMed  Google Scholar 

  96. Canonica GW, Cox L, Pawankar R, Baena-Cagnani CE, Blaiss M, Bonini S, et al. Sublingual immunotherapy: World Allergy Organization position paper 2013 update. World Allergy Organ J. 2014;7:6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  97. Cox L, Nelson H, Lockey R, Calabria C, Chacko T, Finegold I, et al. Allergen immunotherapy: a practice parameter third update. J Allergy Clin Immunol. 2011;127:S1–55.

    Article  PubMed  Google Scholar 

  98. Allam JP, Novak N. Immunological mechanisms of sublingual immunotherapy. Curr Opin Allergy Clin Immunol. 2014;14:564–9.

    Article  CAS  PubMed  Google Scholar 

  99. Cavkaytar O, Akdis CA, Akdis M. Modulation of immune responses by immunotherapy in allergic diseases. Curr Opin Pharmacol. 2014;17:30–7.

    Article  CAS  PubMed  Google Scholar 

  100. Ozdemir C, Kucuksezer UC, Akdis M, Akdis CA. Mechanisms of Aeroallergen Immunotherapy: subcutaneous immunotherapy and sublingual immunotherapy. Immunol Allergy Clin N Am. 2016;36:71–86.

    Article  Google Scholar 

  101. Passalacqua G. Specific immunotherapy: beyond the clinical scores. Ann Allergy Asthma Immunol. 2011;107:401–6.

    Article  PubMed  Google Scholar 

  102. Malling H, Weeke B. Position paper of the European Academy of Allergy and Clinical Immunol. Position paper: immunotherapy. Allergy. 1993;48:9–35.

    Article  Google Scholar 

  103. Sussman GL, Moote DW, Danoff D, Drouin MA, Leith, ES, Payton KB, et al. Guidelines for the use of allergen immunotherapy. Canadian Society of Allergy and Clinical Immunology. CMAJ. 1995;152:1413–9.

  104. Milgrom H, Huang H. Allergic disorders at a venerable age: a mini review. Gerontology. 2014;60:99–107.

    Article  CAS  PubMed  Google Scholar 

  105. Mathur SK. Allergy and asthma in the elderly. Semin Respir Crit Care Med. 2010;31:587–95.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Pinto JM, Jeswani S. Rhinitis in the geriatric population. Allergy Asthma Clin Immunol. 2010;6:10.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Lombardi C, Raffetti E, Caminati M, Liccardi G, Passalacqua G, Reccardini F, et al. Phenotyping asthma in the elderly: allergic sensitization profile and upper airways comorbidity in patients older than 65 years. Ann Allergy Asthma Immunol. 2016;116:206–11.

    Article  PubMed  Google Scholar 

  108. Bozek A, Jarzab J. Epidemiology of IgE-dependent allergic diseases in elderly patients in Poland. Am J Rhinol Allergy. 2013;27:e140–5.

    Article  PubMed  Google Scholar 

  109. Eaton SM, Burns EM, Kusser K, Randall TD, Haynes L. Age related defects in CD4 T cell cognate helper function lead to reductions in humoral responses. J Exp Med. 2004;200:1613–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Haynes L, Eaton SM. The effect of age on the cognate function of CD4+ T cells. Immunol Rev. 2005;205:220–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Di Lorenzo G, Pacor ML, Esposito Pellitteri M, Listì F, Colombo A, Candore G, et al. A study of age-related IgE pathophysiological changes. Mech Ageing Dev. 2003;124:445–8.

    Article  PubMed  CAS  Google Scholar 

  112. Moro-García MA, Alonso-Arias R, López-Larrea C. Molecular mechanisms involved in the aging of the T-cell immune response. Curr Genomics. 2012;13:589–602.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Listì F, Candore G, Modica MA, Russo M, Di Lorenzo G, Esposito-Pellitteri M, et al. A study of serum immunoglobulin levels in elderly persons that provides new insights into B cell immunosenescence. Ann N Y Acad Sci. 2006;1089:487–95.

    Article  PubMed  CAS  Google Scholar 

  114. Ventura MT, Gelardi M, D’Amato A, Buquicchio R, Tummolo R, Misciagna G, et al. Clinical and cytologic characteristics of allergic rhinitis in elderly patients. Ann Allergy Asthma Immunol. 2012;108:141–4.

    Article  PubMed  Google Scholar 

  115. Bozek A, Ignasiak B, Kasperska-Zajac A, Scierski W, Grzanka A, Jarzab J. Local allergic rhinitis in elderly patients. Ann Allergy Asthma Immunol. 2015;114:199–202.

    Article  PubMed  Google Scholar 

  116. Asero R. Efficacy of injection immunotherapy with ragweed and birch pollen in elderly patients. Int Arch Allergy Immunol. 2004;135:332–5.

    Article  PubMed  Google Scholar 

  117. Marogna M, Bruno ME, Massolo A, Falagiani P. Sublingual immunotherapy for allergic respiratory disease in elderly patients: a retrospective study. Eur Ann Allergy Clin Immunol. 2008;40:22–9.

    CAS  PubMed  Google Scholar 

  118. Bozek A, Kolodziejczyk K, Warkocka-Szoltysek B, Jarzab J. Grass pollen sublingual immunotherapy: a double-blind, placebo-controlled study in elderly patients with seasonal allergic rhinitis. Am J Rhinol Allergy. 2014;28:423–7.

    Article  PubMed  Google Scholar 

  119. Bozek A, Kolodziejczyk K, Krajewska-Wojtys A, Jarzab J. Pre-seasonal, subcutaneous immunotherapy: a double-blinded, placebo-controlled study in elderly patients with an allergy to grass. Ann Allergy Asthma Immunol. 2016;116:156–61.

    Article  PubMed  Google Scholar 

  120. Pitsios C, Demoly P, Bilò MB, Gerth van Wijk R, Pfaar O, Sturm GJ, et al. Clinical contraindications to allergen immunotherapy. an EAACI position paper. Allergy. 2015;70:897–909.

    Article  CAS  PubMed  Google Scholar 

  121. Farage MA, Miller KW, Elsner P, Maibach HI. Functional and physiological characteristics of the aging skin. Aging Clin Exp Res. 2008;20:195–200.

    Article  PubMed  Google Scholar 

  122. Beauregard S, Gilchrest BA. A survey of skin problems and skin care regimens in the elderly. Arch Dermatol. 1987;123:1638–43.

    Article  CAS  PubMed  Google Scholar 

  123. Balato A, Balato N, Costanzo L, Ayala F. Contact sensitization in the elderly. Clin Dermatol. 2011;29:24–30.

    Article  PubMed  Google Scholar 

  124. Zuberbier T, Asero R, Bindslev-Jensen C, Walter Canonica G, Church MK, Giménez-Arnau AM, et al. EAACI/GA(2)LEN/EDF/WAO guideline: management of urticaria. Allergy. 2009;64:1427–43.

    Article  CAS  PubMed  Google Scholar 

  125. Kaplan AP, Greaves M. Pathogenesis of chronic urticaria. Clin Exp Allergy. 2009;39:777–87.

    Article  CAS  PubMed  Google Scholar 

  126. Karakelides M, Monson KL, Volcheck GW, Weiler CR. Monoclonal gammopathies and malignancies in patients with chronic urticaria. Int J Dermatol. 2006;45:1032–8.

    Article  PubMed  Google Scholar 

  127. Malde B, Regalado J, Greenberger PA. Investigation of angioedema associated with the use of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers. Ann Allergy Asthma Immunol. 2007;98:57–63.

    Article  CAS  PubMed  Google Scholar 

  128. Chen YJ, Wu CY, Shen JL, Chen TT, Chang YT. Cancer risk in patients with chronic urticaria: a population-based cohort study. Arch Dermatol. 2012;148:103–8.

    Article  PubMed  Google Scholar 

  129. Magen E, Mishal J, Schlesinger M. Clinical and laboratory features of chronic idiopathic urticaria in the elderly. Int J Dermatol. 2013;52:1387–91.

    Article  PubMed  Google Scholar 

  130. Ventura MT, Napolitano S, Buquicchio R, Cecere R, Arsieni A. An approach to urticaria in the elderly patients. Immunopharmacol Immunotoxicol. 2012;34:530–3.

    Article  CAS  PubMed  Google Scholar 

  131. Ban G-Y, Kim M-Y, Yoo H-S, Nahm D-H, Ye Y-M, Shin Y-S, et al. Clinical feature of elderly chronic urticaria. Korean J Intern Med. 2014;29:800–6.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Ye YM, Kim JE, Nahm DI, Kim SH, Suh CH, Nahm DH, et al. Comparison of clinical characteristics and prognosis of chronic urticaria according to the aspirin sensitivity. Korean J Asthma Allergy Clin Immunol. 2005;25:194–9.

    Google Scholar 

  133. Ventura MT, Cassano N, Romita P, Vestita M, Foti C, Vena GA. Management of chronic spontaneous urticaria in the elderly. Drugs Aging. 2015;32:271–82.

    Article  PubMed  Google Scholar 

  134. Agostini JV, Leo-Summers LS, Inouye SK. Cognitive and other adverse effects of diphenhydramine use in hospitalized older patients. Arch Intern Med. 2001;161:2091–7.

    Article  CAS  PubMed  Google Scholar 

  135. Affrime M, Gupta S, Banfield C, Cohen A. A pharmacokinetic profile of desloratadine in healthy adults, including elderly. Clin Pharmacokinet. 2002;41:13–9.

    Article  CAS  PubMed  Google Scholar 

  136. Jáuregui I, García-Lirio E, Soriano AM, Gamboa PM, Antépara I. An overview of the novel H1-antihistamine bilastine in allergic rhinitis and urticaria. Expert Rev Clin Immunol. 2012;8:33–41.

    Article  PubMed  CAS  Google Scholar 

  137. Maurer M, Rosén K, Hsieh HJ, Saini S, Grattan C, Gimenéz-Arnau A, et al. Omalizumab for the treatment of chronic idiopathic or spontaneous urticaria. N Engl J Med. 2013;368:924–35.

    Article  CAS  PubMed  Google Scholar 

  138. Saini SS, Bindslev-Jensen C, Maurer M, Grob JJ, Bülbül Baskan E, Bradley MS, et al. Efficacy and safety of omalizumab in patients with chronic idiopathic/spontaneous urticaria who remain symptomatic on H1 antihistamines: a randomized, placebo-controlled study. J Investig Dermatol. 2015;135:67–75.

    Article  PubMed  Google Scholar 

  139. Kaplan A, Ledford D, Ashby M, Canvin J, Zazzali JL, Conner E, et al. Omalizumab in patients with symptomatic chronic idiopathic/spontaneous urticaria despite standard combination therapy. J Allergy Clin Immunol. 2013;132:101–9.

    Article  CAS  PubMed  Google Scholar 

  140. Metz M, Ohanyan T, Church MK, Maurer M. Omalizumab is an effective and rapidly acting therapy in difficult-to-treat chronic urticaria: a retrospective clinical analysis. J Dermatol Sci. 2014;73:57–62.

    Article  CAS  PubMed  Google Scholar 

  141. Wöhrl S, Stingl G. Underestimation of allergies in elderly patients. Lancet. 2004;363:249.

    Article  PubMed  Google Scholar 

  142. Diesner SC, Untersmayr E, Pietschmann P, Jensen-Jarolim E. Food allergy: only a pediatric disease? Gerontology. 2011;57:28–32.

    Article  CAS  PubMed  Google Scholar 

  143. Campbell RL, Hagan JB, Li JT, Vukov SC, Kanthala AR, Smith VD, et al. Anaphylaxis in emergency department in patients 50 or 65 or older. Ann Allergy Asthma Immunol. 2011;106:401–6.

    Article  PubMed  Google Scholar 

  144. Weiskopf D, Weinberger B, Grubeck-Loebenstein B. The aging of the immune system. Transpl Int. 2009;22:1041–50.

    Article  CAS  PubMed  Google Scholar 

  145. De Faria AM, Ficker SM, Speziali E, Menezes JS, Stransky B, Silva Rodrigues V, et al. Aging affects oral tolerance induction but not its maintenance in mice. Mech Ageing Dev. 1998;102:67–80.

    Article  PubMed  Google Scholar 

  146. Sato S, Kiyono H, Fujihashi K. Mucosal immunosenescence in the gastrointestinal tract: a mini-review. Gerontology. 2015;61:336–42.

    Article  CAS  PubMed  Google Scholar 

  147. Ma TY, Hollander D, Dadufalza V, Krugliak P. Effect of aging and caloric restriction on intestinal permeability. Exp Gerontol. 1992;27:321–33.

    Article  CAS  PubMed  Google Scholar 

  148. Al-Sadi R, Ye D, Said HM, Ma TY. IL-1beta-induced increase in intestinal epithelial tight junction permeability is mediated by MEKK-1 activation of canonical NF-kappaB pathway. Am J Pathol. 2010;177:2310–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Ershler WB. Biological interactions of aging and anemia: a focus on cytokines. J Am Geriatr Soc. 2003;51:S18–21.

    Article  PubMed  Google Scholar 

  150. Tran L, Greenwood-Van Meerveld B. Age-associated remodeling of the intestinal epithelial barrier. J Gerontol A Biol Sci Med Sci. 2013;68:1045–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Macpherson AJ, McCoy KD. Stratification and compartmentalisation of immunoglobulin responses to commensal intestinal microbes. Semin Immunol. 2013;25:358–63.

    Article  CAS  PubMed  Google Scholar 

  152. Thoreux K, Owen RL, Schmucker DL. Intestinal lymphocyte number, migration and antibody secretion in young and old rats. Immunology. 2000;101:161–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Man AL, Gicheva N, Nicoletti C. The impact of ageing on the intestinal epithelial barrier and immune system. Cell Immunol. 2014;289:112–8.

    Article  CAS  PubMed  Google Scholar 

  154. Ventura MT, Napolitano S, Menga R, Cecere R, Asero R. Anisakis simplex is associated with chronic urticaria in endemic areas. Int Arch Allergy Immunol. 2013;160:297–300.

    Article  CAS  PubMed  Google Scholar 

  155. Ventura MT, Tummolo RA, Di Leo E, D’Erasmo M, Arsieni A. Immediate and cell-mediated reactions in parasitic infections by Anisakis simplex. J Investig Allergol Clin Immunol. 2008;18:253–9.

    CAS  PubMed  Google Scholar 

  156. Ventura MT, Polimeno L, Amoruso AC, Gatti F, Annoscia E, Marinaro M, et al. Intestinal permeability in patients with adverse reactions to food. Dig Liver Dis. 2006;38:732–6.

    Article  CAS  PubMed  Google Scholar 

  157. Prasad A. Zinc and immunity. Mol Cell Biochem. 1998;188:63–9.

    Article  CAS  PubMed  Google Scholar 

  158. Ahluwalia N, Sun J, Krause D, Mastro A, Handte G. Immune function is impaired in iron-deficient, homebound, older women. Am J Clin Nutr. 2004;79:516–21.

    CAS  PubMed  Google Scholar 

  159. Chambers ES, Hawrylowicz CM. The impact of vitamin D on regulatory T cells. Curr Allergy Asthma Rep. 2011;11:29–36.

    Article  CAS  PubMed  Google Scholar 

  160. Untersmayr E, Diesner SC, Bramswig KH, Knittelfelder R, Bakos N, Gundacker C, et al. Characterization of intrinsic and extrinsic risk factors for celery allergy in immunosenescence. Mech Ageing Dev. 2008;129:120–8.

    Article  CAS  PubMed  Google Scholar 

  161. Lazarou J, Pomeranz BH, Corey PN. Incidence of adverse drug reactions in hospitalized patients: a meta-analysis of prospective studies. JAMA. 1998;279:1200.

    Article  CAS  PubMed  Google Scholar 

  162. Ventura MT, Calogiuri GF, Muratore L, Di Leo E, Buquicchio R, Ferrannini A, et al. Cross-reactivity in cell-mediated and IgE-mediated hypersensitivity to glucocorticoids. Curr Pharm Design. 2006;12:3383–91.

    Article  CAS  Google Scholar 

  163. Johansson SG, Bieber T, Dahl R, Friedmann PS, Lanier BQ, Lockey RF, et al. Revised nomenclature for allergy for global use: report of the Nomenclature Review Committee of the World Allergy Organization, October 2003. J Allergy Clin Immunol. 2004;113:832.

    Article  CAS  PubMed  Google Scholar 

  164. Ben m’rad M, Leclerc-Mercier S, Blanche P, Franck N, Rozenberg F, Fulla Y, et al. Drug-induced hypersensitivity syndrome: clinical and biologic disease patterns in 24 patients. Medicine. 2009;88:131.

    Article  PubMed  CAS  Google Scholar 

  165. Mallet L, Spinewine A, Huang A. The challenge of managing drug interactions in elderly people. Lancet. 2007;370:185.

    Article  CAS  PubMed  Google Scholar 

  166. Alhawassi TM, Krass I, Bajorek BV, Pont LG. A systematic review of the prevalence and risk factors for adverse drug reactions in the elderly in the acute care setting. Clin Interv Aging. 2014;9:2079–86.

    PubMed  PubMed Central  Google Scholar 

  167. Budniz DS, Lovegrove MC, Shehab N, Richards CL. Emerging hospitalizations fr adverse drug events im older Americans. N Engl J Med. 2011;365:2001.

    Google Scholar 

  168. Beijer HJ, de Blaey CJ. Hospitalizations caused by adverse drug reactons (ADR): a meta-analysis of observational studies. Pharm World Sci. 2002;24:46.

    Article  CAS  PubMed  Google Scholar 

  169. Muhlberger N, Schneeweiss S, Hasford J. Adverse drug reaction monitoring—cost and benefit considerations. Part 1: frequency of ADRs leading to hospital admissions. Pharmacoepidemiol Drug Saf. 1997;6:S71–7.

    Article  Google Scholar 

  170. Schneeweiss S, Hasford J, Göttler M, Hoffmann A, Riethling AK, Avorn J. Admissions caused by adverse drug events to internal medicine and emergency departments in hospitals: a longitudinal population-based study. Eur J Clin Pharmacol. 2002;58:285–91.

    Article  PubMed  Google Scholar 

  171. Winterstein AG, Sauer BC, Hepler CD. Poole C Preventable drug-related hospital admissions. Ann Pharmacother. 2002;36:1238–48.

    Article  PubMed  Google Scholar 

  172. Sheldon T. Dutch doctors call for action on drug safety. BMJ. 2006;333:1238.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Conforti A, Costantini D, Zanetti F, Moretti U, Grezzana M, Leone R. Adverse drug reactions in older patients: an Italian observational prospective hospital study. Drug Healthc Patient Saf. 2012;4:75–80.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Hanlon JT, Schmader KE, Koronkowski MJ, Weinberger M, Landsman PB, Samsa GP, et al. Adverse drug events in high risk older outpatients. J Am Geriatr Soc. 1997;45:945–8.

    Article  CAS  PubMed  Google Scholar 

  175. Wilson RM, Runciman WB, Gibberd RW, Harrison BT, Newby L, Hamilton JD. The quality in Australian health care study. Med J Aust. 1995;163:458–71.

    CAS  PubMed  Google Scholar 

  176. Heng YK, Lim YL. Cutaneos adverse drug reactions in elderly. Curr Opin Allergy Clin Immunol. 2015;15:300–7.

    Article  CAS  PubMed  Google Scholar 

  177. Pirmohamed M, James S, Meakin S, Green C, Scott AK, Walley TJ, et al. Adverse drug reactions as cause of admission to hospital: prospective analysis of 18,820 patients. BMJ. 2004;329:15–9.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Gurwitz JH, Field TS, Harrold LR, Rothschild J, Debellis K, Seger AC, et al. Incidence and preventability of adverse drug events among older persons in the ambulatory setting. JAMA. 2003;289:1107–16.

    Article  PubMed  Google Scholar 

  179. Gandhi TK, Weingart SN, Borus J, Seger AC, Peterson J, Burdick E, et al. Adverse drug events in ambulatory care. N Engl J Med. 2003;348:1556–64.

    Article  PubMed  Google Scholar 

  180. Ventura MT, Sanapo F, Calogiuri GF, Satriano F. Anaphylaxis induced by intramuscular betamethasone disodium phosphate: reflections on a clinical case. Int J Immunopathol Pharmacol. 2007;20:387–91.

    Article  CAS  PubMed  Google Scholar 

  181. Onder G, Pedone C, Landi F, Cesari M, Della Vedova C, Bernabei R, et al. Adverse drug reactions as cause of hospital admissionis: results from the Italian Group of Pharmacoepidemiology in the Elderly (GIFA). J Am Geriatr Soc. 2002;50:1962.

    Article  PubMed  Google Scholar 

  182. Ventura MT, Laddaga R, Cavallera P, Pugliese P, Tummolo RA, Buquicchio R, et al. Adverse drug reactions as the cause of emergency department admission: focus on the elderly. Immunopharmacol Immunotoxicol. 2010;32:426–9.

    Article  PubMed  Google Scholar 

  183. Pichler WJ, Franklin Adkinson N Jr, Feldweg AM. An approach to the patient with drug allergy. UpTodate. Wolters Kluwer.

  184. Joint Task Force on Practice Parameters, American Academy of Allergy, Asthma and Immunology, American College of Allergy, Asthma and Immunology, Joint Council of Allergy, Asthma and Immunology. Drug allergy: an updated practice parameter. Ann Allergy Asthma Immunol. 2010;105:259–73.

    Article  Google Scholar 

  185. Demoly P, Adkinson NF, Brockow K, Castells M, Chiriac AM, Greenberger PA, et al. International consensus on drug allergy. Allergy. 2014;69:420–37.

    Article  CAS  PubMed  Google Scholar 

  186. Sicherer SH, Leung DY. Advances in allergic skin disease, anaphylaxis, and hypersensitivity reactions to foods, drugs, and insects in 2014. J Allergy Clin Immunol. 2015;135:357–67.

    Article  PubMed  Google Scholar 

  187. Beers MH. Explicit criteria for determining potentially inappropriate medication use by the elderly. An update. Arch Intern Med. 1997;157:1531–6.

    Article  CAS  PubMed  Google Scholar 

  188. Muraro A, Roberts G, Worm M, Bilò MB, Brockow K, Fernández Rivas M, et al. Anaphylaxis: guidelines from the European Academy of Allergy and Clinical Immunology. Allergy. 2014;69:1026–45.

    Article  CAS  PubMed  Google Scholar 

  189. Simons FE, Ardusso LR, Bilò MB, El-Gamal YM, Ledford DK, Ring J, World Allergy Organization, et al. World allergy organization guidelines for the assessment and management of anaphylaxis. World Allergy Organ J. 2011;4:13–37.

    Article  PubMed  PubMed Central  Google Scholar 

  190. Turner PJ, Gowland MH, Sharma V, Ierodiakonou D, Harper N, Garcez T, et al. Increase in anaphylaxis-related hospitalizations but no increase in fatalities: an analysis of United Kingdom national anaphylaxis data, 1992–2012. J Allergy Clin Immunol. 2015;135:956–63.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Ma L, Danoff TM, Borish L. Case fatality and population mortality associated with anaphylaxis in the United States. J Allergy Clin Immunol. 2014;133:1075–83.

    Article  PubMed  Google Scholar 

  192. Triggiani M, Montagni M, Parente R, Ridolo E. Anaphylaxis and cardiovascular diseases: a dangerous liaison. Curr Opin Allergy Clin Immunol. 2014;14:309–15.

    Article  PubMed  Google Scholar 

  193. Triggiani M, Patella V, Staiano RI, Granata F, Marone G. Allergy and the cardiovascular system. Clin Exp Immunol. 2008;153(Suppl 1):7–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Worm M, Edenharter G, Ruëff F, Scherer K, Pföhler C, Mahler V, et al. Symptom profile and risk factors of anaphylaxis in Central Europe. Allergy. 2012;67:691–8.

    Article  CAS  PubMed  Google Scholar 

  195. Lee S, Hess EP, Nestler DM, Bellamkonda Athmaram VR, Bellolio MF, Decker WW, et al. Antihypertensive medication use is associated with increased organ system involvement and hospitalization in emergency department patients with anaphylaxis. J Allergy Clin Immunol. 2013;131:1103–8.

    Article  PubMed  Google Scholar 

  196. Brown SG, Stone SF, Fatovich DM, Burrows SA, Holdgate A, Celenza A, et al. Anaphylaxis: clinical patterns, mediator release, and severity. J Allergy Clin Immunol. 2013;132:1141–9.

    Article  CAS  PubMed  Google Scholar 

  197. Tenbrook JA, Wolf MP, Hoffman SN, Rosenwasser LJ, Konstam MA, Salem DN, et al. Should beta-blockers be given to patients with heart disease and peanut-induced anaphylaxis? A decision analysis. J Allergy Clin Immunol. 2004;113:977–82.

    Article  CAS  PubMed  Google Scholar 

  198. de Araujo AL, Silva LC, Fernandes JR, Benard G. Preventing or reversing immunosenescence: can exercise be an immunotherapy? Immunotherapy. 2013;5:879–93.

    Article  PubMed  CAS  Google Scholar 

  199. Senchina DS, Kohut ML. Immunological outcomes of exercise in older adults. Clin Interv Aging. 2007;2:3–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Smith TP, Kennedy SL, Fleshner M. Influence of age and physical activity on the primary in vivo antibody and T cell-mediated responses in men. J Appl Physiol. 2004;97:491–8.

    Article  CAS  PubMed  Google Scholar 

  201. Kohut ML, Senchina DS. Reversing age-associated immunosenescence via exercise. Exerc Immunol Rev. 2004;10:6–41.

    PubMed  Google Scholar 

  202. Guenette JA, Diane Lougheed M, Webb KA, O’Donnell DE. Can an 86-year-old woman with advanced lung disease be a world class athlete? Respir Physiol Neurobiol. 2012;181:162–6.

    Article  PubMed  Google Scholar 

  203. Gomieiro LT, Nascimento A, Tanno LK, Agondi R, Kalil J, Giavina-Bianchi P. Respiratory exercise program for elderly individuals with asthma. Clinics. 2011;66:1163–9.

    Article  PubMed  Google Scholar 

  204. Turner S, Eastwood P, Cook A, Jenkins S. Improvements in symptoms and quality of life following exercise training in older adults with moderate/severe persistent asthma. Respiration. 2011;81:302–10.

    Article  PubMed  Google Scholar 

  205. Ram FS, Robinson SM, Black PN, Picot J. Physical training for asthma. Cochrane Database Syst Rev. 2005:CD001116.

  206. Eichenberger PA, Diener SN, Kofmehl R, Spengler CM. Effects of exercise training on airway hyperreactivity in asthma: a systematic review and meta-analysis. Sports Med. 2013;43:1157–70.

    Article  PubMed  Google Scholar 

  207. Nellessen A, Hernandes NA, Pitta F. Physiotherapy and rehabilitative interventions in patients with chronic respiratory diseases: exercise and non-exercise treatment. Panminerva Med. 2013;55(2):197–209.

    CAS  PubMed  Google Scholar 

  208. Rohrer V, Schmidt-Trucksass A. Impact of exercise, sport and rehabilitation therapy in asthma and COPD. Ther Umsch. 2014;71:295–300.

    Article  PubMed  Google Scholar 

  209. Emtner M, Finne M, Stalenheim G. A 3-year follow-up of asthmatic patients participating in a 10-week rehabilitation program with emphasis on physical training. Arch Phys Med Rehabil. 1998;79:539–44.

    Article  CAS  PubMed  Google Scholar 

  210. Scichilone N, Ventura MT, Bonini M, Braido F, Bucca C, Caminati M, et al. Choosing wisely: practical considerations on treatment efficacy and safety of asthma in the elderly. Clin Mol Allergy. 2015;13:7.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  211. Franco MR, Tong A, Howard K, Sherrington C, Ferreira PH, Pinto RZ, et al. Older people’s perspectives on participation in physical activity: a systematic review and thematic synthesis of qualitative literature. Br J Sports Med. 2015;49:1268–76.

    Article  PubMed  Google Scholar 

Download references

Authors’ contributions

MTV and SG made substantial contributions to conception and design of this review. MTV, SG and PLM participated in critical revision and drafting of final version of the manuscript. MTV, NS, RP, PLM, VP, MB, GP, CL, LS, ER, SRDG participated in literature search, acquisition of data and manuscript writing. GWC gave the final approval of the version of the manuscript. All authors read and approved the final manuscript.

Acknowledgements

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Paola Lucia Minciullo.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ventura, M.T., Scichilone, N., Paganelli, R. et al. Allergic diseases in the elderly: biological characteristics and main immunological and non-immunological mechanisms. Clin Mol Allergy 15, 2 (2017). https://doi.org/10.1186/s12948-017-0059-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12948-017-0059-2

Keywords