Abstract
The International Collaboration in Asthma, Allergy and Immunology initiated an international coalition among the American Academy of Allergy, Asthma & Immunology; the European Academy of Allergy and Clinical Immunology; the World Allergy Organization; and the American College of Allergy, Asthma & Immunology on common variable immunodeficiency. An author group was formed and then divided into individual committees. Within the committee, teams of authors were subgrouped to generate content for specific sections of the document. Content was derived from literature searches, relevant published guidelines, and clinical experience. After a draft of the document was assembled, it was collectively reviewed and revised by the authors. Where evidence was lacking or conflicting, the information presented represents the consensus expert opinion of the group. The full document was then independently reviewed by 5 international experts in the field, none of whom was among the authors of the original. The comments of these reviewers were incorporated before submission for publication.
DEFINITION
The term “common variable immunodeficiency” (CVID) was coined in 1971 by a World Health Organization committee to separate less well-defined antibody deficiency syndromes from others with a more coherent clinical description and Mendelian inheritance.1,2 Therefore, the hypogammaglobulinemic syndrome of CVID became a diagnosis of exclusion. Since then, the International Union of Immunological Societies Expert Primary Immunodeficiency Committee redefined the conditions in 2009 as “common variable immunodeficiency disorders”, thus retaining the CVID acronym but emphasizing the heterogeneous nature of these hypogammaglobulinemic states.3 According to the proposal by the European Society for Immunodeficiencies and the Pan American Group for Immunodeficiency in 1999, CVID was defined as follows:
-
CVID is probable in a male or female patient who has a marked decrease of IgG (at least 2 SD below the mean for age) and a marked decrease in at least one of the isotypes IgM or IgA, and fulfills all of the following criteria:
- Onset of immunodeficiency at greater than 2 years of age
- Absent isohemagglutinins and/or poor response to vaccines
- Defined causes of hypogammaglobulinemia have been excluded according to a list of differential diagnosis (Table I).
As will be discussed further below, CVID encompasses a group of heterogeneous primary antibody failure syndromes characterized by hypogammaglobulinemia. The number of potential distinct entities within this group is still unknown, and the diagnosis remains one of exclusion. Monogenic forms have been described, but polygenic inheritance is likely in most cases.4–6 Despite the fact that several monogenic defects underlying apparent CVID have been defined, because of the rarity of each defect and the lack in most cases of significant impact on management, as well as the cost of testing, genetic studies are not considered appropriate for routine use in patients with CVID at this time.
TABLE I.
Drug induced |
Antimalarial agents |
Captopril |
Carbamazepine |
Glucocorticoids |
Fenclofenac |
Gold salts |
Penicillamine |
Phenytoin |
Sulfasalazine |
Anti-CD20 mAbs (rituximab) |
Single gene and other defects |
Ataxia telangiectasia |
Autosomal-recessive forms of SCID and other forms of combined immunodeficiency |
Hyper-IgM syndromes |
Transcobalamin II deficiency and hypogammaglobulinemia |
X-linked agammaglobulinemia |
X-linked lymphoproliferative disorder (EBV-associated) |
X-linked SCID |
Some metabolic disorders |
Chromosomal anomalies |
Chromosome 18q-syndrome |
Monosomy 22 |
Trisomy 8 |
Trisomy 21 |
Infectious diseases |
HIV |
Congenital infection with rubella virus |
Congenital infection with cytomegalovirus |
Congenital infection with Toxoplasma gondii |
EBV |
Malignancy |
Chronic lymphocytic leukemia |
Immunodeficiency with thymoma |
Non-Hodgkin lymphoma |
Monoclonal gammopathy (mutiple myeloma, Waldenstrom macroglobulinemia) |
Other systemic disorders |
Immunodeficiency caused by excessive loss of immunoglobulins (nephrosis, severe burns, lymphangiectasia, protein-losing enteropathy) |
SCID, Severe combined immunodeficiency
The onset of the varied clinical manifestations and laboratory abnormalities do not necessarily coincide, and may occur at any age from early childhood to old age. Given (1) the broad differential diagnosis of hypogammaglobulinemia (Table I),7 (2) the challenge of differentiating some of these in early childhood (particularly regarding definitive assessment of vaccine responses), and (3) that CVID is considered a diagnosis of exclusion, it is best not to confer this diagnosis before at least age 4 years.
Antibody production is always disturbed in CVID. This is often the result of B-cell dysfunction, but may also result primarily from impairment of T-cell function and lack of sufficient help for antibody production. Infection susceptibility is mainly to encapsulated extracellular bacteria in the respiratory tract, but there may also occur various other clinical manifestations affecting many organ systems. The phenotype is very broad, ranging from only bacterial infections, to progression from a CVID-like condition to severe disease similar to a combined immunodeficiency, possibly having a different etiology.8,9 Some patients may also have distinct initial presentations, such as autoimmune disease, granulomatous disease, or enteropathy without recurrent infections (discussed in detail below).10,11
The normal range of IgG serum levels varies in different age groups; therefore, it is critical that this be defined according to the age-adjusted reference range for the population. An absolute lower limit value of IgG at 4.5 g/L for adults has been proposed, because nearly 95% of the patients with CVID in a European cohort fulfilled this criterion.12 However, it is recognized that some patients with CVID have relatively high residual IgG levels (up to 6 g/L) at diagnosis while still showing impaired specific antibody formation.13 Furthermore, the normal range of IgG levels may also vary according to race or ethnicity.14 Thus, for practical purposes, the definition of hypogammaglobulinemia depends on the local or regional reference range applicable to the patient. In addition to a low IgG level, IgA or IgM level must be low for a definite diagnosis of CVID.15 Note that not all clinical immunologists agree regarding these laboratory criteria. Some do not confer a diagnosis of CVID if the IgA level is normal. We publish the less stringent criteria here because it is an accepted standard for many practitioners. It is of critical importance that all immunoglobulin measurements be interpreted according to age-specific normal ranges and that levels be consistently low on repeated measurements at least 3 weeks apart.
Vaccine responses will be discussed later in greater depth. Impairment of IgG vaccine responses is an extremely important element of the definition of CVID. Note that depending on circumstances, some immunologists forego antibody measurement, for example, if the total IgG level is very low (<100 mg/dL) or if the clinical presentation and other laboratory features are highly characteristic; see section on Diagnosis).
Consensus definition of CVID
Most patients will have at least 1 of the characteristic clinical manifestations (infection, autoimmunity, lymphoproliferation). However, a diagnosis of CVID may be conferred on asymptomatic individuals who fulfill criteria 2 to 5, especially in familial cases.
Hypogammaglobulinemia should be defined according to the age-adjusted reference range for the laboratory in which the measurement is performed. The IgG level must be repeatedly low in at least 2 measurements more than 3 weeks apart in all patients. Repeated measurement may be omitted if the level is very low (<100–300 mg/dL depending on age), other characteristic features are present, and it is considered in the best interest of the patient to initiate therapy with IgG as quickly as possible.
IgA or IgM level must also be low. (Note that some experts prefer a more narrow definition requiring low IgA level in all patients.)
It is strongly recommended that all patients with an IgG level of more than 100 mg/dL should be studied for responses to T-dependent (TD) and T-independent (TI) antigens, whenever possible. In all patients undergoing such testing, there must be a demonstrable impairment of response to at least 1 type of antigen (TD or TI). At the discretion of the practitioner, specific antibody measurement may be dispensed with if all other criteria are satisfied and if the delay incurred by prevaccination and postvaccination antibody measurement is thought to be deleterious to the patient’s health.
Other causes of hypogammaglobulinemia must be excluded (Table I).
Genetic studies to investigate monogenic forms of CVID or for disease-modifying polymorphisms are not generally required for diagnosis and management in most of the patients, especially those who present with infections only without immune dysregulation, autoimmunity, malignancy, or other complications. In these latter groups of patients, however, single gene defects may be amenable to specific therapies (eg, stem cell therapy) and molecular genetic diagnosis should be considered when possible.
Ameratunga et al16 have recently proposed a distinct set of diagnostic criteria for CVID. Differences between the criteria of Ameratunga et al16 and those stated above are summarized in Table II. Although the criteria presented here do not define “possible” or “probable” forms of CVID, we recognize that some patients with a low IgG level and impaired vaccine responses may not fulfill our criteria for CVID (at least at initial evaluation) because IgA or IgM level is not low. This is a form of hypogammaglobulinemia with antibody deficiency that should not be called CVID (it may be called “unspecified IgG deficiency” or “unspecified hypogammaglobulinemia”). Alternatively, IgG and IgA levels may be low, but vaccine responses may appear normal by standard criteria. In all these cases, patients should be assessed repeatedly over time because immunoglobulin levels and antibody function may wane to the point that the above criteria are met and a diagnosis of CVID may be conferred. Regardless of whether there is deterioration over time, many patients with abnormal immunoglobulin levels and/or functional antibody responses not meeting criteria for CVID may have a significant burden of infections and should be assessed for benefit from IgG replacement.
TABLE II.
CVID ICON | Ameratunga et al16 |
---|---|
The diagnosis is “definite” if all criteria are met. | The diagnosis is “probable” if all criteria are met. |
“Probable” or “possible” CVID is not defined. | Both “probable” and “possible” CVID are defined; there is no “definite” CVID. |
Serum IgG level must be below local/regional clinical laboratory norms. |
Serum IgG must be <5 g/L for all. |
IgA or IgM must be low (IgA low preferred). | IgA or IgM may be low, but neither is required to be low. |
Impaired vaccine response is required for the diagnosis. | Impaired vaccine response may be supportive of the diagnosis, but is not required. |
The diagnosis may be established solely on the basis of laboratory criteria. |
Some symptom/clinical manifestation must be present for “probable” CVID. |
The diagnosis is not influenced by additional laboratory criteria. |
“Probable” CVID may be established with supportive laboratory criteria (low IgG3, low memory B cells, increased CD21, low B cells, autoantibodies) and genetic alterations (TACI, BAFFR, MSH5, and others). |
The diagnosis does not depend on histological criteria. | “Probable” CVID may be established with supportive histological criteria (granulomas, lung or gastrointestinal disease). |
ICON, International Consensus.
We do not see a benefit to defining possible or probable categories of diagnosis on the basis of laboratory or histopathologic criteria. The requirement for low IgA (or IgM) level provides additional diagnostic specificity. We believe that it is more appropriate to define hypogammaglobulinemia according to regional and ethic laboratory norms rather than using a single numeric criterion across the board.
EPIDEMIOLOGY
Primary immunodeficiency disorders are usually considered to be “rare diseases.” However, as a whole, this group of diseases may not be as rare as once thought. Neither the true incidence nor the true prevalence of primary immunodeficiency is known. Most studies are geographically limited and based on survey data or record review of diagnosed and/or registered cases.17–21 The number of undiagnosed cases is unknown because a comprehensive population-based screening process for defined primary immunodeficiency has not been developed anywhere in the world.
Almost every primary immunodeficiency registry available for consultation reports a predominance of antibody defects (generally >50%), and in most of them, CVID is the most frequent symptomatic antibody deficiency diagnosed in adulthood.20,22 IgA deficiency occurs with higher frequency overall, but most patients with IgA deficiency are asymptomatic.23 Note that in children, specific antibody deficiency is more often diagnosed.24 In 2007, a Latin American Group for Immunodeficiency registry report revealed that in a total of 3321 patients registered, the most common form of primary immunodeficiency disease was predominantly antibody deficiency (53.2%), with IgA deficiency and CVID reported as the most frequent types.20 A recent report from the European Society for Immunodeficiencies registry database including 13,708 patients from 41 countries established that CVID represents the most common entity with 2880 patients or 21% of all entries.18,25 The United States Immunodeficiency Network registry contains 3459 subjects, with 1049 subjects with CVID (30%).26
There are no precise data on the prevalence of CVID, but it has been estimated at between 1:100,000 and 1:10,000 of the population.12 The European Society for Immunodeficiencies registry estimates a total primary immunodeficiency diseases (PIDD) prevalence ranging from 1.3 (Poland) to 5 (France) per 100,000 living persons in European countries.18,25 The calculated prevalence of CVID in these European countries ranges from 0.07 to 0.98 patients per 100,000 inhabitants. These observed differences between countries are thought to be a consequence of underreporting in those showing the lower rates of prevalence.18 The reported prevalence in Japan is also within this range (0.25:100,000).27 Discrepancies between these reports are likely a result of different methodologies and their influences on various forms of ascertainment bias. Additional factors in apparent geographic variance in incidence/prevalence could be due to access to health care, rate at which patients are properly diagnosed, or population genetic differences.
NATURAL HISTORY
Pathogenesis
With the exception of some known monogenic forms of CVID (discussed below), no cause for the immune defect has been found in 98% of the patients with CVID. With the hallmark of hypogammaglobulinemia, the immune defect common to all patients with CVID is loss of B-cell function. This is either intrinsic to B cells, or a result of insufficient help from other cells for antibody production. In particular, there is a reduction in the number and percentage of isotype-switched B cells in a majority (not all)28,29 and a loss of plasma cells in both bone marrow and mucosal tissues.30,31 The causes of these abnormalities remain largely obscure.
Patients with CVID have often been stratified on the basis of peripheral blood B- and T-cell phenotype (discussed below) and in vitro B-cell function, using a number of stimuli. In most cases, B cells of some subjects produce normal amounts of immunoglobulin in culture, others produce only IgM, and others are unable to produce any immunoglobulin at all.32 One recent analysis using flow cytometric phenotyping and measurement of kappa-deleting recombination excision circles (KRECs, see below) distinguished 5 possible subgroups or defects: (1) B-cell production, (2) early peripheral B-cell maturation or survival, (3) B-cell activation and proliferation, (4) germinal center, and (5) postgerminal center.33 In some patients, calcium flux after activation of the B-cell receptor may be impaired (note that these correspond to low switched memory B cells) with expansion of the CD21low population (see Table III).34 Others have demonstrated distinct signaling abnormalities in subpopulations with CVID. For example, stimulation of B cells and/or plasmacytoid dendritic cells via Toll-like receptors 7, 8, and 9 is inhibited in some patients; this correlates with lower levels of switched memory B cells.35
TABLE III.
Nearly absent B cells (<1%†) | Includes all patients with severe defects in B-cell differentiation |
Low switched memory B cells (<2%) | Indicates a defective germinal center development similar to |
CD27+IgM−IgD− | • ICOS deficiency |
• CD40L deficiency | |
Increased risk: | |
• Splenomegaly | |
• Granulomatous disease | |
Expansion of transitional B cells (>9%) CD38hiIgMhi |
Associated with lymphadenopathy |
Expansion of CD21low B cells (>10%) | Associated with splenomegaly |
ICOS, Inducible T cell co-stimulator.
Adapted from Wehr et al.29
Expressed as a percentage of total B cells (CD19+ or CD20+).
A relative loss of T-cell function in many subjects has been demonstrated, including a lack of circulating CD4 T cells,9 and especially, naive CD4 T cells,36 antigen-specific T cells,37,38 and impaired proliferation, activation,39 and secretion of some cytokines (IL-2, IFN-γ, and IL-10)40,41 with increases in IL-642 and perhaps IL-12.43 More recently, defects in thymic maturation,44–46 monocyte/dendritic cell defects,47 and impaired innate immune responses48 including loss of natural killer cells49 have also been demonstrated. It is not simple to dissect the primary and secondary changes observed in CVID, especially because the mechanisms are likely to be heterogeneous.
Studies of the bone marrow of patients with CVID show depletion of plasma cells in 94%; the degree of depletion correlates with serum immunoglobulin levels.30 The presence of aggregates of T cells in the bone marrow correlates with autoimmune cytopenias. In 9 of 25 (36%) patients, a block in the pre-B I-II transition correlated with lower peripheral blood B-cell numbers.
Clinical manifestations
In a recent large (2212 patients) study from Europe, a large proportion (34%) of the patients had disease onset before age 10 years.50 There was a reported 2:1 male predominance before age 11 years, this was less pronounced up to age 30 years, and there was a slight female predominance (1.3:1) after age 30 years. Mendelian inheritance is relatively infrequent (between 5% and 25%), displaying both autosomal-dominant and autosomal-recessive patterns and depending at least partly on regional genetic factors11,50,51 In a large American cohort of 248 patients, the age of onset of symptoms was found to be bimodal, with peaks in the first and third decades.52 However, in a European cohort of 413 patients, the age of onset was found to be a continuous curve, with a mean age of 35.3 years and median of 33 years.53 The more recent European cohort suggested again a slightly bimodal onset curve similar to the earlier American report.50 The early peak may reflect the disproportionately higher rate of diagnosis in young males.
There may be significant delay between the onset of symptoms and the establishment of the diagnosis of CVID. In the American cohort, this delay was approximately 5 to 6 years.52 In an older report of a European cohort, there was a mean diagnostic delay of 7.5 years (median, 5 years; range, 0–61 years)53 and 8.9 years in an Italian cohort.11 In a more recent European report, the delay was approximately 4 to 5 years, overall.50 In this cohort, there was no reduction in the delay in patients diagnosed after the year 2000 in comparison to those diagnosed before. However, the delay was considerably shorter in those patients with onset of symptoms after age 10 years, in comparison to those who became ill at younger ages (3.1 vs 7.2 years, respectively).
The first literature report of CVID appeared in 1954 and described a 39-year-old woman with agammaglobulinemia who had many of the observed complications of this immune defect, including chronic bronchitis, episodes of bacterial pneumonia, Haemophilus influenzae meningitis, chronic diarrhea and malabsorption leading to weight loss, and severe hypocalcemia.54 Frequent and severe infections are common at the time of diagnosis of CVID. Complications related to immune dysregulation are not present in all patients. Depending on details in how various patient cohorts were studied, approximately 33% to 80% do not develop these complications at all (“infection-only” phenotype), though they may have infection-related structural changes such as bronchiectasis.8,9,55 The other 20% to 67% of the patients may develop various additional clinical problems including autoimmunity, interstitial lung disease, granulomatous disease, liver disease, gastrointestinal inflammatory disease, lymphoid hyperplasia, and/or cancer or lymphoma. These are all discussed in greater detail below.
There are significant differences in the prevalence of these complications between countries in Europe and in comparison to other large cohorts.11,53,56,57 With increased reporting of patients with CVID from newly established registries around the world, new patterns of complications of CVID may emerge.
Unusual infections
Recurrent urinary tract or uterine cervical infections due to Ureaplasma urealyticum have been described.58 Arthritides and lung infections may also be caused by Ureaplasma or Mycoplasma organisms, and these should be considered when other common pathogens are not found.59–61 Enteroviral infections may cause meningoencephalitis or a dermatomyositis-like syndrome in undiagnosed patients not receiving IgG, or in those who are receiving lower (inadequate) doses (0.1–0.3 g/kg/month).62 The mortality of this complication is high (50%). The occurrence of other opportunistic infections should raise suspicion for a combined immunodeficiency or diagnosis other than CVID.9
Chronic lung disease
Airway inflammation is common in CVID and may progress over time to obstructive or restrictive disease and bronchiectatic changes evident on computed tomography (CT).11,53,63 Bronchoscopy may be needed to identify pathogenic organisms. Lavage culture may include organisms potentially not susceptible to antibody clearance such as nontypeable H Influenzae and/or viruses.64 Mycoplasma and Ureaplasma organisms are prominent pathogens for sinopulmonary as well as nonrespiratory infections (see below).61
Mediastinal lymphadenopthy is common in CVID. When nodes are large, or coupled with larger or persistent nodules in the lung fields, an open lung biopsy is required to determine whether the lung collections are scars, lymphoid cells, clonal proliferations, granulomatous infiltrates resembling sarcoidosis, or malignancy. Lymphoid interstitial pneumonia or follicular bronchitis/bronchiolitis may lead to fatigue, cough, shortness of breath, and alveolar damage resulting in reduced gas transfer.65 On high-resolution chest CT, pulmonary interstitial infiltrates appear as reticulonodular changes, marked linear opacities, fibrosis, and a ground glass appearance.66
Although granulomas may be found in many organs in CVID, in the lung, the granulomas are commonly accompanied by lymphoid infiltrations, leading to what has been termed “granulomatous lymphocytic interstitial lung disease.”67,68 In a series, open lung biopsy revealed that 11 of 12 patients with clinical/radiographic diagnosis of granulomatous lymphocytic interstitial lung disease had granulomas and all had follicular bronchiolitis and lymphoid interstitial pneumonitis (J.M. Routes et al., unpublished data, 2015). The granulomas, although not necrotizing, are not perilymphatic as in sarcoidosis. Furthermore, follicular bronchiolitis and lymphoid interstitial pneumonitis are not prominent in sarcoidosis, suggesting that granulomas in the lung represent a different pathophysiology. Other series have shown different combinations of pathologies, suggesting various etiologies depending on the locality or genetic background of the patients.65,69 The evolution of granulomas or interstitial pneumonitis in CVID is not yet well understood. Early stages may be asymptomatic. It is recommended that patients have a chest CT scan and measurement of gas transfer obtained at some time relatively close to the time of diagnosis to provide a baseline for comparison in the future. Whenever suggestive findings occur, this diagnosis (or other) must be formally established by histopathological analysis. Ongoing inflammation leads to lung damage and diminished survival. Chronic lung disease is a major cause of mortality in CVID.8,50,70,71
Diffuse granulomatous disease
Granulomatous disease or atypical sarcoidlike lesions occur in 8% to 22% of the patients with CVID and may be discovered on the basis of various organ biopsies years before the recognition of the immune defect.68,72–78 Lungs, lymph nodes, and spleen are commonly affected. However, skin, liver, bone marrow, kidney, gastrointestinal tract, eyes, and/or brain may also be involved.74,79–81 The granulomas in CVID are noncaseating, and microbial associations have not been described. The cause of these tissue collections remains unknown.
In individuals who already carry a diagnosis of CVID, the constellation of symptoms and signs described here is not to be confused with sarcoidosis. However, confusion may occur in patients who have undiagnosed CVID. Hematologic abnormalities such as variable cytopenias (generally nonimmune) are common in sarcoidosis, as is monocytosis. Importantly, hyper-gammaglobulinemia is frequently seen in sarcoidosis, and most of the remainder have normal immunoglobulin levels, in contrast to the hypogammaglobulinemia of a patient with CVID. Additional differences between CVID and sarcoidosis include the distribution and size of pulmonary nodules and the very high prevalence of splenomegaly in the former.82 All patients being evaluated for sarcoidosis should have measurement of serum immunoglobulins.
Autoimmunity
Autoimmunity occurs in approximately 25% to 30% of the patients with CVID.50,55 Quinti et al11 described autoimmunity as one of the presenting manifestations of CVID in 17% of 224 patients, and in 2.3%, autoimmune disease was the only clinical complication at the time of diagnosis of CVID.10 The most common of these include cytopenias: immune thrombocytopenia purpura (ITP), autoimmune hemolytic anemia (AIHA), or both at the same or different times (Evans syndrome). Autoimmune neutropenia is much less common.83–85 For unclear reasons, these patients may not have a typical history of recurrent infections and severe cytopenia may be the first manifestation of the immune defect.8 Patients with CVID with granulomatous disease and evidence of lymphoproliferation are more likely to have had episodes of ITP and/or AIHA.72,78 In the recent European cohort report, autoimmunity was correlated with enteropathy, granulomatous disease, splenomegaly and splenectomy, low IgA level, and later age of onset.50 Note that autoimmune manifestations may even occur before the appearance of hypogammaglobulinemia. These patients may appear similar to those with syndromes of immune dysregulation such as autoimmune lymphoproliferative syndrome.86
Other autoimmune diseases in patients with CVID include inflammatory bowel disease, seronegative arthritis, pernicious anemia, Sjogren syndrome, uveitis, vasculitis, thyroiditis, alopecia, vitiligo, hepatitis, primary biliary cirrhosis, sicca syndrome, or systemic lupus erythematosus.10,55,87,88 These do not appear to be linked to the occurrence of cytopenias. Some other common autoimmune conditions, such as insulin-dependent diabetes, psoriasis, celiac disease, hypothyroidism, and seropositive rheumatoid arthritis, do not appear to be increased in CVID.10,50,88–90
Gastrointestinal disease
Some form of enteropathy was found in 9% of the patients studied in the recent European report.50 This was correlated with autoimmunity, splenomegaly, lobectomy, low IgM level, and age of onset. Enteropathy in CVID has a high rate of nonmalignant mortality, possibly due to malabsorption.53,71 Bacterial, protozoal (mainly Giardia lamblia), and viral gastrointestinal infections occur in CVID and often respond to standard medical treatments. Eradication may be difficult for some patients. Even more challenging is a form of chronic small bowel inflammation that occurs in 4% to 12% of the patients, depending on the cohort studied. This is associated with unexplained persistent chronic diarrhea, weight loss, steatorrhea, and malabsorption with loss of both minerals and fatsoluble vitamins.91–93 Vitamin A deficiency is also important as a cause of night blindness as well as having a negative effect on intrinsic immunoglobulin production.94 This has been referred to as CVID-associated autoimmune enteropathy. Bacterial overgrowth is common and can lead to bloating and worsening diarrhea.
Although the pathogenesis of AIE is unclear, on biopsy, the mucosa shows villous blunting and crypt distortion with increased lymphocytes (usually CD8 T cells), lymphoid aggregates (lymphoid hyperplasia), and loss of plasma cells.91,95 Note that lack of plasma cells is a frequent histological feature in CVID.
Some authors conclude that the absence of plasma cells and increased lymphoid hyperplasia reliably distinguish AIE from celiac disease.92 Others suggest that celiac disease–associated HLA-DQ markers in patients with CVID may help distinguish AIE due to immune dysregulation.96 That is, if celiac disease markers are absent, a diagnosis of AIE is more likely. Although MHC genetic tests may be helpful, serologic tests (transglutaminase and other antibodies) usually are not. Mild hepatomegaly with persistently increased liver enzyme levels, including alkaline phosphatase, is frequent in CVID. The most commonly identified cause of liver abnormalities is nodular regenerative hyperplasia.97–99 Although previously thought to have a relatively benign course, a significant proportion of patients progress to autoimmune hepatitis with typical histopathological changes and/or develop portal hypertension. These patients frequently have hypersplenism with neutropenia and many succumb to infection. Gall bladder disease with cholestasis, primary biliary cirrhosis, and autoimmune or granulomatous hepatitis are also potential causes of liver abnormalities. Hepatitis B and C are uncommon in CVID unless risk factors are present.
Allergic disease
Recent studies of allergic respiratory diseases in CVID have revealed low incidences of allergic asthma, 6.5% of 62 patients as a whole and 22.2% of the 18 patients with a clinical history suggestive of asthma.100 Likewise, the same group found allergic rhinitis to be equally unusual.101 Although 82% of the patients had a history of chronic or episodic rhinitis or rhinosinusitis, an allergic cause was confirmed only by detection of specific IgE to aeroallergens in 5.6% of the 72 patients with CVID with a clinical history suggestive of the diagnosis. This is in contrast to patients not having CVID with low or absent IgA levels with or without IgG subclass deficiency in whom allergic diseases, particularly asthma, are more common.102–104
Lymphoid hyperplasia
Cervical, mediastinal, and abdominal lymphoid hyperplasia and/or splenomegaly are found in at least 20% of the patients with CVID105,106; splenomegaly was reported in 26% of the recent European cohort.50 Biopsies of lymph nodes usually show atypical lymphoid hyperplasia, reactive lymphoid hyperplasia, or granulomatous inflammation. Lack of plasma cells and the presence of ill-defined germinal centers in lymph nodes and other lymphoid tissues are characteristic.107 These tissues need to be examined for B- and T-cell clonality, using fluorescence markers, cytogenetics, and/or molecular analysis to rule out lymphoid malignancy. For B-cell infiltrates, examination for EBV-encoded RNAs by in situ hybridization can be performed. Because subjects with CVID can have unusual lymphoid structures with loss of characteristic boundaries, it is important for an experienced pathologist to examine these tissues. However, the presence of clonal lymphocytes is not diagnostic of lymphoma because these can be found in CVID lymphoid tissue showing reactive hyperplasia.106
As noted above, an increased spleen size is a common finding in patients with CVID but neither its causes nor its consequences are well understood. Splenomegaly can be massive and yet not cause clinical symptoms. Histological descriptions of spleen abnormalities after splenectomy showed granulomatous lesions, congestive red pulp, follicular hyperplasia, and atrophic germinal centers/white pulp. These abnormalities might contribute to a splenic sequestration as the cause of thrombocytopenias in patients with CVID.105,106
Malignancy
Malignancies of all types are increased in patients with CVID compared with the general population, occurring in possibly 6% to 9% of the patients. Most of the literature to date indicates that lymphomas are the most common form of malignancy in CVID. For 176 subjects in a European study, the observed to expected ratio for lymphoma in CVID was 12:1 and for stomach cancer was 10:3.108 However, suggesting a potential downward trend of stomach cancer in recent years, there were 3 stomach cancers in 476 patients (0.6%) in contrast to 32 non-Hodgkin lymphomas (6.7%) and 4 cases of Hodgkin disease.71 In a country where H pylori is prevalent, this organism was found in 14 of 34 subjects with gastric symptoms, 1 of whom had gastric cancer.109 In a large Italian cohort of 353 patients, adenocarcinoma was an initial presentation in 5 (1.4%).70 A recent report from a large (2,212 patients) European database found solid tumors outnumbering lymphoma at a rate of 5% versus 3%, respectively.50
Other complications
Various additional clinical problems may arise in patients with CVID. The relationship of these to the underlying pathophysiology remains unknown. One such potential association is osteoporosis in association with increased bone turnover.110,111 Hypothyroidism is also occasionally seen, although not with increased frequency compared with the general population.112
Laboratory manifestations
Despite the heterogeneity of patients with CVID, large population retrospective studies both in the United States and in Europe have suggested that IgG levels of less than 4.5 g/L are found in most patients with CVID (85%–94%, respectively).11,52,53 IgM levels are variable. In the series from the United States, approximately 80% of the patients had IgM levels of less than 0.25 g/L, whereas the mean IgM level was 0.4 g/dL in an Italian cohort of 224 patients with CVID. In one study, females with CVID tended to have higher levels of IgM.71 IgA levels are low or undetectable in CVID, with 70% of the patients demonstrating values of less than 0.1 g/L in the US cohort vs 49% of the patients with IgA levels of less than 0.07 g/L in the European cohort. (As noted in the section on Definition, some experts require that the IgA level be below the age-adjusted normal range for all patients given a diagnosis of CVID.) There are several reported cases of patients with selective IgA deficiency with normal IgG level at initial evaluation in whom IgG levels slowly decline until they fulfill laboratory criteria for CVID.113 Finally, up to 21% of the patients with CVID may have very low levels or absence of all immunoglobulin isotypes at presentation.29,50
Specific antibody production may be variable in some patients with CVID. Antigen-specific IgG levels or vaccine responses in patients suspected to have CVID may be within normal limits at initial presentation, but may decrease over time, ultimately becoming consistent with the diagnosis. In a small study of childhood CVID, a large proportion of children (73%) retained normal isohemagglutinin titers and specific antibody responses to protein antigens were protective at the time of diagnosis in 44% to 62%.114 In contrast, absent responses to pneumococcal polysaccharide antigens was noted in 71% of children, whereas such response was impaired in 21% of children. In a study of 21 adults with CVID and receiving IgG replacement, about half responded to at least 1 of 5 different protein/peptide or conjugate vaccines.115 Four of 21 responded to more than 1 protein vaccine, and 3 of 17 made some measurable antibody to pneumococcal polysaccharide.
Some patients will have clinical manifestations of CVID, but may not fulfill laboratory criteria because of their IgG or other isotype level being too high, or their vaccine responses appearing to be adequate. These patients must be followed longitudinally. As noted above, milder laboratory phenotypes such as IgA deficiency or IgG subclass deficiency may evolve over time until laboratory criteria are met and a diagnosis of CVID is appropriate.
Most patients with CVID will have normal levels of total circulating T cells and natural killer cells in peripheral blood.49,52 However, B-cell numbers are variable. In the 2008 retrospective review of European data comprising mainly adult patients with CVID, 54% of the patients had normal levels (6%–16%) of circulating B cells, 19% had increased levels (>17%), 12% had reduced levels (1%–6%), and 12% had undetectable levels.53
Efforts to study patients with CVID have been fraught by the heterogeneous nature of the disorder. Thus, for more than a decade now, efforts have been put forth to systematically classify patients with CVID by using easily accessible standard flow cytometry criteria (Table III).28,29,116,117 Such immunophenotypic classifications have permitted insights into the pathogenesis of clinical manifestations. Thus, the EUROclass trial was designed to bring consensus among several classification schemes.29 Based on B-cell immunophenotyping, levels of class-switched memory B cells, defined by the expression of cell surface markers CD27+IgM−IgD−, have been found to be associated with splenomegaly, granulomatous disease, possibly chronic lung disease, and autoimmunity. The added value of identification of transitional B cells (CD38hiIgMhi) and CD21low B cells also appear to aid in defining subcategories having potential clinical relevance such as lymphadenopathy and splenomegaly, respectively.
Variable dysfunction within the T-cell compartment has been reported in groups of patients with CVID.36 One study found that CD4 T cells were decreased in 29% of the 473 patients with CVID, while 50% of this cohort demonstrated abnormal proliferative responses to at least 1 mitogen.71 Reduced numbers of naive T cells are also commonly observed as reported by the French DEFI group of 311 patients with CVID and others.9,118 This decrease in the naive CD4 population (CD45RA+CCR7+CD4+) is most pronounced among those patients who meet criteria for late-onset combined immune deficiency (LOCID, see below). Measurement of T-cell receptor excision circles may distinguish CVID subgroups (see below).
Restricted T-cell receptor repertoires, oligoclonality, increased T-cell apoptosis, and reduced expression of CD40L have been noted in some patients, indicating that T- cell function is not normal in subgroups of patients with CVID.119–121 Because B cells require help from CD4 T cells for response to (glyco-) protein antigens, T-cell abnormalities may contribute to defective antibody production in patients with CVID. However, in many patients, T cells may be able to undergo activation and terminal differentiation, with ability to generate specificity to cytomegalovirus, EBV, and influenza virus, supporting a potential benefit of influenza subunit vaccination for most patients with CVID.122
Dendritic cells and regulatory T cells may also have roles in the pathogenesis of some phenotypes or complications of CVID, such as autoimmune or inflammatory manifestations.41,123,124 The mechanisms underlying these cellular abnormalities in CVID are yet to be fully elucidated.
In a French cohort of 311 patients with CVID, 9% had either an opportunistic infection, a CD4 T-cell count of less than 200 cells/µL, or both. In this group of patients, intestinal disease, splenomegaly, lymphomas, and granulomas occurred with greater frequency than in the remainder of the cohort.9 This phenotype was named LOCID and is now classified as a combined immunodeficiency by the International Union of Immunological Societies. It is notable that consanguinity was more common in this group, suggesting possible Mendelian genetic defect(s). This phenotype is very similar to Good syndrome although thymoma, characteristic of Good syndrome, is not seen in LOCID.125
GENETICS
CVID is a complex, multifocal disease, the genetic origins of which are beginning to be at least partially understood. Most cases of CVID are sporadic. Approximately 5% to 25% are familial, with an autosomal-dominant pattern of inheritance being more frequently observed.51,126 Rarely, families exhibiting autosomal-recessive inheritance have been reported.
A number of studies concentrated on CVID/IgAD families revealed several putative susceptibility loci identified within the HLA region on chromosome 6p.127 Linkage analysis indicated a susceptibility locus termed IGAD1. Other selected HLA-DQ/DR haplotypes conferred either protection or susceptibility to IgAD and CVID. The strong influence of the MHC region has been noted in several other cohorts. In one, most of the patients inherited HLA *DQ2, *DR7, *DR3, *B8, and/or *B44. B44 was present in almost half and was the most common susceptibility allele.128 Genetic linkage studies in such families have found evidence of causative mutations on chromosome arms 4q51 and 16q,129 but disease-associated genes have not been identified.
To date, mutations in various nonredundant genes have been shown to be disease causing in some patients who fulfill criteria for CVID. Some of the clinical and laboratory features of these disorders are summarized in Table IV. However, these disease-causing mutations currently account for only a very small fraction (about 2%) of the population with CVID. Furthermore, many patients with mutations in some of these genes (eg, CTLA4 and LRBA) exhibit combined immunodeficiency with clinical features or laboratory abnormalities not often seen in CVID. These have been segregated in the CVID classification (from those with no known defect) by the International Union of Immunological Societies and are classified as distinct entities on the basis of defined genetic defects.154
TABLE IV.
Gene | Clinical features | Autoimmunity | Laboratory features | References |
---|---|---|---|---|
CD19 | Recurrent respiratory infections | No | Low IgG and IgA level, poor PS vaccine response, normal total B cells, low memory B cells |
130,131 |
CD20 | Recurrent respiratory infections | No | Low IgG level, poor PS vaccine response, normal total B cells, low switched memory B cells |
132 |
CD21 | Recurrent respiratory infections, diarrhea | No | Low IgG level, normal vaccine responses, normal total B cells, low switched memory B cells |
133 |
CD27 | EBV-associated LPD, lymphoma, recurrent sinusitis in some |
No | Low IgG or IgA level in some, poor vaccine responses, T-cell dysfunction, normal total B cells, absent memory B cells |
134,135 |
CD81 | Recurrent respiratory infections, Henoch-Schonlein purpura, glomerulonephritis |
ITP | Low IgA level, poor vaccine response, normal total B cells, low memory B cells |
136 |
CTLA4 | Autosomal-dominant, respiratory infections, diarrhea, lymphoid organ infiltration |
Various | Low IgG and/or IgA levels, poor vaccine response, low total B cells, and low switched memory B cells |
137,138 |
ICOS | Recurrent respiratory, gastrointestinal infections | Present in some | Low IgG and IgA levels (low IgM level in some), poor vaccine response, low total B cells, low memory B cells, poor CD4/CD8 effector T-cell function |
139,140 |
IL21 | Recurrent respiratory infections, cryptosporidium, other severe bacterial/viral infections |
No | Low IgG level, high IgE level, poor vaccine response, low B cells, low memory B cells, poor T-cell cytokine production, low natural killer–cell cytotoxicity |
141,142 |
IL21R | Recurrent respiratory infections, IBD | IBD | Low IgG level, high IgE level, poor vaccine response, low B cells, low memory B cells, low T-cell antigen response |
141,143 |
LRBA | Recurrent respiratory infections, enteropathy | ITP, AIHA, others | Low IgG, IgA, and IgM levels, poor vaccine responses, low/normal total B cells, low switched memory B cells |
144–146 |
NFKB2 | Autosomal-dominant, recurrent respiratory infections, meningococcal meningitis, adrenal insufficiency |
ITP | In some: low IgG and/or IgA/IgM levels, poor PS vaccine response, normal total B cells, low switched memory B cells |
147,148 |
PIK3CD | Autosomal-dominant, recurrent respiratory infections, bronchiectasis, severe herpesvirus infections, lymphoma |
No | Low IgG level, low IgG2 level, poor PS vaccine response, variable low T cells and B cells, low switched memory B cells, high transitional B cells |
149–151 |
RAC2 | Recurrent respiratory infections, post-strep. GN | Thyroid | Low IgG, IgA, and IgM levels, poor PS vaccine response, low/normal B cells, low naive T cells |
152 |
TWEAK | Autosomal-dominant, recurrent respiratory infections, pneumococcal meningitis, warts |
No | Low IgG (or low IgG2/4), IgA, and IgM levels, poor vaccine response, low/normal total B cells, low memory B cells |
153 |
IBD, Inflammatory bowel disease; LPD, lymphoproliferative disease; post-strep. GN, post–streptococcal glomerulonephritis; PS, polysaccharide. All are autosomal recessive unless otherwise specified.
DNA repair variants may account for some forms of CVID. With inappropriate DNA repair, a predisposition to radiation damage and cancer are plausible outcomes. Subjects with CVID have an increased propensity to cancer, and some appear to have increased cellular radiosensitivity. Genes encoding elements important for DNA mismatch repair have been associated with IgA deficiency and CVID.155
Particular polymorphisms in the TACI (TNFRSF13B) and MSH5 genes may affect the phenotype of about 5% to 8% of the patients with CVID, though they are also present in a significant number of healthy individuals (~1%).154,156 TACI polymorphisms may impair T-cell independent class-switch recombination because interactions between TACI and its ligands (APRIL [a proliferation-inducing ligand] and BAFF [B cell activating factor]) are important for this process.157,158 Most individuals with biallelic TNFRSF13B polymorphisms develop CVID, whereas those with single-allele mutations or rare variants are at an increased risk for developing CVID and autoimmune phenomena.159,160 One kindred with a CVID-like phenotype has been found to have mutations in TNFRSF13C (BAFFR), with both siblings having reduced IgG and IgM levels but persistently normal IgA levels.161 A recent report indicates that polymorphisms in this gene are fairly common and may contribute to an altered B-cell response.162
Thus far, the defects described have mainly been monogenic, but it is anticipated that many of the genetic causes of CVID yet to be characterized will be polygenic, and modifier genes may play crucial roles in the development of disease. For example, polymorphisms in genes encoding IL-10 and TNF have been associated with granuloma formation.163 Polymorphisms in genes encoding alpha-1 antitrypsin and mannose-binding lectin may affect the occurrence of bronchiectasis and pulmonary fibrosis.164,165
The identification of novel disease-causing genes is further complicated by the fact that the same clinical disorder may be caused by mutations in different genes and that mutations in different locations of the same gene may give rise to completely different primary immunodeficiency disorders.154 In addition, epigenetic overlay, as illustrated by IgA-deficient discordant monozygotic twins,166 may be expected to add to the etiological complexity.
The heterogeneous nature of CVID is further confirmed after a genomewide association study identified multiple potential susceptibility loci for CVID.4 Three hundred and sixty-three patients with CVID were genotyped. Single nucleotide polymorphism (SNP) associations and copy number variations were recognized and distinguished the CVID cases from control subjects. The strongest associations with CVID were found in MHC and disintegrin/metalloproteinase gene loci. There were 16 associated gene duplications/deletions and many (~100) unique intraexonic duplications and deletions. A total of approximately 1,000 composite SNPs predicted the CVID phenotype. SNP analysis revealed genes associated with particular phenotypes, including mitogen-activated protein 3 kinase 7-interacting protein 3 (MAP3K7IP3) significantly associated with low IgA level. Genes significantly associated with lymphoma including PFTK1, HAVCR1, and KIAA0834 were also found. The gene CACNA1C (calcium channel, voltage-dependent, L type, alpha 1C subunit) was common to both patients with CVID and enteropathy and patients with inflammatory bowel diseases.167
Note again that in our current consensus definition of CVID (see item 6), molecular genetic analysis of patients is not a requirement for conferring the diagnosis. However, in light of the possible definitive or supportive therapies (eg, stem cell therapy and cytokine therapies) that may be afforded to patients with specific genetic defects, consideration should be given to pursuing a molecular diagnosis, if possible. This applies especially to patients exhibiting clinical or laboratory features that are unusual in CVID. Assessment of patients’ relatives may distinguish familial from sporadic CVID.
DIAGNOSIS
Clinicians must maintain an index of suspicion for antibody deficiency in patients of all ages. Conditions associated with acquired hypogammaglobulinemia should always be considered during the evaluation of a patient with a suspected antibody deficiency. These include drugs (chronic glucocorticosteroid use, antiepileptic drugs, rituximab therapy), malignancies (chronic lymphocytic leukemia, lymphomas), nephrotic syndrome, protein-losing enteropathy, and congenital lymphangiectasias. See Table I for these and a more complete list of diagnoses to be excluded before making a determination of CVID.
Regarding glucorticoid use, daily doses of more than 20 mg for 14 days in individuals (or doses >2 mg/kg in children weighing <10 kg) or lower doses over longer periods (months to years) may lead to hypogammaglobulinemia.168,169 There is also a possibility that vaccine responses may be impaired when steroids are used in high doses and for prolonged periods. Although hypogammaglobulinemia with systemic steroid use is well documented, impaired vaccine response is more theoretical; there has been little formal prospective assessment of vaccine responses in patients on systemic steroid therapy. One study found no effect on responses to yellow fever vaccine.170 Chronic inhaled steroids did not impair responses to influenza vaccine in another study.171
In light of the definition of CVID discussed above, the minimum laboratory studies that should be performed during evaluation include measurement of serum immunoglobulin levels and vaccine responses to at least 1 T-dependent and 1 T-independent antigen. For maximum diagnostic specificity, flow cytometry analysis of peripheral circulating lymphocytes to delineate T-, B-, and natural killer–cell populations to exclude combined immunodeficiency is essential. Measurement of B-cell subtypes may also be helpful for differentiating CVID from other disorders.
Serum immunoglobulins
Levels of 4 major isotypes—IgG, IgM, IgA, and IgE—should be measured. At birth, IgG levels are high due to transfer of maternal IgG during gestation. Levels fall to a nadir between age 3 and 6 months and then increase steadily thereafter. Thus, age-related local normal ranges must be used. Wherever possible, population-specific ranges should also be applied to account for racial or environmental differences. Normal values are usually reported as 2 SDs above or below the mean or the 5th to 95th percentile intervals. Because the distribution of serum immunoglobulins is nonparametric, percentile criteria for normality may be preferred, but this is not the rule in all laboratories. Regardless, it is important to recognize that 2.5% to 5% of normal subjects may fall below the defined norms for each age group.
It is common to find discrepancies between laboratories. In addition, intercurrent illnesses, fluid shifts, or other processes may affect immunoglobulin levels. Because of such variability, diagnosis merits demonstration of persistently low serum immunoglobulin levels (as discussed previously) before starting IgG therapy. However, IgG therapy should not be unduly delayed if there is reasonable suspicion of the disease based on other features and delay might be harmful to the patient’s health.
Quantitation of IgG subclasses is not relevant to the diagnosis of CVID. Measurement of IgE level is not usually necessary to make a diagnosis of CVID, but an elevated concentration of IgE is unusual in this setting and should prompt consideration of an alternative diagnosis and/or investigation of immune dysregulation. When IgM levels are either normal or elevated in conjunction with low IgG and IgA levels, the clinician should consider one of the hyper-IgM syndromes or defects of class switch recombination before establishing a diagnosis of CVID.
Assessment of vaccine responses
Vaccine responses should be determined in all patients except those who present with very low or undetectable IgG levels. Even with very low IgG levels (100–200 mg/dL), adequate levels of vaccine-specific IgG may be found and could lead one to consider secondary hypogammaglobulinemia or other diagnoses. Of course, this evaluation should always be performed before initiating IgG therapy. Note that some experts do not require measurement of vaccine responses in all patients. For example, in a patient with characteristic clinical features having low IgG and IgA levels and abnormally low memory B cells with severe sinopulmonary infection, the practitioner may choose to forego the month needed to assess vaccine response in favor of immediate initiation of IgG infusions for therapeutic benefit. In other patients, detection of specific antibodies to known/documented previous infections (such as measles and herpes simplex) or previous immunizations may suffice to rule out primary antibody failure. These decisions must be made on a case-by-case basis. The consensus of the authors of this document is that vaccine responses should be measured in all patients except in cases in which the treating physician judges the potential morbidity of delaying IgG therapy to be unacceptable.
The quality of an antibody response is complex and encompasses many different aspects of antibody function including clonal diversity, binding affinity/avidity, opsonizing and neutralizing capacity, and the effective development of immunologic memory capable of eliciting an anamnestic response. In clinical practice, a patient’s ability to mount normal functional antibody responses is assessed by measuring the serum levels of antigen-specific IgG antibody in response to vaccine antigens or documented infection, and should include evaluation of both T-dependent responses (to protein or glycoprotein antigens) and T-independent responses (to polysaccharide antigens).
Common vaccines available in most countries include tetanus and diphtheria toxoids, Haemophilus influenza type B, and pneumococcus. The first 3 of these are composed of protein antigens or a polysaccharide antigen coupled to a protein carrier (conjugate vaccine), and measurement of specific IgG levels to these vaccine antigens provides information on T-dependent responses. Pneumococcal vaccines may be either conjugate (eg, Prevnar-13 [United States] or Prevenar-13 [Europe]) or pure polysaccharide (eg, Pneumovax). T-independent responses are evaluated by measurement of the specific IgG response to pure polysaccharide antigens contained within the pneumococcal polysaccharide vaccine. Other vaccines of diagnostic value include those for meningococcus (both protein-conjugate and polysaccharide preparations) and pure polysaccharide salmonella vaccine. Measurement of antibodies for other routine childhood vaccines such as measles, mumps, rubella, polio, hepatitis B, and varicella may sometimes be helpful. Assessment of responses to pathogens the patient is known to be infected with (eg, herpes simplex virus, cytomegalovirus, and EBV) may also be informative.
The approach to evaluation of functional antibody responses to T-dependent antigens is to first measure serum levels of specific IgG to various vaccines. If antigen-specific antibody levels are low on initial measurement, immunization followed by repeat measurement of specific antibody levels 3 to 6 weeks later (4 weeks is often considered standard) should be performed to assess production of specific antibody. Diphtheria and tetanus toxoid vaccines are the most commonly used vaccines to evaluate T-cell–dependent responses in both children and adults.
Evaluation of functional antibody responses to T-independent antigens requires measurement of postimmunization specific IgG level because high titers of naturally occurring cross-reactive antipolysaccharide antibodies that correlate poorly with functional activity are commonly present in preimmunization sera.172 The polysaccharide pneumococcal vaccine is the most commonly used vaccine to assess T-independent response and may be used reliably in adults and in children older than 12 months.173,174 Recent studies demonstrate normal functional antibody responses to Pneumovax in 12-month-old infants, supporting its use in the evaluation of T-independent responses in even younger children.174,175
Protective levels of specific IgG to many protein and conjugate vaccine antigens have been established. Common examples include diphtheria and tetanus toxoids (0.15 IU/mL) and Haemophilus influenzae type B (HIB) polysaccharide (1 µg/mL). Levels below these thresholds are suggested to indicate impaired specific responses to these antigens, although this has not been formally established. Some patients with CVID receiving tetanus do make a “protective” response (by concentration criteria) but still need replacement immunoglobulin therapy to prevent bacterial infections.115
Studies from immunization trials evaluating the efficacy of pneumococcal conjugate vaccines suggest that a titer of 0.35 µg/mL or more is protective against invasive pneumococcal disease (not otitis media, sinusitis, or nasal colonization) in healthy infants.176 Specific criteria regarding the number or fraction of serotypes to which an individual responds after receiving a conjugate pneumococcal vaccine have not been developed, if serotype-specific assays are used. Specific IgA and IgM antibody responses to pneumococcal capsular polysaccharides are being studied, but these tests are not routinely available in clinical practice. In the future, these specific IgA and IgM responses may provide an additional way to assess antibody responses in patients on IgG therapy and further characterize CVID phenotypes.115,177
Criteria for the interpretation of specific antibody responses to pure pneumococcal polysaccharide antigens have been developed. Although the level of protective antibody for an individual serotype may be defined, there is tremendous individual variability in healthy people with respect to the response pattern across serotypes. Following immunization with a pure polysaccharide pneumococcal vaccine, 1.3 µg/mL type-specific IgG is considered protective against invasive disease for that type.178 Recently, consensus criteria have been proposed by an expert group regarding the proportion of types to which an individual responds and the change in preimmunization and postimmunization levels.179 Four response phenotypes are defined (summarized in Table V). Note that these criteria exclude serotypes contained within pneumococcal conjugate vaccines if previously vaccinated with these (within ~5 years) because of the effect of priming. These new criteria have not been formally validated. Many European laboratories use either whole Pneumovax or Typhum Vi vaccines for evaluating polysaccharide responses.180
TABLE V.
Phenotype* | PPV23 response, age >6 y |
PPV23 response, age <6 y | Notes |
---|---|---|---|
Severe | <2 protective titers (>1.3 mg/mL) | <2 protective titers (>1.3 mg/mL) | Protective titers present are low |
Moderate | <70% of the serotypes are protective (>1.3 mg/mL) |
<50% of the serotypes are protective (>1.3 mg/mL) |
Protective titers present to >3 serotypes |
Mild | Failure to generate protective titers to multiple serotypes or failure of a 2-fold increase in 70% of the serotypes |
Failure to generate protective titers to multiple serotypes or failure of a 2-fold increase in 50% of the serotypes |
2-Fold increases assume a prevaccination titer of less than cutoff values of 4.4–10.3 µg/mL, depending on serotype |
Memory | Loss of response within 6 mo | Loss of response within 6 mo | Adequate initial response to >50% of the serotypes in children aged <6 y and >70% in those aged >6 y |
All phenotypes assume a history of infection. Reproduced from Orange et al179 with permission.
It should also be noted that simply measuring the level of antibody does not give complete information regarding the quality of the immune response. It must be considered that in some individuals, currently available clinical laboratory tests may not always adequately define impairment of antibody production. For example, some individuals with “protective” levels of pathogen-specific IgG antibody may nevertheless not be protected by virtue of producing an antibody with low avidity or low opsonophagocytic activity.181,182 However, there are currently no accepted clinical criteria regarding normal vaccine response based on measurements other than specific serum IgG antibody concentration.
Measuring titers of isohemagglutinins, naturally occurring antibodies of IgM and IgG isotypes to polysaccharide blood group antigens, may be an alternative clinically relevant analysis to assess T-independent antibody function in young patients, or in those who have already been started on IgG therapy. Rabies or tick-borne encephalitis vaccines may also be useful diagnostically in patients who are already receiving IgG and have not received these vaccines previously.130 It is also possible to use standard vaccines to assess responsiveness in patients receiving IgG therapy (see section on Laboratory Manifestations above), although interpretation of results must be adjusted.115 This is not considered a standard practice. Alternative methods using flow cytometric measurement of B-cell activation following immunization are being developed.183
MANAGEMENT
Immunoglobulin replacement therapy
IgG can be given by intravenous or subcutaneous route and at varying intervals to suit the patient’s specific needs.184 Both types of preparations are efficacious, safe for infusion at home, and widely available, being on the World Health Organization Essential Medicines Lists for adults and children. Patients require monitoring of breakthrough bacterial infections and serum trough or steady-state IgG levels because each individual has a unique threshold level of IgG to prevent breakthrough bacterial infections.8,185,186
The required IgG dose for an individual patient is unknown, so most national and international guidelines suggest a starting dose of 0.4 to 0.5 g/kg/month for both intravenous immunoglobulin (IVIG) and 0.4 to 0.6 g/kg/month for subcutaneous immunoglobulin (SCIG).187 If there is preexisting bronchiectasis, there is evidence for using 0.6 g/kg/month.188 Some practitioners also recommend higher doses (0.6–0.8 g/kg/month) for patients with enteropathy or splenomegaly.8,189,190 Higher rates of respiratory infection and poorer outcomes are associated with underdosing IgG and vice versa.50,191,192
IVIG infusions are usually given at 3- or 4-week intervals. Adverse events tend to be associated with rapid infusion rates, concurrent acute infections, and previously untreated patients, when a significant time has elapsed between infusions (>6 weeks), or when there is a change in product.193,194
Typical acute reactions associated with IVIG infusions include headaches, nausea and vomiting, flushing, hives, chills, myalgia, arthralgia, or abdominal and/or back pain; these usually resolve with antihistamine therapy or acetaminophen or nonsteroidal anti-inflammatory drugs and slowing of the infusion rate. Delayed reactions (fatigue, headache, myalgias) can occur up to 72 hours after the infusion. Pretreatment and posttreatment with the same medications can also frequently alleviate these delayed minor adverse events. Moderate acute and delayed adverse effects may require the addition of corticosteroids to preinfusion and postinfusion medications. Severe, life-threatening anaphylactoid reactions are very rare and are treated acutely with epinephrine, antihistamines, and corticosteroids.194 Occasionally, switching to a different IVIG product may alleviate adverse effects.
Some patients with CVID with absent serum IgA (<7 mg/dL) may develop IgG anti-IgA antibodies. It is possible that high levels of such antibodies may be associated with anaphylactic reactions to IVIG.23,195,196 However, such reactions are quite rare, and it is not routine to study IgA-deficient patients with CVID for the presence of such antibodies before initiating therapy.
SCIG doses in adults vary from 100 to 200 mg/kg/week and dosing intervals from daily to twice weekly, once a week, or every 2 weeks.197,198 Absorption of IgG administered subcutaneously takes 3 to 7 days.199,200 Some practitioners prefer to initiate therapy with IVIG until the patient is stable and then transition to SCIG, whereas others initiate SCIG directly. IVIG and SCIG can also be given on the same day, particularly if a loading dose is desired at the same time as training in self-infusion of SCIG. Therapy can also be started with SCIG on a daily basis to provide rapid increases in IgG levels while training parents or patients in administration.197,200 With weekly SCIG dosing, steady-state IgG levels are reached after approximately 6 months; the steady-state level can be reached more quickly with 5 daily infusions of the weekly dose followed by weekly infusions.200 It is possible to use SCIG in patients who previously had severe adverse effects with IVIG and in those with high titers of IgG anti-IgA antibodies.23,195,201 Systemic reactions to SCIG are rare; local tissue reactions are common.202 However, these decline over time.203 More recently, it was shown that using a new infusion area (eg, changing from abdomen to thighs) increases the occurrence of local tissue reactions.204
SCIG may also be administered with facilitation by hyaluronidase.205 In this technique, a 10% IgG solution is administered subcutaneously at the same site after infusion of recombinant human hyaluronidase. The enzyme permits infusion and absorption of much larger volumes in comparison to unfacilitated SCIG. Hyaluronidase SCIG is administered according to the same dose regimen as IVIG, the only difference being subcutaneous versus intravenous administration.
IVIG may be administered in a health care facility, by an infusion service in the patient’s home, or self-infused by the patient at home after training. SCIG is usually self-infused at home by a patient or parent. Patient acceptance and safety are proven, quality of life is improved, and there may be cost savings with SCIG, as well.193,206 The push method without a pump has been used for SCIG in Europe for some time because pumps are expensive and cost savings have also been confirmed in Canada.207 Educational programs in specialist centers, with the stated aims that patients should be able to self-infuse at home and that parents be able to perform infusions with their child, have increased compliance with therapy by patients and parents.208
Complications due to transmission of infectious agents, such as hepatitis B or hepatitis C, are now extremely rare.209 However, it is still important to continue to monitor patients on replacement therapy for transmission of unknown or new pathogenic agents. Measurement of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) should be performed once or twice annually; significant persistent elevation should be investigated. Serum may be stored for retrospective analysis in the event of future suspected disease transmission.
Renal complications have been observed in patients receiving IVIG. Most of these are associated with sucrose-containing products and in patients with preexisting kidney disease.210 However, renal injury can occur in any patient with any product. Serum blood urea nitrogen (BUN) and/or creatinine should also be monitored regularly in patients receiving IgG therapy. Hemolysis and thromboembolic phenomena are also rare but important complications of IVIG and SCIG therapies. These require regular monitoring of hematologic parameters and vigilance for thrombosis.211,212
Immunization
The effectiveness of common vaccines has not been formally evaluated in patients with CVID. Routine boosters of tetanus or diphtheria toxoids or pneumococcus are probably not necessary for individuals receiving IgG replacement therapy because these antibodies are present in high amounts in therapeutic polyclonal IgG.213 Less commonly used inactivated vaccines do not pose any risk to patients, but efficacy is unknown. Because cellular immunity is thought to be largely intact in many patients with CVID, and inactivated vaccines may induce some cellular immunity, these may be administered according to the individual needs of the patient. In particular, some immunologists recommend routine administration of the inactivated influenza vaccine.214
Therapeutic IgG contains significant amounts of neutralizing antibodies to the most commonly used live viral vaccines such as measles, mumps, rubella, polio, and varicella.215 Thus, most of these vaccines are neutralized and rendered ineffective in patients receiving IgG. Note that some of these vaccines, especially live attenuated polio, may cause disease in patients with CVID if they have not yet been diagnosed or if IgG therapy has not been initiated; only inactivated or subunit vaccines should be given if CVID is suspected. Note also that the amount of antibody against these agents varies from product to product or from lot to lot and that caution regarding exposure is still advisable when patients live in or travel to areas where these diseases are still common.216 Efficacy and safety of less frequently used live-agent vaccines have not been studied in patients with CVID, and they are usually considered to be contraindicated. The risk of transmission of routine (and other) live-agent vaccines is usually low, and therapeutic IgG is neutralizing for measles, mumps, rubella (MMR) and varicella vaccines, so they may be used routinely in family members of patients with CVID. Vaccine polio transmission may occur from family contacts, and this does pose a risk to individuals with CVID because replacement IgG therapy may not be protective.217
Treatment of complications
Rhinosinusitis
Recurrent infections in the upper respiratory tract result in chronic sinusitis and nasal polyps. Patients with CVID in whom bacterial sinus infections persist after the commencement of adequate replacement immunoglobulin therapy should have sinus imaging and consultation with an otolaryngologist, removal of any polyps, and possible surgical restoration of sinus drainage. Note that a role for functional endoscopic sinus surgery in the management of any antibody deficiency, including CVID, has not been formally evaluated. Routine saline nasal rinses may be used to aid in mucus clearance. In addition, antibiotic prophylaxis may be required for periods of months, or longer.
Regarding antibiotic prophylaxis for chronic or recurrent rhinosinusitis and/or otitis media, no controlled trials have been conducted in antibody-deficient patients. There is no general consensus among practitioners whether intermittent, continuous, or rotating antibiotics provide improved therapy in subjects with CVID. However, many treating physicians with broad experience with immunodeficiency do use antibiotic prophylaxis for those patients with chronic sinus disease.218 Various agents and regimens may be considered. Suitable drugs include (but are not necessarily limited to) amoxicillin, trimethoprim sulfa, and macrolides. The latter may provide additional anti-inflammatory effects.219
Bronchiectasis
A baseline chest CT and lung function tests are essential to identify bronchiectasis or scarring, assess severity, and discover other pathologies (interstitial disease, granulomas).53,220 Bronchiectasis should be managed with appropriate measures including physiotherapy and sometimes prophylactic antibiotics, depending on the severity.220 Recent studies in patients with bronchiectasis (excluding cystic fibrosis) suggest that prophylactic use of azithromycin in patients with frequent exacerbations is useful on a trial basis at least.221,222 Sputum cultures before the institution of azithromycin prophylaxis may be taken to exclude nontuberculous mycobacteria and ascertain sensitivity to azithromycin. Patients may require monitoring for cardiac or hearing problems, and macrolides should be used with caution in patients at risk for prolonged Q-T interval. In extreme bronchiectasis, 7% hypertonic saline in conjunction with pulmonary hygiene has been found to be useful.223 Aminoglycoside antibiotics delivered via a nebulizer may also be considered (authors’ personal observations). Patients may benefit from joint management with a pulmonary specialist.
Unusual infections
Ureaplasma or Mycoplasma infections should be treated with a macrolide antibiotic to which the organism is sensitive.58 In the case of urogenital infections, it may often be useful to treat the patient’s partner as well. Mycoplasma in joints or lungs may require intravenous antibiotics.58,59 There are no consistently effective therapies for enteroviral infections causing meningoencephalitis. High-dose IVIG with measurable titer antibody to the infecting serotype may be helpful.62,224 The survival rate is only 50%. Fortunately, these infections do not appear to occur in patients maintaining relatively high (>800 mg/dL) IgG trough levels. Giardia infections are treated with high- dose tinidazole or metronidazole, with the addition of paromomycin in resistant cases.225
Autoimmune cytopenias
For autoimmune cytopenias, steroids in standard doses (1–2 mg/kg daily prednisone equivalent in divided doses) are usually sufficient. Higher doses of IgG (1–2 g/kg/month) can be used to supplement baseline therapy with oral steroids tapered over several weeks or more.226 Antirhesus antigen globulin may also be effective.227 Rituximab in standard doses has been successful for both refractory or recurrent ITP and/or AIHA.228 A repeat course of rituximab was needed in some patients.
Severe infections have occurred in some patients after splenectomy.52,229 However, this was not observed in all series.84 Splenectomy has been curative of ITP or AIHA in CVID and may be needed if other therapies fail. Recent reports indicate that splenectomy does not increase mortality in CVID if adequate immunoglobulin replacement is used.71,230
Organ-specific autoimmune disorders in patients with CVID are also treated as they would be in otherwise immunocompetent patients.
Granulomatous disease
Treatment of granulomas is empirical.78,231,232 Corticosteroids, in low doses for long periods to avoid steroid-related complications, have been found to result in short-term resolution in sites that are easily monitored by endoscopy or imaging. Steroid-sparing agents, such as azathioprine, cyclosporine, or other agents, have been tried with varying success. Infliximab has been successful for skin granulomas, but caution is advised in view of the increased risk of pyogenic or mycobacterial infections.233
Enteropathy
Biopsies of the intestinal mucosa should be performed in patients with CVID with chronic enteritis. Although the pathology is similar to that of celiac disease, there are histological differences.92 Unlike celiac disease, the enteropathy more often seen in CVID is not responsive to gluten withdrawal.75,234 IgG replacement alone does not generally ameliorate gastrointestinal involvement.235 Agents such as 5-aminosalicylic acid and/or nonabsorbed oral steroids such as budesonide are commonly used.91 Low-dose corticosteroids such as prednisone can be used in doses of 10 mg/day; however, higher doses can lead to a significant risk of opportunistic infections. Immunomodulators, such as azathioprine or 6-mercaptopurine, can be used safely because the doses used (as for Crohn disease) are low and do not appear to affect standard T- and B-cell function tests. Infliximab has also been used with some benefit in severe enteropathy, although its effectiveness may not be long-lasting.236 Bacterial overgrowth may be ameliorated by broad-spectrum antibiotics (metronidazole or tinidizole or ciprofloxacin). It is important to treat deficiency of fat-soluble vitamins even in those without enteropathy or malabsorption.93
Interstitial lung disease
This complication in CVID has a high mortality, so it is important to detect it as early as possible.53,67 All patients should have at least 1 high-resolution CT scan at diagnosis. Those without evidence of disease may be monitored at least annually with spirometry. The frequency of follow-up imaging in the absence of functional changes is unclear. Evidence of interstitial or other lung disease should be followed by full pulmonary function testing including diffusion capacity at least annually with imaging according to functional changes.
It is important to define the histopathology of lung disease so that the most potentially effective therapy can be used.53,67 There are no controlled studies that define optimal therapy, but a combination of azathioprine and rituximab was found to be effective in subjects with granulomatous/lymphocytic infiltrates in a small open-label study.237 In forms with a prominent T-cell infiltration, corticosteroids, with or without cyclosporine, have been reported to be successful.238 Anti-TNF antibody therapy has been successfully used in isolated cases of complex interstitial lung disease.233
Lymphoproliferation
More than 30% of the patients with CVID have an enlarged spleen, either palpable or on imaging, though symptoms of hypersplenism are often mild. There may be no need to treat these patients on the basis of splenomegaly alone. Likewise, persistent hypertrophy of lymph nodes should result in review of the diagnosis to exclude lymphoma or autoimmune lymphoproliferative syndrome but does not require treatment per se. Corticosteroids usually lead to regression of these phenomena, but they may recur when steroids are tapered. An acute increase in adenopathy or splenomegaly should prompt evaluation for possible malignant transformation.
Lymphoma
Clinicians must maintain a high index of suspicion for lymphoma in patients with CVID. Lymphoma may be difficult to distinguish from polyclonal lymphoid proliferation. Clonal analysis can be misleading because oligoclonal lymphocyte populations have been demonstrated in biopsies irrespective of histology.106 Treatment follows the current protocols for immunocompetent patients.
Stem cell therapy
Until now, bone marrow transplantation or other forms of hemopoietic stem cell therapy (HSCT) have not been considered to have an important role in the treatment of CVID, and experience has been limited. One study in severely ill patients with complex CVID showed that HSCT can be successful, but there was significant procedure-related mortality in those receiving HSCT equal to that of those remaining on the waiting list.239 More recently, a retrospective study of 25 patients with CVID receiving HSCT in 14 worldwide centers has been reported.240 The indication for HSCT was hematologic malignancy in 7 patients; 6 (83%) of these survived. The other 18 patients had 1 or more of the following conditions refractory to conventional therapies: autoimmune cytopenias, respiratory or gastrointestinal infections, interstitial/granulomatous lung disease, and/or autoimmune enteropathy. Only 33% of these patients survived. Of the total 12 surviving patients, half were able to discontinue IgG therapy and the indication for transplantation resolved in 11 (92%). With improved laboratory predictors, it may be possible in the near future to make better prognostic assessments and so transplant those in whom survival is likely to be considerably reduced.
Special considerations
Children
The main goal of therapeutic management in children with CVID is to decrease the morbidity and mortality associated with recurrent infections. The consensus among pediatric immunologists is that close clinical monitoring and appropriate IgG replacement can ultimately extend the life expectancy of these young patients to approach that of the general population. The optimal dose and interval of replacement immunoglobulin needed to achieve this aim is still under investigation.
In a multicenter randomized, double-blind cross-over study of 18 children (and 25 adults) with antibody deficiencies, the patients who received 800 mg/kg IVIG every 4 weeks had significantly reduced number and duration of infections versus those who received 400 mg/kg every 4 weeks.241 These findings suggest that in children, as in adults, higher doses of IgG replacement therapy result in fewer infections. Similar conclusions were reached in 2 meta-analyses of infection incidence in relation to IVIG and SCIG dosing.189,190
Although the general concerns regarding IgG therapy affect adults and children equally, reduced blood volumes and immature renal function places neonates and infants at risk of developing electrolytic imbalances or volume overload. For these patients, selection of IVIG products with a higher protein concentration, low osmolality, and neutral pH constitute the best option.242 In children, periodic dose adjustments are required during periods of accelerated growth.
Complications in children may be more common than in adult patients with CVID.243,244 Furthermore, recurrent infections, chronic inflammation, chronic nutritional deficits, and associated gastrointestinal abnormalities may pose threats to optimal growth in children. Bronchial hyperreactivity associated with allergen sensitization may be seen in some children with CVID.100 Children with asthma who have other features suggestive of CVID should be screened for antibody deficiency. Although CVID is not common in the general population with asthma, other milder forms of antibody deficiency may also require IgG therapy. As in adults, it is also important to distinguish early structural damage and overt bronchiectasis from interstitial lung disease.245 Many children already have bronchiectasis by the time they are diagnosed with CVID. In a series of 22 children, bronchiectasis was present in 7 cases (age range, 2.5–15 years) and detected before CVID diagnosis in 5 children.246 Similar findings were observed in another pediatric cohort.244
To prevent organ damage, the importance of screening of family members of children with CVID is emphasized in a recent report demonstrating that a considerable proportion displayed either a positive clinical history with symptoms suggestive of primary immunodeficiency or alterations in humoral immunity including CVID.247
Pregnancy
Plasma dilution in the third trimester of pregnancy results in a modest reduction in serum IgG trough levels. At the same time, the fetus is receiving maternal IgG via transport across the placenta. It is advisable to increase the replacement IgG dose during this time and for delivery. Authorities differ in their recommendations for dose increase, ranging anywhere from 10% to 50%248–250 Female patients can be assured that SCIG can be delivered into the abdominal wall safely during pregnancy. Depending on body mass index, alternative sites may be chosen for comfort.
Travel
Although many Web sites relating to travel medicine propose specific recommendations regarding immunocompromised travelers, these do not necessarily apply to those with primary antibody failure patients receiving adequate IgG replacement therapy. Patients should discuss their plans for travel with their specialist physician or pediatrician well in advance. Avoidance of infectious agents to which patients with CVID are particularly susceptible, such as water-borne Giardia, Campylobacter, and Salmonella, should be avoided by using only clean water and eschewing uncooked food in countries where these pathogens are common. Caution should be advised regarding travel to areas where certain infections (eg, meningococcal meningitis) are endemic and protection by IgG may not be adequate.
Support
The aim of management is to enable patients to live in a near-normal fashion as much as possible, and to take charge of the disease and the treatment. Lifestyle considerations (sleep, exercise, smoking, drug use, etc) must be emphasized regularly. Children should be encouraged to play outdoors with other children, attend school, and participate in sports and other extracurricular activities, especially those that promote aerobic exercise. There may be circumstances in which the frequency of infections or exposure to other irritant agents may render day care (especially in larger group settings) and school attendance problematic for individual patients. Similar considerations apply to certain work occupations and environments for adult patients. Decisions regarding such attendance or work must be made on a case-by-case basis with the participation of all involved. The goal should always be to promote as much social integration as possible.
Participation in support groups (Table VI) provides essential information relating to work, schooling, insurance, and access to medical and expert care. Other patients and families offer perspectives and mutual support not obtainable from medical personnel. Such groups can help with the social, economic, emotional, and psychological issues of chronic disease.
TABLE VI.
Organization | Web site address (URL) |
---|---|
The Jeffrey Modell Foundation (JMF) | jmfworld.org |
The Immune Deficiency Foundation (IDF) | primaryimmune.org |
International Patient Organization for Primary Immunodeficiencies (IPOPI) |
ipopi.org |
IPOPI provides support for the national patient organizations in each of 50 countries. It will provide contact information for any of the member country organizations upon request. |
PROGNOSIS
In the first UK Medical Research Council Report in 1979, survival of patients with CVID 12 years after diagnosis was only 30%.251 By the end of the 1990s, with the advent of high-dose IVIG therapy, survival 20 years after diagnosis was 64% for males and 67% for females, as compared with the expected 92% and 94% population survival, respectively.52 Since the year 2000, as the standard replacement dose of IgG has steadily increased, more and more patients have had more normal IgG levels for the majority of their lives, leading to an expected survival overall of 58% 45 years after diagnosis.53,71 In a large Italian cohort, overall survival was 35% at 40 years. However, while no patients with cancer survived beyond 35 years after diagnosis, patients without malignancy had overall survival of 65% at 40 years.70 Individual prognosis depends on the clinical phenotype of the patient. Those without disease-related complications have an almost normal life expectancy.12 Patients with unexplained enteropathy, chronic lung disease, polyclonal lymphoproliferation, or cytopenia have reduced survival (see below).53
Data obtained in 2 large surveys over a period of 4 decades showed that causes of death included respiratory failure from chronic lung disease accounting for about 35%, lymphomas accounting for 18%, and cancers other than lymphomas 10% to 33%.70,71 The importance of cancers other than lymphomas as CVID-associated diseases was further underlined by observations that intestinal adenocarcinomas were the first clinical presentation in some and that in about 3% of the patients with CVID more than 1 distinct primary malignancy occurred.
IgG replacement is the standard of care in CVID and significantly reduces infections (see below). However, this therapy does not seem to prevent or treat the poorly understood noninfectious complications that have emerged as the most difficult aspects of patient management.53,186 Subjects with CVID who have had infections as their only manifestation of immune deficiency do well over time, with survival resembling that of age-matched controls. Long-term follow-up reveals that subjects with CVID with inflammatory complications have increased morbidity and mortality despite IgG replacement therapy.52,53,71,252
There are differences among large series reported as to the prognostic value of serum immunoglobulins at presentation. In a cohort of 473 patients followed for 4 decades at Mount Sinai Medical Center (New York, NY), lower baseline levels of serum IgG and higher IgM levels were associated with poor survival.71 In a retrospective study from Europe, serum immunoglobulin levels did not predict survival.53 In both cohorts, elevated levels of serum IgM at diagnosis correlated with the later development of lymphoproliferation or lymphoid malignancy.
In the European cohort, low number of B cells at presentation did not predict survival.53 This is in contrast to the Mount Sinai study in which lower numbers of peripheral B cells continued to be associated with reduced survival, as previously reported.52,71
AREAS FOR FURTHER STUDY
Definition
It is evident from this work and other publications16 that full consensus regarding the definition of CVID does not yet exist. Some groups include clinical criteria in the definition, however, as for other primary immune defects, and considering the diverse manifestations of this disease, we conclude that the diagnosis of CVID should be primarily based on laboratory criteria. It is possible that advances in the areas of biomarkers and genetics (see below) will provide new material for or a new approach to the development of such a consensus.
Biomarkers
In CVID, the immunological and clinical picture might change gradually over time, even in patients receiving treatment. Similarly, the clinical and laboratory presentation at diagnosis might also depend on the time elapsed since the onset of symptoms because additional complications may arise and immunologic alterations may progress after the initial manifestations of the disease.8 In particular, B-cell function might gradually decrease or improve over time. This is most clearly seen in most of the patients with transient hypogammaglobulinemia of infancy who normalize IgG values, mostly within 2 to 4 years of age.253 A minority of these children develop a form of persistent undefined hypogammaglobulinemia that may not meet diagnostic criteria for CVID. As noted above, other forms of antibody deficiency may progress in severity, as illustrated by patients with selective IgA deficiency who ultimately develop CVID over time.113 Thus, immunological abnormalities, including immunoglobulin levels and antibody responses, should be periodically assessed. The evaluation of the latter criterion in most patients will be performed before IgG replacement therapy and will need to account for the altered vaccination schedule in many countries, including conjugated vaccination against S pneumoniae during childhood, which can complicate the evaluation of the polysaccharide response.
The possibility of measuring IgM and IgA antibody responses, instead of specific IgG, would overcome the difficulty of studying vaccine responses in patients who are already on IgG therapy. Antipneumococcal polysaccharide IgM and IgA assays might be useful in the distinction between responders and nonresponders. Such assays could also help stratify patients with CVID with respect to residual B-cell capacity to mount an antibody response and the risk of developing invasive infections. Thus, they might serve as prognostic markers in patients with CVID. Study of mucosal immunity at the tissue level may improve understanding of the immunological abnormalities in patients with CVID. These data might provide additional prognostic markers.
Lymphocyte phenotyping via flow cytometry and other methods will likely continue to reveal finer detail in peripheral blood populations that may have relevance for disease severity, clinical complications, and progression.
TRECs and KRECs as clinical markers for CVID
TRECs and signal joint immunoglobulin KRECs are nuclear episomal DNA by-products of V(D)J recombination during T-cell and B-cell neogenesis. Impairment of T- or B-lymphocyte development leads to a reduction in the proportion of TREC- or KREC-containing T or B cells in the periphery. Real-time PCR-based quantification of TRECs and KRECs is being applied for newborn screening of severe combined immunodeficiency254 and B-lymphocyte deficiency (agammaglobulinemia).255 In addition to the newborn screening, it has been reported that TRECs and KRECs can be used as clinical markers for the severity and progression of various primary immunodeficiency diseases.46
As discussed above, although CVID is still considered to be a disease with predominant B-cell dysfunction, many individuals also exhibit important abnormalities of T-cell development and function and suffer from complications that may be related to T-cell deficiency, including opportunistic infections, autoimmune diseases, and malignancies.5,53 It can be challenging to draw the line between T-cell dysfunction in CVID and combined immunodeficiency (CID) “misdiagnosed” as CVID.5 Therefore, identifying novel markers to better classify CVID and distinguish CID from CVID is desirable (also see the discussion of LOCID above).
Measurement of TRECs and KRECs may provide useful clinical markers to distinguish CID from CVID. In a report, 37 patients with CVID all receiving IgG therapy were analyzed for TRECs and KRECs.46 Many patients in this study had opportunistic infections, suggesting that CID may often be misdiagnosed as CVID.
On the basis of TREC and KREC copy numbers, patients were classified into 4 groups: A: TREC(+) KREC(+), B: TREC(+) KREC(−), C: TREC(−) KREC(+), and D: TREC(−) KREC(−). Correlation was found with the TREC/KREC-based classification and clinical symptoms in each patient group. The cumulative events of complications (opportunistic infections, autoimmune diseases, and malignancies) per 10 patient-years were highest in group D, followed by group C, group B, and group A. Events in groups D and C were significantly higher than those in group A. TREC/KREC-based classification also correlated with prognosis. One patient in group D died of Pneumocystis jirovecii pneumonia, and 2 other patients in the same group received HSCT following complications due to EBV-related lymphoproliferative disorder. In contrast to group D patients, group A patients remained relatively healthy. One possible explanation is that these patients harbor defects only in terminal B-cell differentiation, but not in T cells. In group B patients, autoimmune diseases were often observed, suggesting that the balance between T and B cells is important in the development of autoimmune diseases in CVID.5 Group C and D patients suffered a high frequency of both opportunistic infections and malignancies, suggesting that these TREC(−) patients have functionally important T-cell deficits. HSCT may be considered as a treatment to “cure” group D patients, as reported in severe CVID/CID cases, because event-free survival is poor.239
Genetics
Genetic variation may be further studied in relation to clinical symptoms (ie, infection-predominant, inflammation-predominant, autoimmune-predominant), in relation to disease severity (ie, disease course, treatment efficacy), and in relation to immunological parameters (ie, immunophenotyping and/or function in vitro/in vivo).
Complete genome or exome sequencing may be highly informative for elucidating more common disease-causing genetic alterations as well as the assessment of relevant disease-modifying loci or genes. Further large cohort studies using genomic sequencing and other high-throughput methods (assessment of copy number variations and SNPs) applied in international studies with subjects from diverse genetic backgrounds will be needed in the future to illuminate the (possibly) many causes of this disease and possible therapeutic targets.
Management
What is the best practice for following subjects with CVID over time? Because of the heterogeneity of this disease, there are no set rules aside from regularly scheduled follow-ups and periodic monitoring of serum IgG levels. Full chemistry panels and complete blood cell counts are also important to check for problems that can arise over time (see above). Best practice for monitoring subjects for lung disease has been controversial, and there is no current consensus. The significance of possible radiosensitivity needs to be further studied in CVID.256 Magnetic resonance imaging may represent a reliable radiation-free technique for diagnosis and follow-up.257 Further attention must also be paid to early diagnosis of lymphoid and nonlymphoid cancers. Additional prospective data from large cohorts are needed to better define the most efficient and cost-effective strategies.
How it is possible to individualize immunoglobulin replacement in terms of intervals and doses? Health care delivery systems are quickly changing in response to economic pressures and concerns about quality of care. The system of care is itself an important determinant of patient outcomes. Elucidating the effects of the system of care on patient outcomes requires new methodologic approaches to identify what works in which setting and under what conditions. Personalized health research presents further methodologic challenges because emphasis is placed on the individual response rather than on the population.258
The Oxford group proposed to increase the immunoglobulin dose by 0.15 g/kg/month when patients present with a serious infection, persistent infection, or 3 or more moderate infections over a year.188 Other factors contributing to infections may need to be assessed before these increased doses are made permanent for a specific patient. Other options include shortening the interval between IgG doses with or without increase in the cumulative IgG monthly dose.
It may be that replacement therapy in primary immunodeficiencies is not a mere passive transfer of antibodies exclusively to prevent recurrent infections but also has an active role in regulating autoimmune and inflammatory responses through modulating B-cell or other-cell functions.259 Consequently, we need a better understanding of the biological effects of IgG replacement even if administered at lower doses than usually used for inflammatory disease.
From existing data, it is not possible at the moment to provide clear assessment of the outcome of HSCT as a potentially curative approach in patients with CVID with poor prognosis. Knowing that the supportive care and efficacy of newer antimicrobial drugs will continue to improve clinical outcomes, it has remained difficult to advise patients with CVID to undergo HSCT. Elaboration of prognostic scores and better diagnostic tools (eg, for stromal cell defects) will be of utmost importance for early selection of suitable patients to better inform choices in the future.239
Acknowledgments
We respectfully and fondly dedicate this work to the memory of our colleague, coauthor, and friend Dr Isil Barlan.
F. A. Bonilla is on the Immune Deficiency Foundation Board; has received consultancy fees from Baxter, The Cowan Group Inc, CSL Behring, Gerson-Lehrman Group Inc, Grand Rounds Health Inc, and the Immune Deficiency Foundation; has received research support from CSL Behring; has received lecture fees from Albany Medical College, Finger Lakes Allergy Society, and Eastern Allergy Conference; received royalties from UpToDate; has received travel support from American Public Health Laboratories; and has received other funds from Octapharma Inc. H. Chapel is on the Baxter board and has received consultancy fees from Biotest and LFB. M. T. de la Morena has received consultancy fees from the Atlantic Research Group. F. J. Espinosa-Rosalesis on the Baxter board; has received lecture fees from Grifols and CSL Behring; and has received travel support from Grifols, CSL Behring, and Octapharma. I. Quinti is on the Baxter board; has received consultancy fees from FKB; has received research support from the Plasma Protein Therapeutics Association; has received lecture fees from Baxter and Kedrion; and has received travel support from Baxter. J. M. Routes has received research support from the National Institutes of Health/National Institute of Allergy and Infectious Diseases (granulomatous lymphocytic interstitial lung disease in common variable immunodeficiency grant—R34 planning grant). M. L. K. Tang is on the Nestle Nutrition Institute Medical Advisory Board Oceania; is on the Danone Nutritia Medical Advisory Board Australia New Zealand and Global Scientific Advisory Board; has received consultancy fees from GLG Consultants and Deerfield Institute; has received lecture fees from Danone Nutricia and Health Ed; has a patent on a method for inducing tolerance; received royalties from Wiley as a coauthor of Kids Food Allergies for Dummies; and has received payment for developing an educational module for MD Linx. K. Warnatz has received lecture fees from Baxter, GlaxoSmithKline, CSL Behring, Pfizer, the American Academy of Allergy, Asthma & Immunology, Biotest, Novartis, Stallergenes, Roche, Meridian Health Comms, and Octapharma; has received payment for manuscript preparation from UCB Pharma; and has received payment for developing educational presentations from the European Society for Immunodeficiency.
Abbreviations used
- AIE
autoimmune enteropathy
- AIHA
autoimmune hemolytic anemia
- CID
combined immunodeficiency
- CT
computed tomography
- CVID
common variable immunodeficiency disorders
- HSCT
hemopoietic stem cell therapy
- ITP
immune thrombocytopenic purpura
- IVIG
intravenous immunoglobulin
- LOCID
late-onset combined immune deficiency
- SCIG
subcutaneous immunoglobulin
- SNP
single nucleotide polymorphism
- TREC
T-cell receptor excision circle
Footnotes
This article is a product of an international coalition between the American Academy of Allergy, Asthma & Immunology; the European Academy of Allergy and Clinical Immunology; the World Allergy Organization; and the American College of Allergy, Asthma & Immunology on common variable immunodeficiency.
Conflicts of interest: The rest of the authors declare that they have no relevant conflicts of interest.
REFERENCES
- 1.Fudenberg H, Good RA, Goodman HC, Hitzig W, Kunkel HG, Roitt IM, et al. Primary immunodeficiencies: report of a World Health Organization Committee. Pediatrics. 1971;47:927–946. [PubMed] [Google Scholar]
- 2.Cooper MD, Faulk WP, Fudenberg HH, Good RA, Hitzig W, Kunkel H, et al. Classification of primary immunodeficiencies. N Engl J Med. 1973;288:966–967. doi: 10.1056/NEJM197305032881814. [DOI] [PubMed] [Google Scholar]
- 3.Notarangelo LD, Fischer A, Geha RS, Casanova JL, Chapel H, Conley ME, et al. Primary immunodeficiencies: 2009 update. J Allergy Clin Immunol. 2009;124:1161–1178. doi: 10.1016/j.jaci.2009.10.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Orange JS, Glessner JT, Resnick E, Sullivan KE, Lucas M, Ferry B, et al. Genome-wide association identifies diverse causes of common variable immunodeficiency. J Allergy Clin Immunol. 2011;127:1360.e6–1367.e6. doi: 10.1016/j.jaci.2011.02.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Yong PF, Thaventhiran JE, Grimbacher B. “A rose is a rose is a rose,” but CVID is not CVID common variable immune deficiency (CVID), what do we know in 2011? Adv Immunol. 2011;111:47–107. doi: 10.1016/B978-0-12-385991-4.00002-7. [DOI] [PubMed] [Google Scholar]
- 6.Keller MD, Jyonouchi S. Chipping away at a mountain: genomic studies in common variable immunodeficiency. Autoimmun Rev. 2013;12:687–689. doi: 10.1016/j.autrev.2012.10.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kutukculer N, Karaca NE, Demircioglu O, Aksu G. Increases in serum immunoglobulins to age-related normal levels in children with IgA and/or IgG subclass deficiency. Pediatr Allergy Immunol. 2007;18:167–173. doi: 10.1111/j.1399-3038.2006.00491.x. [DOI] [PubMed] [Google Scholar]
- 8.Chapel H, Lucas M, Patel S, Lee M, Cunningham-Rundles C, Resnick E, et al. Confirmation and improvement of criteria for clinical phenotyping in common variable immunodeficiency disorders in replicate cohorts. J Allergy Clin Immunol. 2012;130:1197.e9–1198.e9. doi: 10.1016/j.jaci.2012.05.046. [DOI] [PubMed] [Google Scholar]
- 9.Malphettes M, Gerard L, Carmagnat M, Mouillot G, Vince N, Boutboul D, et al. Late-onset combined immune deficiency: a subset of common variable immunodeficiency with severe T cell defect. Clin Infect Dis. 2009;49:1329–1338. doi: 10.1086/606059. [DOI] [PubMed] [Google Scholar]
- 10.Agarwal S, Cunningham-Rundles C. Autoimmunity in common variable immunodeficiency. Curr Allergy Asthma Rep. 2009;9:347–352. doi: 10.1007/s11882-009-0051-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Quinti I, Soresina A, Spadaro G, Martino S, Donnanno S, Agostini C, et al. Long-term follow-up and outcome of a large cohort of patients with common variable immunodeficiency. J Clin Immunol. 2007;27:308–316. doi: 10.1007/s10875-007-9075-1. [DOI] [PubMed] [Google Scholar]
- 12.Chapel H, Cunningham-Rundles C. Update in understanding common variable immunodeficiency disorders (CVIDs) and the management of patients with these conditions. Br J Haematol. 2009;145:709–727. doi: 10.1111/j.1365-2141.2009.07669.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Cunningham-Rundles C. How I treat common variable immune deficiency. Blood. 2010;116:7–15. doi: 10.1182/blood-2010-01-254417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kardar G, Oraei M, Shahsavani M, Namdar Z, Kazemisefat G, Haghi Ashtiani M, et al. Reference intervals for serum immunoglobulins IgG, IgA, IgM and complements C3 and C4 in Iranian healthy children. Iran J Public Health. 2012;41:59–63. [PMC free article] [PubMed] [Google Scholar]
- 15.Ozen A, Baris S, Karakoc-Aydiner E, Ozdemir C, Bahceciler NN, Barlan IB. Outcome of hypogammaglobulinemia in children: immunoglobulin levels as predictors. Clin Immunol. 2010;137:374–383. doi: 10.1016/j.clim.2010.08.010. [DOI] [PubMed] [Google Scholar]
- 16.Ameratunga R, Woon ST, Gillis D, Koopmans W, Steele R. New diagnostic criteria for CVID. Expert Rev Clin Immunol. 2014;10:183–186. doi: 10.1586/1744666X.2014.875274. [DOI] [PubMed] [Google Scholar]
- 17.Aghamohammadi A, Moein M, Farhoudi A, Pourpak Z, Rezaei N, Abolmaali K, et al. Primary immunodeficiency in Iran: first report of the National Registry of PID in Children and Adults. J Clin Immunol. 2002;22:375–380. doi: 10.1023/a:1020660416865. [DOI] [PubMed] [Google Scholar]
- 18.Gathmann B, Binder N, Ehl S, Pourpak Z, Rezaei N, Abolmaali K. The European internet-based patient and research database for primary immunodeficiencies: update 2011. Clin Exp Immunol. 2012;167:479–491. doi: 10.1111/j.1365-2249.2011.04542.x. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 19.Golan H, Dalal I, Garty BZ, Schlesinger M, Levy J, Handzel Z, et al. The incidence of primary immunodeficiency syndromes in Israel. Isr Med Assoc J. 2002;4:868–871. [PubMed] [Google Scholar]
- 20.Leiva LE, Zelazco M, Oleastro M, Carneiro-Sampaio M, Condino-Neto A, Costa-Carvalho BT, et al. Primary immunodeficiency diseases in Latin America: the second report of the LAGID registry. J Clin Immunol. 2007;27:101–108. doi: 10.1007/s10875-006-9052-0. [DOI] [PubMed] [Google Scholar]
- 21.Lim DL, Thong BY, Ho SY, Shek LP, Lou J, Leong KP, et al. Primary immunodeficiency diseases in Singapore–the last 11 years. Singapore Med J. 2003;44:579–586. [PubMed] [Google Scholar]
- 22.Boyle JM, Buckley RH. Population prevalence of diagnosed primary immunodeficiency diseases in the United States. J Clin Immunol. 2007;27:497–502. doi: 10.1007/s10875-007-9103-1. [DOI] [PubMed] [Google Scholar]
- 23.Rachid R, Bonilla FA. The role of anti-IgA antibodies in causing adverse reactions to gamma globulin infusion in immunodeficient patients: a comprehensive review of the literature. J Allergy Clin Immunol. 2012;129:628–634. doi: 10.1016/j.jaci.2011.06.047. [DOI] [PubMed] [Google Scholar]
- 24.Javier FC, III, Moore CM, Sorensen RU. Distribution of primary immunodeficiency diseases diagnosed in a pediatric tertiary hospital. Ann Allergy Asthma Immunol. 2000;84:25–30. doi: 10.1016/S1081-1206(10)62736-6. [DOI] [PubMed] [Google Scholar]
- 25.Immunodeficiencies ESf. New ESID Registry 2013. [Accessed November 15, 2014]; Available from: http://esid.org/Working-Parties/Registry/New-ESID-Registry. [Google Scholar]
- 26.Network USI. USIDNET Registry. Towson, MD: Immune Deficiency Foundation; 2014. [Accessed September 22, 2014]. Available from: http://www.usidnet.org/pub/Registries-Info. [Google Scholar]
- 27.Ishimura M, Takada H, Doi T, Imai K, Sasahara Y, Kanegane H, et al. Nationwide survey of patients with primary immunodeficiency diseases in Japan. J Clin Immunol. 2011;31:968–976. doi: 10.1007/s10875-011-9594-7. [DOI] [PubMed] [Google Scholar]
- 28.Warnatz K, Denz A, Drager R, Braun M, Groth C, Wolff-Vorbeck G, et al. Severe deficiency of switched memory B cells (CD27(+)IgM(−)IgD(−)) in subgroups of patients with common variable immunodeficiency: a new approach to classify a heterogeneous disease. Blood. 2002;99:1544–1551. doi: 10.1182/blood.v99.5.1544. [DOI] [PubMed] [Google Scholar]
- 29.Wehr C, Kivioja T, Schmitt C, Ferry B, Witte T, Eren E, et al. The EUROclass trial: defining subgroups in common variable immunodeficiency. Blood. 2008;111:77–85. doi: 10.1182/blood-2007-06-091744. [DOI] [PubMed] [Google Scholar]
- 30.Ochtrop ML, Goldacker S, May AM, Rizzi M, Draeger R, Hauschke D, et al. T and B lymphocyte abnormalities in bone marrow biopsies of common variable immunodeficiency. Blood. 2011;118:309–318. doi: 10.1182/blood-2010-11-321695. [DOI] [PubMed] [Google Scholar]
- 31.Taubenheim N, von Hornung M, Durandy A, Warnatz K, Corcoran L, Peter HH, et al. Defined blocks in terminal plasma cell differentiation of common variable immunodeficiency patients. J Immunol. 2005;175:5498–5503. doi: 10.4049/jimmunol.175.8.5498. [DOI] [PubMed] [Google Scholar]
- 32.Bryant A, Calver NC, Toubi E, Webster AD, Farrant J. Classification of patients with common variable immunodeficiency by B cell secretion of IgM and IgG in response to anti-IgM and interleukin-2. Clin Immunol Immunopathol. 1990;56:239–248. doi: 10.1016/0090-1229(90)90145-g. [DOI] [PubMed] [Google Scholar]
- 33.Driessen GJ, van Zelm MC, van Hagen PM, Hartwig NG, Trip M, Warris A, et al. B-cell replication history and somatic hypermutation status identify distinct pathophysiologic backgrounds in common variable immunodeficiency. Blood. 2011;118:6814–6823. doi: 10.1182/blood-2011-06-361881. [DOI] [PubMed] [Google Scholar]
- 34.Foerster C, Voelxen N, Rakhmanov M, Keller B, Gutenberger S, Goldacker S, et al. B cell receptor-mediated calcium signaling is impaired in B lymphocytes of type Ia patients with common variable immunodeficiency. J Immunol. 2010;184:7305–7313. doi: 10.4049/jimmunol.1000434. [DOI] [PubMed] [Google Scholar]
- 35.Marron TU, Yu JE, Cunningham-Rundles C. Toll-like receptor function in primary B cell defects. Front Biosci (Elite Ed) 2012;4:1853–1863. doi: 10.2741/507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Giovannetti A, Pierdominici M, Aiuti F. T-cell homeostasis: the dark(ened) side of common variable immunodeficiency. Blood. 2008;112:446. doi: 10.1182/blood-2008-03-145045. author reply 446–7. [DOI] [PubMed] [Google Scholar]
- 37.Funauchi M, Farrant J, Moreno C, Webster AD. Defects in antigen-driven lymphocyte responses in common variable immunodeficiency (CVID) are due to a reduction in the number of antigen-specific CD4+ T cells. Clin Exp Immunol. 1995;101:82–88. doi: 10.1111/j.1365-2249.1995.tb02281.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Stagg AJ, Funauchi M, Knight SC, et al. Failure in antigen responses by T cells from patients with common variable immunodeficiency (CVID) Clin Exp Immunol. 1994;96:48–53. doi: 10.1111/j.1365-2249.1994.tb06228.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Thon V, Eggenbauer H, Wolf HM, Fischer MB, Litzman J, Lokaj J, et al. Antigen presentation by common variable immunodeficiency (CVID) B cells and monocytes is unimpaired. Clin Exp Immunol. 1997;108:1–8. doi: 10.1046/j.1365-2249.1997.d01-989.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Fischer MB, Hauber I, Vogel E, Wolf HM, Mannhalter JW, Eibl MM. Defective interleukin-2 and interferon-gamma gene expression in response to antigen in a subgroup of patients with common variable immunodeficiency. J Allergy Clin Immunol. 1993;92:340–352. doi: 10.1016/0091-6749(93)90178-i. [DOI] [PubMed] [Google Scholar]
- 41.Holm AM, Aukrust P, Aandahl EM, Muller F, Tasken K, Froland SS. Impaired secretion of IL-10 by T cells from patients with common variable immunodeficiency–involvement of protein kinase A type I. J Immunol. 2003;170:5772–5777. doi: 10.4049/jimmunol.170.11.5772. [DOI] [PubMed] [Google Scholar]
- 42.Adelman DC, Matsuda T, Hirano T, Kishimoto T, Saxon A. Elevated serum interleukin-6 associated with a failure in B cell differentiation in common variable immunodeficiency. J Allergy Clin Immunol. 1990;86:512–521. doi: 10.1016/s0091-6749(05)80207-6. [DOI] [PubMed] [Google Scholar]
- 43.Martinez-Pomar N, Raga S, Ferrer J, Pons J, Munoz-Saa I, Julia MR, et al. Elevated serum interleukin (IL)-12p40 levels in common variable immunodeficiency disease and decreased peripheral blood dendritic cells: analysis of IL-12p40 and interferon-gamma gene. Clin Exp Immunol. 2006;144:233–238. doi: 10.1111/j.1365-2249.2006.03063.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bateman EA, Ayers L, Sadler R, Lucas M, Roberts C, Woods A, et al. T cell phenotypes in patients with common variable immunodeficiency disorders: associations with clinical phenotypes in comparison with other groups with recurrent infections. Clin Exp Immunol. 2012;170:202–211. doi: 10.1111/j.1365-2249.2012.04643.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Guazzi V, Aiuti F, Mezzaroma I, Mazzetta F, Andolfi G, Mortellaro A, et al. Assessment of thymic output in common variable immunodeficiency patients by evaluation of T cell receptor excision circles. Clin Exp Immunol. 2002;129:346–353. doi: 10.1046/j.1365-2249.2002.01893.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kamae C, Nakagawa N, Sato H, Honma K, Mitsuiki N, Ohara O, et al. Common variable immunodeficiency classification by quantifying T-cell receptor and immunoglobulin kappa-deleting recombination excision circles. J Allergy Clin Immunol. 2013;131:1437.e5–1440.e5. doi: 10.1016/j.jaci.2012.10.059. [DOI] [PubMed] [Google Scholar]
- 47.Scott-Taylor TH, Green MR, Raeiszadeh M, Workman S, Webster AD. Defective maturation of dendritic cells in common variable immunodeficiency. Clin Exp Immunol. 2006;145:420–427. doi: 10.1111/j.1365-2249.2006.03152.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Yu JE, Knight AK, Radigan L, Marron TU, Zhang L, Sanchez-Ramon S, et al. Toll-like receptor 7 and 9 defects in common variable immunodeficiency. J Allergy Clin Immunol. 2009;124:349–356. 356.e1–356.e3. doi: 10.1016/j.jaci.2009.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Aspalter RM, Sewell WA, Dolman K, Gerard L, Oksenhendler E, Warnatz K, et al. Deficiency in circulating natural killer (NK) cell subsets in common variable immunodeficiency and X-linked agammaglobulinaemia. Clin Exp Immunol. 2000;121:506–514. doi: 10.1046/j.1365-2249.2000.01317.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gathmann B, Mahlaoui N, Ceredih, Gerard L, Oksenhendler E, Warnatz K, et al. Clinical picture and treatment of 2212 patients with common variable immunodeficiency. J Allergy Clin Immunol. 2014;134:116–126. doi: 10.1016/j.jaci.2013.12.1077. [DOI] [PubMed] [Google Scholar]
- 51.Finck A, Van der Meer JW, Schaffer AA, Pfannstiel J, Fieschi C, Plebani A, et al. Linkage of autosomal-dominant common variable immunodeficiency to chromosome 4q. Eur J Hum Genet. 2006;14:867–875. doi: 10.1038/sj.ejhg.5201634. [DOI] [PubMed] [Google Scholar]
- 52.Cunningham-Rundles C, Bodian C. Common variable immunodeficiency: clinical and immunological features of 248 patients. Clin Immunol. 1999;92:34–48. doi: 10.1006/clim.1999.4725. [DOI] [PubMed] [Google Scholar]
- 53.Chapel H, Lucas M, Lee M, Bjorkander J, Webster D, Grimbacher B, et al. Common variable immunodeficiency disorders: division into distinct clinical phenotypes. Blood. 2008;112:277–286. doi: 10.1182/blood-2007-11-124545. [DOI] [PubMed] [Google Scholar]
- 54.Sanford JP, Favour CB, Tribeman MS. Absence of serum gamma globulins in an adult. N Engl J Med. 1954;250:1027–1029. doi: 10.1056/NEJM195406172502403. [DOI] [PubMed] [Google Scholar]
- 55.Boileau J, Mouillot G, Gerard L, Carmagnat M, Rabian C, Oksenhendler E, et al. Autoimmunity in common variable immunodeficiency: correlation with lymphocyte phenotype in the French DEFI study. J Autoimmun. 2011;36:25–32. doi: 10.1016/j.jaut.2010.10.002. [DOI] [PubMed] [Google Scholar]
- 56.Oksenhendler E, Gerard L, Fieschi C, Malphettes M, Mouillot G, Jaussaud R, et al. Infections in 252 patients with common variable immunodeficiency. Clin Infect Dis. 2008;46:1547–1554. doi: 10.1086/587669. [DOI] [PubMed] [Google Scholar]
- 57.Resnick ES, Cunningham-Rundles C. The many faces of the clinical picture of common variable immune deficiency. Curr Opin Allergy Clin Immunol. 2012;12:595–601. doi: 10.1097/ACI.0b013e32835914b9. [DOI] [PubMed] [Google Scholar]
- 58.Webster AD, Taylor-Robinson D, Furr PM, Asherson GL. Chronic cystitis and urethritis associated with ureaplasmal and mycoplasmal infection in primary hypogammaglobulinaemia. Br J Urol. 1982;54:287–291. doi: 10.1111/j.1464-410x.1982.tb06977.x. [DOI] [PubMed] [Google Scholar]
- 59.Franz A, Webster AD, Furr PM, Taylor-Robinson D. Mycoplasmal arthritis in patients with primary immunoglobulin deficiency: clinical features and outcome in 18 patients. Br J Rheumatol. 1997;36:661–668. doi: 10.1093/rheumatology/36.6.661. [DOI] [PubMed] [Google Scholar]
- 60.Pitcher DG, Windsor D, Windsor H, Bradbury JM, Yavari C, Jensen JS, et al. Mycoplasma amphoriforme sp. nov., isolated from a patient with chronic bronchopneumonia. Int J Syst Evol Microbiol. 2005;55:2589–2594. doi: 10.1099/ijs.0.63269-0. [DOI] [PubMed] [Google Scholar]
- 61.Gelfand EW. Unique susceptibility of patients with antibody deficiency to mycoplasma infection. Clin Infect Dis. 1993;17:S250–S253. [PubMed] [Google Scholar]
- 62.Halliday E, Winkelstein J, Webster AD. Enteroviral infections in primary immunodeficiency (PID): a survey of morbidity and mortality. J Infect. 2003;46:1–8. doi: 10.1053/jinf.2002.1066. [DOI] [PubMed] [Google Scholar]
- 63.Busse PJ, Razvi S, Cunningham-Rundles C. Efficacy of intravenous immunoglobulin in the prevention of pneumonia in patients with common variable immunodeficiency. J Allergy Clin Immunol. 2002;109:1001–1004. doi: 10.1067/mai.2002.124999. [DOI] [PubMed] [Google Scholar]
- 64.Kainulainen L, Nikoskelainen J, Vuorinen T, Tevola K, Liippo K, Ruuskanen O. Viruses and bacteria in bronchial samples from patients with primary hypogammaglobulinemia. Am J Respir Crit Care Med. 1999;159:1199–1204. doi: 10.1164/ajrccm.159.4.9807067. [DOI] [PubMed] [Google Scholar]
- 65.Maglione PJ, Overbey JR, Radigan L, Bagiella E, Cunningham-Rundles C. Pulmonary radiologic findings in common variable immunodeficiency: clinical and immunological correlations. Ann Allergy Asthma Immunol. 2014;113:452–459. doi: 10.1016/j.anai.2014.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Gregersen S, Aalokken TM, Mynarek G, Fevang B, Holm AM, Ueland T, et al. Development of pulmonary abnormalities in patients with common variable immunodeficiency: associations with clinical and immunologic factors. Ann Allergy Asthma Immunol. 2010;104:503–510. doi: 10.1016/j.anai.2010.04.015. [DOI] [PubMed] [Google Scholar]
- 67.Bates CA, Ellison MC, Lynch DA, Cool CD, Brown KK, Routes JM. Granulomatous-lymphocytic lung disease shortens survival in common variable immunodeficiency. J Allergy Clin Immunol. 2004;114:415–421. doi: 10.1016/j.jaci.2004.05.057. [DOI] [PubMed] [Google Scholar]
- 68.Morimoto Y, Routes JM. Granulomatous disease in common variable immunodeficiency. Curr Allergy Asthma Rep. 2005;5:370–375. doi: 10.1007/s11882-005-0008-x. [DOI] [PubMed] [Google Scholar]
- 69.Maarschalk-Ellerbroek LJ, de Jong PA, van Montfrans JM, Lammers JW, Bloem AC, Hoepelman AI, et al. CT screening for pulmonary pathology in common variable immunodeficiency disorders and the correlation with clinical and immunological parameters. J Clin Immunol. 2014;34:642–654. doi: 10.1007/s10875-014-0068-6. [DOI] [PubMed] [Google Scholar]
- 70.Quinti I, Agostini C, Tabolli S, Brunetti G, Cinetto F, Pecoraro A, et al. Malignancies are the major cause of death in patients with adult onset common variable immunodeficiency. Blood. 2012;120:1953–1954. doi: 10.1182/blood-2012-05-431064. [DOI] [PubMed] [Google Scholar]
- 71.Resnick ES, Moshier EL, Godbold JH, Cunningham-Rundles C. Morbidity and mortality in common variable immune deficiency over 4 decades. Blood. 2012;119:1650–1657. doi: 10.1182/blood-2011-09-377945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ardeniz O, Cunningham-Rundles C. Granulomatous disease in common variable immunodeficiency. Clin Immunol. 2009;133:198–207. doi: 10.1016/j.clim.2009.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Cunningham-Rundles C. Common variable immunodeficiency. Curr Allergy Asthma Rep. 2001;1:421–429. doi: 10.1007/s11882-001-0027-1. [DOI] [PubMed] [Google Scholar]
- 74.Fasano MB, Sullivan KE, Sarpong SB, Wood RA, Jones SM, Johns CJ, et al. Sarcoidosis and common variable immunodeficiency: report of 8 cases and review of the literature. Medicine (Baltimore) 1996;75:251–261. doi: 10.1097/00005792-199609000-00002. [DOI] [PubMed] [Google Scholar]
- 75.Knight AK, Cunningham-Rundles C. Inflammatory and autoimmune complications of common variable immune deficiency. Autoimmun Rev. 2006;5:156–159. doi: 10.1016/j.autrev.2005.10.002. [DOI] [PubMed] [Google Scholar]
- 76.Mullighan CG, Fanning GC, Chapel HM, Welsh KI. TNF and lymphotoxinalpha polymorphisms associated with common variable immunodeficiency: role in the pathogenesis of granulomatous disease. J Immunol. 1997;159:6236–6241. [PubMed] [Google Scholar]
- 77.Park JH, Levinson AI. Granulomatous-lymphocytic interstitial lung disease (GLILD) in common variable immunodeficiency (CVID) Clin Immunol. 2010;134:97–103. doi: 10.1016/j.clim.2009.10.002. [DOI] [PubMed] [Google Scholar]
- 78.Boursiquot JN, Gerard L, Malphettes M, Fieschi C, Galicier L, Boutboul D, et al. Granulomatous disease in CVID: retrospective analysis of clinical characteristics and treatment efficacy in a cohort of 59 patients. J Clin Immunol. 2013;33:84–95. doi: 10.1007/s10875-012-9778-9. [DOI] [PubMed] [Google Scholar]
- 79.Lin JH, Liebhaber M, Roberts RL, Dyer Z, Stiehm ER. Etanercept treatment of cutaneous granulomas in common variable immunodeficiency. J Allergy Clin Immunol. 2006;117:878–882. doi: 10.1016/j.jaci.2006.01.034. [DOI] [PubMed] [Google Scholar]
- 80.Mechanic LJ, Dikman S, Cunningham-Rundles C. Granulomatous disease in common variable immunodeficiency. Ann Intern Med. 1997;127:613–617. doi: 10.7326/0003-4819-127-8_part_1-199710150-00005. [DOI] [PubMed] [Google Scholar]
- 81.Misbah SA, Spickett GP, Esiri MM, Hughes JT, Matthews WB, Thompson RA, et al. Recurrent intra-cranial granulomata presenting as space-occupying lesions in a patient with common variable immunodeficiency. Postgrad Med J. 1992;68:359–362. doi: 10.1136/pgmj.68.799.359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Bouvry D, Mouthon L, Brillet PY, Kambouchner M, Ducroix JP, Cottin V, et al. Granulomatosis-associated common variable immunodeficiency disorder: a case-control study versus sarcoidosis. Eur Respir J. 2013;41:115–122. doi: 10.1183/09031936.00189011. [DOI] [PubMed] [Google Scholar]
- 83.Cunningham-Rundles C. Hematologic complications of primary immune deficiencies. Blood Rev. 2002;16:61–64. doi: 10.1054/blre.2001.0185. [DOI] [PubMed] [Google Scholar]
- 84.Michel M, Chanet V, Galicier L, Ruivard M, Levy Y, Hermine O, et al. Autoimmune thrombocytopenic purpura and common variable immunodeficiency: analysis of 21 cases and review of the literature. Medicine (Baltimore) 2004;83:254–263. doi: 10.1097/01.md.0000133624.65946.40. [DOI] [PubMed] [Google Scholar]
- 85.Wang J, Cunningham-Rundles C. Treatment and outcome of autoimmune hematologic disease in common variable immunodeficiency (CVID) J Autoimmun. 2005;25:57–62. doi: 10.1016/j.jaut.2005.04.006. [DOI] [PubMed] [Google Scholar]
- 86.Oliveira JB, Bleesing JJ, Dianzani U, Fleisher TA, Jaffe ES, Lenardo MJ, et al. Revised diagnostic criteria and classification for the autoimmune lymphoproliferative syndrome (ALPS): report from the 2009 NIH International Workshop. Blood. 2010;116:e35–e40. doi: 10.1182/blood-2010-04-280347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Podjasek JC, Abraham RS. Autoimmune cytopenias in common variable immunodeficiency. Front Immunol. 2012;3:189. doi: 10.3389/fimmu.2012.00189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Warnatz K, Voll RE. Pathogenesis of autoimmunity in common variable immunodeficiency. Front Immunol. 2012;3:210. doi: 10.3389/fimmu.2012.00210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Xiao X, Miao Q, Chang C, Gershwin ME, Ma X. Common variable immunodeficiency and autoimmunity—an inconvenient truth. Autoimmun Rev. 2014;13:858–864. doi: 10.1016/j.autrev.2014.04.006. [DOI] [PubMed] [Google Scholar]
- 90.Maarschalk-Ellerbroek LJ, Hoepelman AI, van Montfrans JM, Ellerbroek PM. The spectrum of disease manifestations in patients with common variable immunodeficiency disorders and partial antibody deficiency in a university hospital. J Clin Immunol. 2012;32:907–921. doi: 10.1007/s10875-012-9671-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Agarwal S, Mayer L. Pathogenesis and treatment of gastrointestinal disease in antibody deficiency syndromes. J Allergy Clin Immunol. 2009;124:658–664. doi: 10.1016/j.jaci.2009.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Malamut G, Verkarre V, Suarez F, Viallard JF, Lascaux AS, Cosnes J, et al. The enteropathy associated with common variable immunodeficiency: the delineated frontiers with celiac disease. Am J Gastroenterol. 2010;105:2262–2275. doi: 10.1038/ajg.2010.214. [DOI] [PubMed] [Google Scholar]
- 93.Aslam A, Misbah SA, Talbot K, Chapel H. Vitamin E deficiency induced neurological disease in common variable immunodeficiency: two cases and a review of the literature of vitamin E deficiency. Clin Immunol. 2004;112:24–29. doi: 10.1016/j.clim.2004.02.001. [DOI] [PubMed] [Google Scholar]
- 94.Aukrust P, Muller F, Ueland T, Svardal AM, Berge RK, Froland SS. Decreased vitamin A levels in common variable immunodeficiency: vitamin A supplementation in vivo enhances immunoglobulin production and down-regulates inflammatory responses. Eur J Clin Invest. 2000;30:252–259. doi: 10.1046/j.1365-2362.2000.00619.x. [DOI] [PubMed] [Google Scholar]
- 95.Daniels JA, Lederman HM, Maitra A, Montgomery EA. Gastrointestinal tract pathology in patients with common variable immunodeficiency (CVID): a clinicopathologic study and review. Am J Surg Pathol. 2007;31:1800–1812. doi: 10.1097/PAS.0b013e3180cab60c. [DOI] [PubMed] [Google Scholar]
- 96.Venhoff N, Emmerich F, Neagu M, Salzer U, Koehn C, Driever S, et al. The role of HLA DQ2 and DQ8 in dissecting celiac-like disease in common variable immunodeficiency. J Clin Immunol. 2013;33:909–916. doi: 10.1007/s10875-013-9892-3. [DOI] [PubMed] [Google Scholar]
- 97.Malamut G, Ziol M, Suarez F, Beaugrand M, Viallard JF, Lascaux AS, et al. Nodular regenerative hyperplasia: the main liver disease in patients with primary hypogammaglobulinemia and hepatic abnormalities. J Hepatol. 2008;48:74–82. doi: 10.1016/j.jhep.2007.08.011. [DOI] [PubMed] [Google Scholar]
- 98.Ward C, Lucas M, Piris J, Collier J, Chapel H. Abnormal liver function in common variable immunodeficiency disorders due to nodular regenerative hyperplasia. Clin Exp Immunol. 2008;153:331–337. doi: 10.1111/j.1365-2249.2008.03711.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Fuss IJ, Friend J, Yang Z, He JP, Hooda L, Boyer J, et al. Nodular regenerative hyperplasia in common variable immunodeficiency. J Clin Immunol. 2013;33:748–758. doi: 10.1007/s10875-013-9873-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Agondi RC, Barros MT, Rizzo LV, Kalil J, Giavina-Bianchi P. Allergic asthma in patients with common variable immunodeficiency. Allergy. 2010;65:510–515. doi: 10.1111/j.1398-9995.2009.02211.x. [DOI] [PubMed] [Google Scholar]
- 101.Agondi RC, Barros MT, Kokron CM, Cohon A, Oliveira AK, Kalil J, et al. Can patients with common variable immunodeficiency have allergic rhinitis? Am J Rhinol Allergy. 2013;27:79–83. doi: 10.2500/ajra.2013.27.3855. [DOI] [PubMed] [Google Scholar]
- 102.Ozcan C, Metin A, Erkocoglu M, Kocabas CN. Allergic diseases in children with primary immunodeficiencies. Turk J Pediatr. 2014;56:41–47. [PubMed] [Google Scholar]
- 103.Aghamohammadi A, Cheraghi T, Gharagozlou M, Movahedi M, Rezaei N, Yeganeh M, et al. IgA deficiency: correlation between clinical and immunological phenotypes. J Clin Immunol. 2009;29:130–136. doi: 10.1007/s10875-008-9229-9. [DOI] [PubMed] [Google Scholar]
- 104.Edwards E, Razvi S, Cunningham-Rundles C. IgA deficiency: clinical correlates and responses to pneumococcal vaccine. Clin Immunol. 2004;111:93–97. doi: 10.1016/j.clim.2003.12.005. [DOI] [PubMed] [Google Scholar]
- 105.Elenitoba-Johnson KS, Jaffe ES. Lymphoproliferative disorders associated with congenital immunodeficiencies. Semin Diagn Pathol. 1997;14:35–47. [PubMed] [Google Scholar]
- 106.Gompels MM, Hodges E, Lock RJ, Angus B, White H, Larkin A, et al. Lymphoproliferative disease in antibody deficiency: a multi-centre study. Clin Exp Immunol. 2003;134:314–320. doi: 10.1046/j.1365-2249.2003.02253.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Unger S, Seidl M, Schmitt-Graeff A, Bohm J, Schrenk K, Wehr C, et al. Ill-defined germinal centers and severely reduced plasma cells are histological hallmarks of lymphadenopathy in patients with common variable immunodeficiency. J Clin Immunol. 2014;34:615–626. doi: 10.1007/s10875-014-0052-1. [DOI] [PubMed] [Google Scholar]
- 108.Mellemkjaer L, Hammarstrom L, Andersen V, Yuen J, Heilmann C, Barington T, et al. Cancer risk among patients with IgA deficiency or common variable immunodeficiency and their relatives: a combined Danish and Swedish study. Clin Exp Immunol. 2002;130:495–500. doi: 10.1046/j.1365-2249.2002.02004.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Zullo A, Romiti A, Rinaldi V, Vecchione A, Tomao S, Aiuti F, et al. Gastric pathology in patients with common variable immunodeficiency. Gut. 1999;45:77–81. doi: 10.1136/gut.45.1.77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ueland T, Froland SS, Bollerslev J, Aukrust P. Increased levels of biochemical markers of bone turnover in relation to persistent immune activation in common variable immunodeficiency. Eur J Clin Invest. 2001;31:72–78. doi: 10.1046/j.1365-2362.2001.00768.x. [DOI] [PubMed] [Google Scholar]
- 111.Baris S, Ozen A, Ercan H, Karakoc-Aydiner E, Cagan H, Ozdemir C, et al. Osteoporosis: an ignored complication of CVID. Pediatr Allergy Immunol. 2011;22:676–683. doi: 10.1111/j.1399-3038.2011.01187.x. [DOI] [PubMed] [Google Scholar]
- 112.Ogershok PR, Hogan MB, Welch JE, Corder WT, Wilson NW. Spectrum of illness in pediatric common variable immunodeficiency. Ann Allergy Asthma Immunol. 2006;97:653–656. doi: 10.1016/S1081-1206(10)61096-4. [DOI] [PubMed] [Google Scholar]
- 113.Aghamohammadi A, Mohammadi J, Parvaneh N, Rezaei N, Moin M, Espanol T, et al. Progression of selective IgA deficiency to common variable immunodeficiency. Int Arch Allergy Immunol. 2008;147:87–92. doi: 10.1159/000135694. [DOI] [PubMed] [Google Scholar]
- 114.Robinson M, Smart J, Tang M. Common variable immune deficiency disorders: a paediatric experience. Curr Trends Immunol. 2008;9:85–91. [Google Scholar]
- 115.Goldacker S, Draeger R, Warnatz K, Huzly D, Salzer U, Thiel J, et al. Active vaccination in patients with common variable immunodeficiency (CVID) Clin Immunol. 2007;124:294–303. doi: 10.1016/j.clim.2007.04.011. [DOI] [PubMed] [Google Scholar]
- 116.Piqueras B, Lavenu-Bombled C, Galicier L, Bergeron-van der Cruyssen F, Mouthon L, Chevret S, et al. Common variable immunodeficiency patient classification based on impaired B cell memory differentiation correlates with clinical aspects. J Clin Immunol. 2003;23:385–400. doi: 10.1023/a:1025373601374. [DOI] [PubMed] [Google Scholar]
- 117.Warnatz K, Schlesier M. Flowcytometric phenotyping of common variable immunodeficiency. Cytometry B Clin Cytom. 2008;74:261–271. doi: 10.1002/cyto.b.20432. [DOI] [PubMed] [Google Scholar]
- 118.Moratto D, Gulino AV, Fontana S, Mori L, Pirovano S, Soresina A, et al. Combined decrease of defined B and T cell subsets in a group of common variable immunodeficiency patients. Clin Immunol. 2006;121:203–214. doi: 10.1016/j.clim.2006.07.003. [DOI] [PubMed] [Google Scholar]
- 119.Farrington M, Grosmaire LS, Nonoyama S, Fischer SH, Hollenbaugh D, Ledbetter JA, et al. CD40 ligand expression is defective in a subset of patients with common variable immunodeficiency. Proc Natl Acad Sci U S A. 1994;91:1099–1103. doi: 10.1073/pnas.91.3.1099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Giovannetti A, Pierdominici M, Mazzetta F, Marziali M, Renzi C, Mileo AM, et al. Unravelling the complexity of T cell abnormalities in common variable immunodeficiency. J Immunol. 2007;178:3932–3943. doi: 10.4049/jimmunol.178.6.3932. [DOI] [PubMed] [Google Scholar]
- 121.Oliva A, Scala E, Quinti I, Paganelli R, Ansotegui IJ, Giovannetti A, et al. IL-10 production and CD40L expression in patients with common variable immunodeficiency. Scand J Immunol. 1997;46:86–90. doi: 10.1046/j.1365-3083.1997.d01-95.x. [DOI] [PubMed] [Google Scholar]
- 122.Kuntz M, Goldacker S, Blum HE, Pircher H, Stampf S, Peter HH, et al. Analysis of bulk and virus-specific CD8+ T cells reveals advanced differentiation of CD8+ T cells in patients with common variable immunodeficiency. Clin Immunol. 2011;141:177–186. doi: 10.1016/j.clim.2011.07.006. [DOI] [PubMed] [Google Scholar]
- 123.Arandi N, Mirshafiey A, Jeddi-Tehrani M, Abolhassani H, Sadeghi B, Mirminachi B, et al. Evaluation of CD4+CD25+FOXP3+ regulatory T cells function in patients with common variable immunodeficiency. Cell Immunol. 2013;281:129–133. doi: 10.1016/j.cellimm.2013.03.003. [DOI] [PubMed] [Google Scholar]
- 124.Horn J, Manguiat A, Berglund LJ, Knerr V, Tahami F, Grimbacher B, et al. Decrease in phenotypic regulatory T cells in subsets of patients with common variable immunodeficiency. Clin Exp Immunol. 2009;156:446–454. doi: 10.1111/j.1365-2249.2009.03913.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Kelesidis T, Yang O. Good’s syndrome remains a mystery after 55 years: a systematic review of the scientific evidence. Clin Immunol. 2010;135:347–363. doi: 10.1016/j.clim.2010.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Hammarstrom L, Vorechovsky I, Webster D. Selective IgA deficiency (SIgAD) and common variable immunodeficiency (CVID) Clin Exp Immunol. 2000;120:225–231. doi: 10.1046/j.1365-2249.2000.01131.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Kralovicova J, Hammarstrom L, Plebani A, Webster AD, Vorechovsky I. Fine-scale mapping at IGAD1 and genome-wide genetic linkage analysis implicate HLA-DQ/DR as a major susceptibility locus in selective IgA deficiency and common variable immunodeficiency. J Immunol. 2003;170:2765–2775. doi: 10.4049/jimmunol.170.5.2765. [DOI] [PubMed] [Google Scholar]
- 128.Johnston DT, Mehaffey G, Thomas J, Young KR, Jr, Wiener H, Li J, et al. Increased frequency of HLA-B44 in recurrent sinopulmonary infections (RESPI) Clin Immunol. 2006;119:346–350. doi: 10.1016/j.clim.2006.02.001. [DOI] [PubMed] [Google Scholar]
- 129.Schaffer AA, Pfannstiel J, Webster AD, Plebani A, Hammarstrom L, Grimbacher B. Analysis of families with common variable immunodeficiency (CVID) and IgA deficiency suggests linkage of CVID to chromosome 16q. Hum Genet. 2006;118:725–729. doi: 10.1007/s00439-005-0101-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.van Zelm MC, Reisli I, van der Burg M, Sira MM, Suga K, Sekiguchi T, et al. An antibody-deficiency syndrome due to mutations in the CD19 gene. N Engl J Med. 2006;354:1901–1912. doi: 10.1056/NEJMoa051568. [DOI] [PubMed] [Google Scholar]
- 131.Kanegane H, Agematsu K, Futatani T, Castano D, van Noesel CJ, van Tol MJ, et al. Novel mutations in a Japanese patient with CD19 deficiency. Genes Immun. 2007;8:663–670. doi: 10.1038/sj.gene.6364431. [DOI] [PubMed] [Google Scholar]
- 132.Kuijpers TW, Bende RJ, Baars PA, Grummels A, Derks IA, Dolman KM, et al. CD20 deficiency in humans results in impaired T cell-independent antibody responses. J Clin Invest. 2010;120:214–222. doi: 10.1172/JCI40231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Thiel J, Kimmig L, Salzer U, Grudzien M, Lebrecht D, Hagena T, et al. Genetic CD21 deficiency is associated with hypogammaglobulinemia. J Allergy Clin Immunol. 2012;129:801.e6–810.e6. doi: 10.1016/j.jaci.2011.09.027. [DOI] [PubMed] [Google Scholar]
- 134.Salzer E, Daschkey S, Choo S, Gombert M, Santos-Valente E, Ginzel S, et al. Combined immunodeficiency with life-threatening EBV-associated lymphoproliferative disorder in patients lacking functional CD27. Haematologica. 2013;98:473–478. doi: 10.3324/haematol.2012.068791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.van Montfrans JM, Hoepelman AI, Otto S, van Gijn M, van de Corput L, de Weger RA, et al. CD27 deficiency is associated with combined immunodeficiency and persistent symptomatic EBV viremia. J Allergy Clin Immunol. 2012;129:787.e6–793.e6. doi: 10.1016/j.jaci.2011.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.van Zelm MC, Smet J, Adams B, Mascart F, Schandene L, Janssen F, et al. CD81 gene defect in humans disrupts CD19 complex formation and leads to antibody deficiency. J Clin Invest. 2010;120:1265–1274. doi: 10.1172/JCI39748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Kuehn HS, Ouyang W, Lo B, Deenick EK, Niemela JE, Avery DT, et al. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science. 2014;345:1623–1627. doi: 10.1126/science.1255904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Schubert D, Bode C, Kenefeck R, Hou TZ, Wing JB, Kennedy A, et al. Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat Med. 2014;20:1410–1416. doi: 10.1038/nm.3746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Grimbacher B, Hutloff A, Schlesier M, Glocker E, Warnatz K, Drager R, et al. Homozygous loss of ICOS is associated with adult-onset common variable immunodeficiency. Nat Immunol. 2003;4:261–268. doi: 10.1038/ni902. [DOI] [PubMed] [Google Scholar]
- 140.Takahashi N, Matsumoto K, Saito H, Nanki T, Miyasaka N, Kobata T, et al. Impaired CD4 and CD8 effector function and decreased memory T cell populations in ICOS-deficient patients. J Immunol. 2009;182:5515–5527. doi: 10.4049/jimmunol.0803256. [DOI] [PubMed] [Google Scholar]
- 141.Kotlarz D, Zietara N, Milner JD, Klein C. Human IL-21 and IL-21R deficiencies: two novel entities of primary immunodeficiency. Curr Opin Pediatr. 2014;26:704–712. doi: 10.1097/MOP.0000000000000160. [DOI] [PubMed] [Google Scholar]
- 142.Kotlarz D, Zietara N, Uzel G, Weidemann T, Braun CJ, Diestelhorst J, et al. Loss-of-function mutations in the IL-21 receptor gene cause a primary immunodeficiency syndrome. J Exp Med. 2013;210:433–443. doi: 10.1084/jem.20111229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Salzer E, Kansu A, Sic H, Majek P, Ikinciogullari A, Dogu FE, et al. Early-onset inflammatory bowel disease and common variable immunodeficiency-like disease caused by IL-21 deficiency. J Allergy Clin Immunol. 2014;133:1651.e12–1659.e12. doi: 10.1016/j.jaci.2014.02.034. [DOI] [PubMed] [Google Scholar]
- 144.Alangari A, Alsultan A, Adly N, Massaad MJ, Kiani IS, Aljebreen A, et al. LPS-responsive beige-like anchor (LRBA) gene mutation in a family with inflammatory bowel disease and combined immunodeficiency. J Allergy Clin Immunol. 2012;130:481.e2–488.e2. doi: 10.1016/j.jaci.2012.05.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Burns SO, Zenner HL, Plagnol V, Curtis J, Mok K, Eisenhut M, et al. LRBA gene deletion in a patient presenting with autoimmunity without hypogammaglobulinemia. J Allergy Clin Immunol. 2012;130:1428–1432. doi: 10.1016/j.jaci.2012.07.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Lopez-Herrera G, Tampella G, Pan-Hammarstrom Q, Herholz P, Trujillo-Vargas CM, Phadwal K, et al. Deleterious mutations in LRBA are associated with a syndrome of immune deficiency and autoimmunity. Am J Hum Genet. 2012;90:986–1001. doi: 10.1016/j.ajhg.2012.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Chen K, Coonrod EM, Kumanovics A, Franks ZF, Durtschi JD, Margraf RL, et al. Germline mutations in NFKB2 implicate the noncanonical NF-kappaB pathway in the pathogenesis of common variable immunodeficiency. Am J Hum Genet. 2013;93:812–824. doi: 10.1016/j.ajhg.2013.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Liu Y, Hanson S, Gurugama P, Jones A, Clark B, Ibrahim MA. Novel NFKB2 mutation in early-onset CVID. J Clin Immunol. 2014;34:686–690. doi: 10.1007/s10875-014-0064-x. [DOI] [PubMed] [Google Scholar]
- 149.Angulo I, Vadas O, Garcon F, Banham-Hall E, Plagnol V, Leahy TR, et al. Phosphoinositide 3-kinase delta gene mutation predisposes to respiratory infection and airway damage. Science. 2013;342:866–871. doi: 10.1126/science.1243292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Crank MC, Grossman JK, Moir S, Pittaluga S, Buckner CM, Kardava L, et al. Mutations in PIK3CD can cause hyper IgM syndrome (HIGM) associated with increased cancer susceptibility. J Clin Immunol. 2014;34:272–276. doi: 10.1007/s10875-014-0012-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Lucas CL, Kuehn HS, Zhao F, Niemela JE, Deenick EK, Palendira U, et al. Dominant-activating germline mutations in the gene encoding the PI(3)K catalytic subunit p110delta result in T cell senescence and human immunodeficiency. Nat Immunol. 2014;15:88–97. doi: 10.1038/ni.2771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Alkhairy OK, Rezaei N, Graham RR, Abolhassani H, Borte S, Hultenby K, et al. RAC2 loss-of-function mutation in 2 siblings with characteristics of common variable immunodeficiency. J Allergy Clin Immunol. 2015;135:1380.e5–1384.e5. doi: 10.1016/j.jaci.2014.10.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Wang HY, Ma CA, Zhao Y, Fan X, Zhou Q, Edmonds P, et al. Antibody deficiency associated with an inherited autosomal dominant mutation in TWEAK. Proc Natl Acad Sci U S A. 2013;110:5127–5132. doi: 10.1073/pnas.1221211110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Al-Herz W, Bousfiha A, Casanova JL, hatila T, Conley ME, Cunningham-Rundles C, et al. Primary immunodeficiency diseases: an update on the classification from the International Union of Immunological Societies Expert Committee for Primary Immunodeficiency. Front Immunol. 2014;5:162. doi: 10.3389/fimmu.2014.00162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Park JH, Resnick ES, Cunningham-Rundles C. Perspectives on common variable immune deficiency. Ann N Y Acad Sci. 2011;1246:41–49. doi: 10.1111/j.1749-6632.2011.06338.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Liadaki K, Sun J, Hammarstrom L, Pan-Hammarstrom Q. New facets of antibody deficiencies. Curr Opin Immunol. 2013;25:629–638. doi: 10.1016/j.coi.2013.06.003. [DOI] [PubMed] [Google Scholar]
- 157.Castigli E, Wilson SA, Garibyan L, Rachid R, Bonilla F, Schneider L, et al. TACI is mutant in common variable immunodeficiency and IgA deficiency. Nat Genet. 2005;37:829–834. doi: 10.1038/ng1601. [DOI] [PubMed] [Google Scholar]
- 158.Salzer U, Chapel HM, Webster AD, Pan-Hammarstrom Q, Schmitt-Graeff A, Schlesier M, et al. Mutations in TNFRSF13B encoding TACI are associated with common variable immunodeficiency in humans. Nat Genet. 2005;37:820–828. doi: 10.1038/ng1600. [DOI] [PubMed] [Google Scholar]
- 159.Pan-Hammarstrom Q, Salzer U, Du L, Bjorkander J, Cunningham-Rundles C, Nelson DL, et al. Reexamining the role of TACI coding variants in common variable immunodeficiency and selective IgA deficiency. Nat Genet. 2007;39:429–430. doi: 10.1038/ng0407-429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Salzer U, Bacchelli C, Buckridge S, Pan-Hammarstrom Q, Jennings S, Lougaris V, et al. Relevance of biallelic versus monoallelic TNFRSF13B mutations in distinguishing disease-causing from risk-increasing TNFRSF13B variants in antibody deficiency syndromes. Blood. 2009;113:1967–1976. doi: 10.1182/blood-2008-02-141937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Warnatz K, Salzer U, Rizzi M, Fischer B, Gutenberger S, Bohm J, et al. B-cell activating factor receptor deficiency is associated with an adult-onset antibody deficiency syndrome in humans. Proc Natl Acad Sci U S A. 2009;106:13945–13950. doi: 10.1073/pnas.0903543106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Pieper K, Rizzi M, Speletas M, Smulski CR, Sic H, Kraus H, et al. A common single nucleotide polymorphism impairs B-cell activating factor receptor’s multimerization, contributing to common variable immunodeficiency. J Allergy Clin Immunol. 2014;133:1222–1225. doi: 10.1016/j.jaci.2013.11.021. [DOI] [PubMed] [Google Scholar]
- 163.Mullighan CG, Marshall SE, Bunce M, Welsh KI. Variation in immunoregulatory genes determines the clinical phenotype of common variable immunodeficiency. Genes Immun. 1999;1:137–148. doi: 10.1038/sj.gene.6363653. [DOI] [PubMed] [Google Scholar]
- 164.Mullighan CG, Marshall SE, Welsh KI. Mannose binding lectin polymorphisms are associated with early age of disease onset and autoimmunity in common variable immunodeficiency. Scand J Immunol. 2000;51:111–122. doi: 10.1046/j.1365-3083.2000.00697.x. [DOI] [PubMed] [Google Scholar]
- 165.Sansom ME, Ferry BL, Sherrell ZP, Chapel HM. A preliminary assessment of alpha-1 antitrypsin S and Z deficiency allele frequencies in common variable immunodeficiency patients with and without bronchiectasis. Clin Exp Immunol. 2002;130:489–494. doi: 10.1046/j.1365-2249.2002.01995.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Alper CA, Husain Z, Larsen CE, Dubey DP, Stein R, Day C, et al. Incomplete penetrance of susceptibility genes for MHC-determined immunoglobulin deficiencies in monozygotic twins discordant for type 1 diabetes. J Autoimmun. 2006;27:89–95. doi: 10.1016/j.jaut.2006.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Cunningham-Rundles C. Human B cell defects in perspective. Immunol Res. 2012;54:227–232. doi: 10.1007/s12026-012-8318-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Fedor ME, Rubinstein A. Effects of long-term low-dose corticosteroid therapy on humoral immunity. Ann Allergy Asthma Immunol. 2006;97:113–116. doi: 10.1016/S1081-1206(10)61380-4. [DOI] [PubMed] [Google Scholar]
- 169.National Center for Immunization and Respiratory Diseases. General recommendations on immunization—recommendations of the Advisory Committee on Immunization Practices (ACIP) MMWR Recomm Rep. 2011;60:1–64. [PubMed] [Google Scholar]
- 170.Kerneis S, Launay O, Ancelle T, Iordache L, Naneix-Laroche V, Mechai F, et al. Safety and immunogenicity of yellow fever 17D vaccine in adults receiving systemic corticosteroid therapy: an observational cohort study. Arthritis Care Res (Hoboken) 2013;65:1522–1528. doi: 10.1002/acr.22021. [DOI] [PubMed] [Google Scholar]
- 171.Hanania NA, Sockrider M, Castro M, Holbrook JT, Tonascia J, Wise R, et al. Immune response to influenza vaccination in children and adults with asthma: effect of corticosteroid therapy. J Allergy Clin Immunol. 2004;113:717–724. doi: 10.1016/j.jaci.2003.12.584. [DOI] [PubMed] [Google Scholar]
- 172.Zielen S, Buhring I, Strnad N, Reichenbach J, Hofmann D. Immunogenicity and tolerance of a 7-valent pneumococcal conjugate vaccine in nonresponders to the 23-valent pneumococcal vaccine. Infect Immun. 2000;68:1435–1440. doi: 10.1128/iai.68.3.1435-1440.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Leinonen M, Sakkinen A, Kalliokoski R, Luotonen J, Timonen M, Makela PH, et al. Antibody response to 14-valent pneumococcal capsular polysaccharide vaccine in pre-school age children. Pediatr Infect Dis. 1986;5:39–44. doi: 10.1097/00006454-198601000-00008. [DOI] [PubMed] [Google Scholar]
- 174.Balloch A, Licciardi PV, Russell FM, Mulholland EK, Tang ML. Infants aged 12 months can mount adequate serotype-specific IgG responses to pneumococcal polysaccharide vaccine. J Allergy Clin Immunol. 2010;126:395–397. doi: 10.1016/j.jaci.2010.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Licciardi PV, Balloch A, Russell FM, Burton RL, Lin J, Nahm MH, et al. Pneumococcal polysaccharide vaccine at 12 months of age produces functional immune responses. J Allergy Clin Immunol. 2012;129:794.e2–800.e2. doi: 10.1016/j.jaci.2011.11.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Siber GR, Chang I, Baker S, Fernsten P, O’Brien KL, Santosham M, et al. Estimating the protective concentration of anti-pneumococcal capsular polysaccharide antibodies. Vaccine. 2007;25:3816–3826. doi: 10.1016/j.vaccine.2007.01.119. [DOI] [PubMed] [Google Scholar]
- 177.Cavaliere FM, Milito C, Martini H, Schlesier M, Drager R, Schutz K, et al. Quantification of IgM and IgA anti-pneumococcal capsular polysaccharides by a new ELISA assay: a valuable diagnostic and prognostic tool for common variable immunodeficiency. J Clin Immunol. 2013;33:838–846. doi: 10.1007/s10875-012-9856-z. [DOI] [PubMed] [Google Scholar]
- 178.Paris K, Sorensen RU. Assessment and clinical interpretation of polysaccharide antibody responses. Ann Allergy Asthma Immunol. 2007;99:462–464. doi: 10.1016/S1081-1206(10)60572-8. [DOI] [PubMed] [Google Scholar]
- 179.Orange JS, Ballow M, Stiehm ER, Ballas ZK, Chinen J, De La Morena M, et al. Use and interpretation of diagnostic vaccination in primary immunodeficiency: a working group report of the Basic and Clinical Immunology Interest Section of the American Academy of Allergy, Asthma & Immunology. J Allergy Clin Immunol. 2012;130:S1–S24. doi: 10.1016/j.jaci.2012.07.002. [DOI] [PubMed] [Google Scholar]
- 180.Ferry BL, Misbah SA, Stephens P, Sherrell Z, Lythgoe H, Bateman E, et al. Development of an anti-Salmonella typhi Vi ELISA: assessment of immunocompetence in healthy donors. Clin Exp Immunol. 2004;136:297–303. doi: 10.1111/j.1365-2249.2004.02439.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Gelfand EW, Ochs HD, Shearer WT. Controversies in IgG replacement therapy in patients with antibody deficiency diseases. J Allergy Clin Immunol. 2013;131:1001–1005. doi: 10.1016/j.jaci.2013.02.028. [DOI] [PubMed] [Google Scholar]
- 182.Fried AJ, Altrich ML, Liu H, Halsey JF, Bonilla FA. Correlation of pneumococcal antibody concentration and avidity with patient clinical and immunologic characteristics. J Clin Immunol. 2013;33:847–856. doi: 10.1007/s10875-013-9870-9. [DOI] [PubMed] [Google Scholar]
- 183.Chovancova Z, Vlkova M, Litzman J, Lokaj J, Thon V. Antibody forming cells and plasmablasts in peripheral blood in CVID patients after vaccination. Vaccine. 2011;29:4142–4150. doi: 10.1016/j.vaccine.2011.03.087. [DOI] [PubMed] [Google Scholar]
- 184.Stiehm ER, Keller MA, Vyas GN. Preparation and use of therapeutic anti-bodies primarily of human origin. Biologicals. 2008;36:363–374. doi: 10.1016/j.biologicals.2008.07.002. [DOI] [PubMed] [Google Scholar]
- 185.Bonagura VR. Using intravenous immunoglobulin (IVIG) to treat patients with primary immune deficiency disease. J Clin Immunol. 2013;33:S90–S94. doi: 10.1007/s10875-012-9838-1. [DOI] [PubMed] [Google Scholar]
- 186.Lucas M, Lee M, Lortan J, Lopez-Granados E, Misbah S, Chapel H. Infection outcomes in patients with common variable immunodeficiency disorders: relationship to immunoglobulin therapy over 22 years. J Allergy Clin Immunol. 2010;125:1354.e4–1360.e4. doi: 10.1016/j.jaci.2010.02.040. [DOI] [PubMed] [Google Scholar]
- 187.Berger M, Jolles S, Orange JS, Sleasman JW. Bioavailability of IgG administered by the subcutaneous route. J Clin Immunol. 2013;33:984–990. doi: 10.1007/s10875-013-9876-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Lucas M, Hugh-Jones K, Welby A, Misbah S, Spaeth P, Chapel H. Immunomodulatory therapy to achieve maximum efficacy: doses, monitoring, compliance, and self-infusion at home. J Clin Immunol. 2010;30:S84–S89. doi: 10.1007/s10875-010-9400-y. [DOI] [PubMed] [Google Scholar]
- 189.Orange JS, Belohradsky BH, Berger M, Borte M, Hagan J, Jolles S, et al. Evaluation of correlation between dose and clinical outcomes in subcutaneous immunoglobulin replacement therapy. Clin Exp Immunol. 2012;169:172–181. doi: 10.1111/j.1365-2249.2012.04594.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Orange JS, Grossman WJ, Navickis RJ, Wilkes MM. Impact of trough IgG on pneumonia incidence in primary immunodeficiency: a meta-analysis of clinical studies. Clin Immunol. 2010;137:21–30. doi: 10.1016/j.clim.2010.06.012. [DOI] [PubMed] [Google Scholar]
- 191.Haddad E, Berger M, Wang EC, Jones CA, Bexon M, Baggish JS. Higher doses of subcutaneous IgG reduce resource utilization in patients with primary immunodeficiency. J Clin Immunol. 2012;32:281–289. doi: 10.1007/s10875-011-9631-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Jolles S, Borte M, Nelson RP, Jr, Rojavin M, Bexon M, Lawo JP, et al. Long-term efficacy, safety, and tolerability of Hizentra(R) for treatment of primary immunodeficiency disease. Clin Immunol. 2014;150:161–169. doi: 10.1016/j.clim.2013.10.008. [DOI] [PubMed] [Google Scholar]
- 193.Brennan VM, Cochrane S, Fletcher C, Hendy D, Powell P. Surveillance of adverse reactions in patients self-infusing intravenous immunoglobulin at home. J Clin Immunol. 1995;15:116–119. doi: 10.1007/BF01541739. [DOI] [PubMed] [Google Scholar]
- 194.Brennan VM, Salome-Bentley NJ, Chapel HM. Immunology Nurses Study. Prospective audit of adverse reactions occurring in 459 primary antibody-deficient patients receiving intravenous immunoglobulin. Clin Exp Immunol. 2003;133:247–251. doi: 10.1046/j.1365-2249.2003.02199.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Horn J, Thon V, Bartonkova D, Salzer U, Warnatz K, Schlesier M, et al. Anti-IgA antibodies in common variable immunodeficiency (CVID): diagnostic workup and therapeutic strategy. Clin Immunol. 2007;122:156–162. doi: 10.1016/j.clim.2006.10.002. [DOI] [PubMed] [Google Scholar]
- 196.Rachid R, Castells M, Cunningham-Rundles C, Bonilla FA. Association of anti-IgA antibodies with adverse reactions to gamma-globulin infusion. J Allergy Clin Immunol. 2011;128:228.e1–230.e1. doi: 10.1016/j.jaci.2011.01.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Borte M, Quinti I, Soresina A, Fernandez-Cruz E, Ritchie B, Schmidt DS, et al. Efficacy and safety of subcutaneous vivaglobin(R) replacement therapy in previously untreated patients with primary immunodeficiency: a prospective, multicenter study. J Clin Immunol. 2011;31:952–961. doi: 10.1007/s10875-011-9588-5. [DOI] [PubMed] [Google Scholar]
- 198.Gustafson R, Gardulf A, Hansen S, Leibl H, Engl W, Linden M, et al. Rapid subcutaneous immunoglobulin administration every second week results in high and stable serum immunoglobulin G levels in patients with primary antibody deficiencies. Clin Exp Immunol. 2008;152:274–279. doi: 10.1111/j.1365-2249.2008.03620.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Bonilla FA. Pharmacokinetics of immunoglobulin administered via intravenous or subcutaneous routes. Immunol Allergy Clin North Am. 2008;28:803–819. doi: 10.1016/j.iac.2008.06.006. ix. [DOI] [PubMed] [Google Scholar]
- 200.Waniewski J, Gardulf A, Hammarstrom L. Bioavailability of gamma-globulin after subcutaneous infusions in patients with common variable immunodeficiency. J Clin Immunol. 1994;14:90–97. doi: 10.1007/BF01541341. [DOI] [PubMed] [Google Scholar]
- 201.Sundin U, Nava S, Hammarstrom L. Induction of unresponsiveness against IgA in IgA-deficient patients on subcutaneous immunoglobulin infusion therapy. Clin Exp Immunol. 1998;112:341–346. doi: 10.1046/j.1365-2249.1998.00571.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Gardulf A, Hammarstrom L, Smith CI. Home treatment of hypogammaglobulinaemia with subcutaneous gammaglobulin by rapid infusion. Lancet. 1991;338:162–166. doi: 10.1016/0140-6736(91)90147-h. [DOI] [PubMed] [Google Scholar]
- 203.Fasth A, Nystrom J. Safety and efficacy of subcutaneous human immunoglobulin in children with primary immunodeficiency. Acta Paediatr. 2007;96:1474–1478. doi: 10.1111/j.1651-2227.2007.00485.x. [DOI] [PubMed] [Google Scholar]
- 204.Gardulf A, Nicolay U. Replacement IgG therapy and self-therapy at home improve the health-related quality of life in patients with primary antibody deficiencies. Curr Opin Allergy Clin Immunol. 2006;6:434–442. doi: 10.1097/01.all.0000246619.49494.41. [DOI] [PubMed] [Google Scholar]
- 205.Wasserman RL. Overview of recombinant human hyaluronidase-facilitated subcutaneous infusion of IgG in primary immunodeficiencies. Immunotherapy. 2014;6:553–567. doi: 10.2217/imt.14.34. [DOI] [PubMed] [Google Scholar]
- 206.Chapel H, Brennan V, Delson E. Immunoglobulin replacement therapy by self-infusion at home. Clin Exp Immunol. 1988;73:160–162. [PMC free article] [PubMed] [Google Scholar]
- 207.Martin A, Lavoie L, Goetghebeur M, Schellenberg R. Economic benefits of subcutaneous rapid push versus intravenous immunoglobulin infusion therapy in adult patients with primary immune deficiency. Transfus Med. 2013;23:55–60. doi: 10.1111/j.1365-3148.2012.01201.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Fasth A, Nystrom J. Quality of life and health-care resource utilization among children with primary immunodeficiency receiving home treatment with subcutaneous human immunoglobulin. J Clin Immunol. 2008;28:370–378. doi: 10.1007/s10875-008-9180-9. [DOI] [PubMed] [Google Scholar]
- 209.Dichtelmuller HO, Biesert L, Fabbrizzi F, Falbo A, Flechsig E, Groner A, et al. Contribution to safety of immunoglobulin and albumin from virus partitioning and inactivation by cold ethanol fractionation: a data collection from Plasma Protein Therapeutics Association member companies. Transfusion. 2011;51:1412–1430. doi: 10.1111/j.1537-2995.2010.03003.x. [DOI] [PubMed] [Google Scholar]
- 210.Orbach H, Katz U, Sherer Y, Shoenfeld Y. Intravenous immunoglobulin: adverse effects and safe administration. Clin Rev Allergy Immunol. 2005;29:173–184. doi: 10.1385/CRIAI:29:3:173. [DOI] [PubMed] [Google Scholar]
- 211.Benadiba J, Robitaille N, Lambert G, Itaj NK, Pastore Y. Intravenous immunoglobulin-associated thrombosis: is it such a rare event? Report of a pediatric case and of the Quebec Hemovigilance System. Transfusion. 2015;55:571–575. doi: 10.1111/trf.12897. [DOI] [PubMed] [Google Scholar]
- 212.Desborough MJ, Miller J, Thorpe SJ, Murphy MF, Misbah SA. Intravenous immunoglobulin-induced haemolysis: a case report and review of the literature. Transfus Med. 2014;24:219–226. doi: 10.1111/tme.12083. [DOI] [PubMed] [Google Scholar]
- 213.Chua I, Lagos M, Charalambous BM, Workman S, Chee R, Grimbacher B. Pathogen-specific IgG antibody levels in immunodeficient patients receiving immunoglobulin replacement do not provide additional benefit to therapeutic management over total serum IgG. J Allergy Clin Immunol. 2011;127:1410–1411. doi: 10.1016/j.jaci.2011.01.035. [DOI] [PubMed] [Google Scholar]
- 214.Junker AK, Bonilla FA, Sullivan KE. How to flee the flu. Clin Immunol. 2004;112:219–220. doi: 10.1016/j.clim.2004.03.013. [DOI] [PubMed] [Google Scholar]
- 215.Buckley RH, Ballas Z, Ballow M, Blaese M, Bonilla FA, Conley ME, et al. Recommendations for live viral and bacterial vaccines in immunodeficient patients and their close contacts. J Allergy Clin Immunol. 2014;133:961–966. doi: 10.1016/j.jaci.2013.11.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Nobre FA, Gonzalez IG, Simao RM, de Moraes Pinto MI, Costa-Carvalho BT. Antibody levels to tetanus, diphtheria, measles and varicella in patients with primary immunodeficiency undergoing intravenous immunoglobulin therapy: a prospective study. BMC Immunol. 2014;15:26. doi: 10.1186/1471-2172-15-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Misbah SA, Lawrence PA, Kurtz JB, Chapel HM. Prolonged faecal excretion of poliovirus in a nurse with common variable hypogammaglobulinaemia. Postgrad Med J. 1991;67:301–303. doi: 10.1136/pgmj.67.785.301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Bonilla FA, Bernstein IL, Khan DA, Ballas ZK, Chinen J, Frank MM, et al. Practice parameter for the diagnosis and management of primary immunodeficiency. Ann Allergy Asthma Immunol. 2005;94:S1–S63. doi: 10.1016/s1081-1206(10)61142-8. [DOI] [PubMed] [Google Scholar]
- 219.Lopez-Boado YS, Rubin BK. Macrolides as immunomodulatory medications for the therapy of chronic lung diseases. Curr Opin Pharmacol. 2008;8:286–291. doi: 10.1016/j.coph.2008.01.010. [DOI] [PubMed] [Google Scholar]
- 220.Ilowite J, Spiegler P, Chawla S. Bronchiectasis: new findings in the pathogenesis and treatment of this disease. Curr Opin Infect Dis. 2008;21:163–167. doi: 10.1097/QCO.0b013e3282f4f237. [DOI] [PubMed] [Google Scholar]
- 221.Altenburg J, de Graaff CS, Stienstra Y, Sloos JH, van Haren EH, Koppers RJ, et al. Effect of azithromycin maintenance treatment on infectious exacerbations among patients with non-cystic fibrosis bronchiectasis: the BAT randomized controlled trial. JAMA. 2013;309:1251–1259. doi: 10.1001/jama.2013.1937. [DOI] [PubMed] [Google Scholar]
- 222.Wong C, Jayaram L, Karalus N, Eaton T, Tong C, Hockey H, et al. Azithromycin for prevention of exacerbations in non-cystic fibrosis bronchiectasis (EMBRACE): a randomised, double-blind, placebo-controlled trial. Lancet. 2012;380:660–667. doi: 10.1016/S0140-6736(12)60953-2. [DOI] [PubMed] [Google Scholar]
- 223.Kellett F, Robert NM. Nebulised 7% hypertonic saline improves lung function and quality of life in bronchiectasis. Respir Med. 2011;105:1831–1835. doi: 10.1016/j.rmed.2011.07.019. [DOI] [PubMed] [Google Scholar]
- 224.Quartier P, Foray S, Casanova JL, Hau-Rainsard I, Blanche S, Fischer A. Enteroviral meningoencephalitis in X-linked agammaglobulinemia: intensive immunoglobulin therapy and sequential viral detection in cerebrospinal fluid by polymerase chain reaction. Pediatr Infect Dis J. 2000;19:1106–1108. doi: 10.1097/00006454-200011000-00020. [DOI] [PubMed] [Google Scholar]
- 225.Onbasi K, Gunsar F, Sin AZ, Ardeniz O, Kokuludag A, Sebik F. Common variable immunodeficiency (CVID) presenting with malabsorption due to giardiasis. Turk J Gastroenterol. 2005;16:111–113. [PubMed] [Google Scholar]
- 226.Spickett GP, Zhang JG, Green T, Shrimankar J. Granulomatous disease in common variable immunodeficiency: effect on immunoglobulin replacement therapy and response to steroids and splenectomy. J Clin Pathol. 1996;49:431–434. doi: 10.1136/jcp.49.5.431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Longhurst HJ, O’Grady C, Evans G, De Lord C, Hughes A, Cavenagh J, et al. Anti-D immunoglobulin treatment for thrombocytopenia associated with primary antibody deficiency. J Clin Pathol. 2002;55:64–66. doi: 10.1136/jcp.55.1.64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Gobert D, Bussel JB, Cunningham-Rundles C, Galicier L, Dechartres A, Berezne A, et al. Efficacy and safety of rituximab in common variable immunodeficiency-associated immune cytopenias: a retrospective multicentre study on 33 patients. Br J Haematol. 2011;155:498–508. doi: 10.1111/j.1365-2141.2011.08880.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Seve P, Bourdillon L, Sarrot-Reynauld F, Ruivard M, Jaussaud R, Bouhour D, et al. Autoimmune hemolytic anemia and common variable immunodeficiency: a case-control study of 18 patients. Medicine (Baltimore) 2008;87:177–184. doi: 10.1097/MD.0b013e31817a90ba. [DOI] [PubMed] [Google Scholar]
- 230.Wong GK, Goldacker S, Winterhalter C, Grimbacher B, Chapel H, Lucas M, et al. Outcomes of splenectomy in patients with common variable immunodeficiency (CVID): a survey of 45 patients. Clin Exp Immunol. 2013;172:63–72. doi: 10.1111/cei.12039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Arnold DF, Wiggins J, Cunningham-Rundles C, Misbah SA, Chapel HM. Granulomatous disease: distinguishing primary antibody disease from sarcoidosis. Clin Immunol. 2008;128:18–22. doi: 10.1016/j.clim.2008.03.510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Hatab AZ, Ballas ZK. Caseating granulomatous disease in common variable immunodeficiency treated with infliximab. J Allergy Clin Immunol. 2005;116:1161–1162. doi: 10.1016/j.jaci.2005.08.041. [DOI] [PubMed] [Google Scholar]
- 233.Thatayatikom A, Thatayatikom S, White AJ. Infliximab treatment for severe granulomatous disease in common variable immunodeficiency: a case report and review of the literature. Ann Allergy Asthma Immunol. 2005;95:293–300. doi: 10.1016/S1081-1206(10)61228-8. [DOI] [PubMed] [Google Scholar]
- 234.Mannon PJ, Fuss IJ, Dill S, Friend J, Groden C, Hornung R, et al. Excess IL-12 but not IL-23 accompanies the inflammatory bowel disease associated with common variable immunodeficiency. Gastroenterology. 2006;131:748–756. doi: 10.1053/j.gastro.2006.06.022. [DOI] [PubMed] [Google Scholar]
- 235.Baris S, Ercan H, Cagan HH, Ozen A, Karakoc-Aydiner E, Ozdemir C, et al. Efficacy of intravenous immunoglobulin treatment in children with common variable immunodeficiency. J Investig Allergol Clin Immunol. 2011;21:514–521. [PubMed] [Google Scholar]
- 236.Chua I, Standish R, Lear S, Harbord M, Eren E, Raeiszadeh M, et al. Anti-tumour necrosis factor-alpha therapy for severe enteropathy in patients with common variable immunodeficiency (CVID) Clin Exp Immunol. 2007;150:306–311. doi: 10.1111/j.1365-2249.2007.03481.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Chase NM, Verbsky JW, Hintermeyer MK, Waukau JK, Tomita-Mitchell A, Casper JT, et al. Use of combination chemotherapy for treatment of granulomatous and lymphocytic interstitial lung disease (GLILD) in patients with common variable immunodeficiency (CVID) J Clin Immunol. 2013;33:30–39. doi: 10.1007/s10875-012-9755-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Davies CW, Juniper MC, Gray W, Gleeson FV, Chapel HM, Davies RJ. Lymphoid interstitial pneumonitis associated with common variable hypogammaglobulinaemia treated with cyclosporin A. Thorax. 2000;55:88–90. doi: 10.1136/thorax.55.1.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Rizzi M, Neumann C, Fielding AK, Marks R, Goldacker S, Thaventhiran J, et al. Outcome of allogeneic stem cell transplantation in adults with common variable immunodeficiency. J Allergy Clin Immunol. 2011;128:1371.e2–1374.e2. doi: 10.1016/j.jaci.2011.07.055. [DOI] [PubMed] [Google Scholar]
- 240.Wehr C, Gennery AR, Lindemans C, Schulz A, Hoenig M, Marks R, et al. Multicenter experience in hematopoietic stem cell transplantation for serious complications of common variable immunodeficiency. J Allergy Clin Immunol. 2015;135:988.e6–997.e6. doi: 10.1016/j.jaci.2014.11.029. [DOI] [PubMed] [Google Scholar]
- 241.Eijkhout HW, van Der Meer JW, Kallenberg CG, Weening RS, van Dissel JT, Sanders LA, et al. The effect of two different dosages of intravenous immunoglobulin on the incidence of recurrent infections in patients with primary hypogammaglobulinemia: a randomized, double-blind, multicenter crossover trial. Ann Intern Med. 2001;135:165–174. doi: 10.7326/0003-4819-135-3-200108070-00008. [DOI] [PubMed] [Google Scholar]
- 242.Garcia-Lloret M, McGhee S, Chatila TA. Immunoglobulin replacement therapy in children. Immunol Allergy Clin North Am. 2008;28:833–849. doi: 10.1016/j.iac.2008.07.001. ix. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Piatosa B, Pac M, Siewiera K, Klaudel-Dreszler M, Heropolitanska-Pliszka E, et al. Common variable immune deficiency in children–clinical characteristics varies depending on defect in peripheral B cell maturation. J Clin Immunol. 2013;33:731–741. doi: 10.1007/s10875-013-9875-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Aydogan M, Eifan AO, Gocmen I, Ozdemir C, Bahceciler NN, Barlan IB. Clinical and immunologic features of pediatric patients with common variable immunodeficiency and respiratory complications. J Investig Allergol Clin Immunol. 2008;18:260–265. [PubMed] [Google Scholar]
- 245.Touw CM, van de Ven AA, de Jong PA, Terheggen-Lagro S, Beek E, Sanders EA, et al. Detection of pulmonary complications in common variable immunodeficiency. Pediatr Allergy Immunol. 2010;21:793–805. doi: 10.1111/j.1399-3038.2009.00963.x. [DOI] [PubMed] [Google Scholar]
- 246.Llobet MP, Soler-Palacin P, Detkova D, Hernandez M, Caragol I, Espanol T. Common variable immunodeficiency: 20-yr experience at a single centre. Pediatr Allergy Immunol. 2009;20:113–118. doi: 10.1111/j.1399-3038.2008.00744.x. [DOI] [PubMed] [Google Scholar]
- 247.Karakoc-Aydiner E, Ozen AO, Baris S, Ercan H, Ozdemir C, Barlan IB. Alterations in humoral immunity is common among family members of CVID patients. J Investig Allergol Clin Immunol. 2014;24:346–351. [PubMed] [Google Scholar]
- 248.Gardulf A, Andersson E, Lindqvist M, Hansen S, Gustafson R. Rapid subcutaneous IgG replacement therapy at home for pregnant immunodeficient women. J Clin Immunol. 2001;21:150–154. doi: 10.1023/a:1011051704960. [DOI] [PubMed] [Google Scholar]
- 249.Brinker KA, Silk HJ. Common variable immune deficiency and treatment with intravenous immunoglobulin during pregnancy. Ann Allergy Asthma Immunol. 2012;108:464–465. doi: 10.1016/j.anai.2012.04.014. [DOI] [PubMed] [Google Scholar]
- 250.Cunningham-Rundles C. Key aspects for successful immunoglobulin therapy of primary immunodeficiencies. Clin Exp Immunol. 2011;164:16–19. doi: 10.1111/j.1365-2249.2011.04390.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Hypogammaglobulinaemia in the United Kingdom: summary report of a Medical Research Council working-party. Lancet. 1969;1:163–168. [PubMed] [Google Scholar]
- 252.Hermaszewski RA, Webster AD. Primary hypogammaglobulinaemia: a survey of clinical manifestations and complications. Q J Med. 1993;86:31–42. [PubMed] [Google Scholar]
- 253.Dorsey MJ, Orange JS. Impaired specific antibody response and increased B-cell population in transient hypogammaglobulinemia of infancy. Ann Allergy Asthma Immunol. 2006;97:590–595. doi: 10.1016/S1081-1206(10)61085-X. [DOI] [PubMed] [Google Scholar]
- 254.Morinishi Y, Imai K, Nakagawa N, Sato H, Horiuchi K, Ohtsuka Y, et al. Identification of severe combined immunodeficiency by T-cell receptor excision circles quantification using neonatal guthrie cards. J Pediatr. 2009;155:829–833. doi: 10.1016/j.jpeds.2009.05.026. [DOI] [PubMed] [Google Scholar]
- 255.Nakagawa N, Imai K, Kanegane H, Sato H, Yamada M, Kondoh K, et al. Quantification of kappa-deleting recombination excision circles in Guthrie cards for the identification of early B-cell maturation defects. J Allergy Clin Immunol. 2011;128:223.e2–225.e2. doi: 10.1016/j.jaci.2011.01.052. [DOI] [PubMed] [Google Scholar]
- 256.Aghamohammadi A, Moin M, Kouhi A, Mohagheghi MA, Shirazi A, Rezaei N, et al. Chromosomal radiosensitivity in patients with common variable immunodeficiency. Immunobiology. 2008;213:447–454. doi: 10.1016/j.imbio.2007.10.018. [DOI] [PubMed] [Google Scholar]
- 257.Serra G, Milito C, Mitrevski M, Granata G, Martini H, Pesce AM, et al. Lung MRI as a possible alternative to CT scan for patients with primary immune deficiencies and increased radiosensitivity. Chest. 2011;140:1581–1589. doi: 10.1378/chest.10-3147. [DOI] [PubMed] [Google Scholar]
- 258.Gabriel SE, Normand SL. Getting the methods right–the foundation of patient-centered outcomes research. N Engl J Med. 2012;367:787–790. doi: 10.1056/NEJMp1207437. [DOI] [PubMed] [Google Scholar]
- 259.Kaveri SV, Maddur MS, Hegde P, Lacroix-Desmazes S, Bayry J. Intravenous immunoglobulins in immunodeficiencies: more than mere replacement therapy. Clin Exp Immunol. 2011;164:2–5. doi: 10.1111/j.1365-2249.2011.04387.x. [DOI] [PMC free article] [PubMed] [Google Scholar]