The Host with Compromised Defenses: The Primary Immunodeficiencies in Adults - A Critical Care Perspective
Abstract
Primary immunodeficiencies (PIDs), once considered exclusively pediatric disorders, are increasingly recognized in adult critical care settings. The survival of patients with PIDs into adulthood, coupled with growing awareness of adult-onset presentations, has transformed the landscape of immunodeficiency management in intensive care units. This review examines key PIDs encountered in adult critical care practice, emphasizing diagnostic approaches, life-threatening complications, and evidence-based management strategies. We focus on Common Variable Immunodeficiency, Chronic Granulomatous Disease, and Hyper-IgE Syndrome, while providing practical guidance on recognizing immunodeficiency in critically ill adults.
Keywords: Primary immunodeficiency, Common Variable Immunodeficiency, Chronic Granulomatous Disease, Hyper-IgE Syndrome, IVIG, critical care
Introduction
Primary immunodeficiencies comprise over 450 distinct genetic disorders affecting innate and adaptive immunity. While historically diagnosed in childhood, approximately 25-30% of PIDs present after age 20 years, with some manifestations occurring well into the sixth decade. The intensivist must maintain a high index of suspicion, as these patients frequently present with sepsis, unusual opportunistic infections, or paradoxical inflammatory complications that confound conventional critical care management.
The epidemiological burden is substantial. CVID affects approximately 1:25,000 to 1:50,000 individuals, making it the most common symptomatic PID in adults. CGD occurs in 1:200,000 to 1:250,000 live births, while Hyper-IgE Syndrome is exceedingly rare at approximately 1:1,000,000. Despite these "rare disease" classifications, the aggregate burden of PIDs in critical care settings warrants systematic understanding.
Common Variable Immunodeficiency (CVID): Beyond Recurrent Infections to Granulomatous Disease and Lymphoma
Pathophysiology and Immunologic Hallmarks
CVID represents a heterogeneous group of disorders characterized by hypogammaglobulinemia (IgG typically <400 mg/dL), reduced IgA and/or IgM, impaired specific antibody responses, and exclusion of other causes of hypogammaglobulinemia. Monogenic causes are identified in only 10-30% of cases, involving genes such as TNFRSF13B (TACI), TNFRSF13C (BAFF-R), CD19, CD20, CD21, CD81, ICOS, and CTLA4.
The immunologic defect extends beyond antibody deficiency. B-cell maturation arrest, T-cell dysfunction, dysregulated T follicular helper cells, and impaired regulatory T-cell function contribute to the protean manifestations. This immune dysregulation explains why CVID patients suffer not only from infections but also from autoimmunity, granulomatous disease, and malignancy.
Pearl #1: The CVID Phenotype Spectrum
CVID patients can be phenotyped into distinct categories based on B-cell flow cytometry (EUROclass classification) and clinical manifestations (Paris classification). The "infection-only" phenotype carries the best prognosis, while patients with granulomatous-lymphocytic interstitial lung disease (GLILD) or enteropathy have significantly increased mortality (hazard ratios of 5-11).
Clinical Presentation in Critical Care
Infectious Complications:
- Bacterial pneumonia: Encapsulated organisms (Streptococcus pneumoniae, Haemophilus influenzae) predominate, but unusual organisms including Mycoplasma and Ureaplasma species occur
- Chronic sinopulmonary infections: Bronchiectasis develops in 30-60% of patients, predisposing to acute exacerbations requiring mechanical ventilation
- Enteropathogenic infections: Giardia lamblia, Campylobacter, Salmonella, and norovirus cause chronic diarrhea and malabsorption
- Meningoencephalitis: Enteroviruses cause chronic CNS infections, often vaccine-derived strains in patients who received live vaccines
Non-Infectious Complications:
Granulomatous-Lymphocytic Interstitial Lung Disease (GLILD): Present in 10-25% of CVID patients, GLILD mimics sarcoidosis with noncaseating granulomas, lymphoid hyperplasia, and follicular bronchiolitis. Critical presentations include acute respiratory failure, pulmonary hypertension, and cor pulmonale. High-resolution CT reveals ground-glass opacities, nodules, lymphadenopathy, and consolidation. Diagnosis requires lung biopsy, though bronchoscopy with lavage and transbronchial biopsy may suffice in appropriate clinical contexts.
Oyster #1: GLILD vs. Infection—The Diagnostic Dilemma
The patient with CVID presenting with new pulmonary infiltrates creates a diagnostic conundrum. Is this bacterial pneumonia requiring antibiotics, GLILD requiring immunosuppression, or both? This distinction is critical as inappropriate immunosuppression in active infection proves catastrophic.
Diagnostic approach:
- Aggressive microbiologic sampling (sputum, bronchoscopy with BAL, blood cultures)
- PET-CT scanning (FDG-avid nodules suggest GLILD; pneumonia typically shows different patterns)
- Serum biomarkers: Elevated soluble IL-2 receptor, IgM, and reduced IgA paradoxically suggest immune activation
- Lung biopsy when feasible
- Empiric broad-spectrum antibiotics while pursuing diagnosis
Autoimmune Cytopenias: Autoimmune hemolytic anemia, immune thrombocytopenia, and Evans syndrome (combined AIHA and ITP) occur in 10-30% of patients. These may precipitate ICU admission through life-threatening bleeding or severe anemia.
Lymphoproliferative Disease: Non-Hodgkin lymphoma risk increases 10-25 fold, with increased incidence of gastric and colon cancer. Lymphoma may present with sepsis-like syndrome, lymphadenopathy-related obstruction, or tumor lysis syndrome.
Management in Critical Care
Immunoglobulin Replacement: Standard IVIG dosing (400-600 mg/kg every 3-4 weeks) targets trough IgG levels >500-600 mg/dL, though some patients require higher troughs (>800 mg/dL) to prevent breakthrough infections. In critically ill patients:
- Consider loading doses (1-2 g/kg over 2-5 days)
- Monitor for adverse effects: thromboembolism, renal dysfunction, hemolysis, aseptic meningitis
- Subcutaneous immunoglobulin (SCIG) may be preferable in stable patients but is impractical in acute critical illness
Hack #1: IVIG in Septic Shock
While high-dose IVIG (1-2 g/kg) in septic shock remains controversial in immunocompetent patients, CVID patients with sepsis and profound hypogammaglobulinemia represent a special case. Consider aggressive IVIG loading alongside source control and antimicrobials. Some centers use both standard IVIG and IgM-enriched preparations, though evidence is anecdotal.
Antimicrobial Management:
- Empiric broad-spectrum coverage must include encapsulated organisms
- Prolonged courses often required due to impaired immune clearance
- Consider atypical organisms (Mycoplasma, Ureaplasma) in pneumonia
- Prophylaxis: Trimethoprim-sulfamethoxazole for PCP, azithromycin for bronchiectasis exacerbations
Immunomodulation for Non-Infectious Complications:
- GLILD: Corticosteroids (prednisone 0.5-1 mg/kg), rituximab (375 mg/m² weekly × 4), azathioprine, or mycophenolate mofetil
- Autoimmune cytopenias: High-dose corticosteroids, IVIG (1-2 g/kg), rituximab
- Refractory cases: Consider abatacept, infliximab, or JAK inhibitors in consultation with immunology
Key References:
- Bonilla FA, et al. International Consensus Document (ICON): Common Variable Immunodeficiency Disorders. J Allergy Clin Immunol Pract. 2016;4(1):38-59.
- Resnick ES, et al. Morbidity and mortality in common variable immune deficiency over 4 decades. Blood.2012;119(7):1650-1657.
- Maglione PJ, et al. Pulmonary manifestations of primary immunodeficiency diseases. Eur Respir Rev.2019;28(154):190055.
Chronic Granulomatous Disease (CGD): The Nitroblue Tetrazolium Test and the Catalase-Positive Organisms
Molecular Basis and Inheritance
CGD results from defects in the NADPH oxidase complex (phagocyte oxidase), which generates superoxide radicals essential for microbial killing. Five genetic subtypes exist, affecting gp91-phox (CYBB, X-linked, ~65%), p47-phox (NCF1, autosomal recessive, ~25%), p67-phox (NCF2, ~5%), p22-phox (CYBA, ~5%), and p40-phox (NCF4, rare). X-linked CGD typically presents earlier and more severely than autosomal recessive forms, though exceptions occur.
The oxidative burst defect specifically impairs killing of catalase-positive organisms. Catalase-negative organisms (most streptococci) generate hydrogen peroxide that phagocytes can use for killing through myeloperoxidase-mediated pathways, partially compensating for NADPH oxidase deficiency. Catalase-positive organisms (Staphylococcus aureus, Burkholderia cepacia complex, Serratia, Nocardia, Aspergillus) destroy their own hydrogen peroxide, eliminating this compensatory mechanism.
Pearl #2: The Catalase Connection
The remarkable specificity of CGD infections for catalase-positive organisms provides a diagnostic clue. When confronting a young adult with Burkholderia cepacia, Serratia marcescens, or Chromobacterium violaceum sepsis, think CGD. These organisms are relatively uncommon in immunocompetent hosts but cause devastating infections in CGD.
Diagnostic Approaches
Nitroblue Tetrazolium (NBT) Test: This historical screening test exploits the oxidative burst defect. Normal neutrophils reduce colorless NBT to blue-black formazan granules upon stimulation. CGD neutrophils fail to reduce NBT, remaining colorless. While simple and inexpensive, NBT testing has limitations:
- Subjective interpretation
- Cannot detect carrier states
- False negatives in X-linked carriers with skewed X-inactivation
- Being supplanted by flow cytometry
Dihydrorhodamine (DHR) Flow Cytometry: The gold standard diagnostic test. DHR-123, a non-fluorescent dye, is oxidized to fluorescent rhodamine-123 by hydrogen peroxide produced during the oxidative burst. CGD neutrophils show absent or markedly reduced fluorescence. DHR offers advantages:
- Quantitative assessment
- Distinguishes X-linked carriers
- Identifies the percentage of normal vs. abnormal neutrophils
- Determines CGD subtype (X-linked vs. autosomal recessive pattern)
Genetic Testing: Confirms diagnosis and enables carrier detection and prenatal testing. Essential for definitive classification and consideration of hematopoietic stem cell transplantation.
Oyster #2: The CGD Patient with "Sterile" Cultures
CGD patients frequently develop granulomatous masses in liver, spleen, lungs, and lymph nodes. Biopsies reveal inflammation and granulomas but sterile cultures. This occurs because:
- Organisms may be dead but incompletely cleared
- Fungi and mycobacteria are difficult to culture from granulomas
- Prior antimicrobial therapy
- Hyperinflammation (excess IL-1, IL-18) causes tissue damage independent of active infection
Clinical approach: Empirically treat for fungal infection (voriconazole or posaconazole) and consider corticosteroids for hyperinflammatory component after excluding active infection. Novel IL-1 blockade (anakinra) shows promise in refractory cases.
Critical Care Presentations
Life-Threatening Infections:
Aspergillus Pneumonia: The leading cause of death in CGD. Aspergillus fumigatus and increasingly azole-resistant species cause necrotizing pneumonia, often requiring surgical resection. CT shows nodules, infiltrates, and pleural involvement. Diagnosis requires bronchoscopy with BAL (galactomannan, PCR, culture) or CT-guided biopsy.
Management:
- Voriconazole (loading 6 mg/kg IV q12h × 2, then 4 mg/kg q12h) or isavuconazole
- Therapeutic drug monitoring essential (voriconazole trough 2-5 μg/mL)
- Liposomal amphotericin B for refractory cases or azole resistance
- Adjunctive granulocyte transfusions (controversial, limited availability)
- Surgical debridement for localized disease
- Recombinant interferon-gamma may augment phagocyte function (50 μg/m² SC three times weekly)
Burkholderia cepacia Complex: Causes pneumonia, bacteremia, and septic shock with high mortality. B. cepacia is intrinsically multidrug-resistant. Treatment requires combination therapy: typically trimethoprim-sulfamethoxazole plus meropenem or ceftazidime, guided by susceptibilities. Duration: 4-6 months minimum.
Staphylococcus aureus Infections: Recurrent skin abscesses, osteomyelitis, hepatic abscesses, and septic arthritis. MRSA is increasingly common. Prolonged therapy (6-12 weeks) required for deep-seated infections.
Mycobacterial Infections: Both tuberculosis and non-tuberculous mycobacteria (especially M. avium complex, M. abscessus) cause disseminated disease. BCG vaccination may cause disseminated BCGosis.
Gastrointestinal Complications:
- CGD colitis: Mimics Crohn's disease with transmural inflammation, strictures, and fistulae
- Gastric outlet obstruction from granulomas
- Hepatic/splenic abscesses
- Perianal abscesses and fistulae
Obstructive Complications: Granulomatous inflammation causes:
- Genitourinary obstruction (ureteral, bladder outlet)
- Gastric outlet or intestinal obstruction
- Bronchial obstruction
Management Principles
Prophylaxis (outpatient, but ICU teams must continue):
- Trimethoprim-sulfamethoxazole: 5 mg/kg/day TMP component
- Itraconazole 200 mg daily (or posaconazole in high-risk patients)
- Interferon-gamma 50 μg/m² three times weekly
- These reduce serious infections by 50-70%
Antimicrobial Selection in Critical Illness:
- Always cover S. aureus (including MRSA) and Aspergillus empirically
- Add Burkholderia coverage (meropenem or ceftazidime) if pneumonia present
- For intra-abdominal sepsis, consider Nocardia and Serratia
- Prolonged courses essential (weeks to months)
Surgical Intervention:
- Abscesses often require drainage despite antimicrobial therapy
- Obstructive lesions may need resection or stenting
- Aspergillus lesions may require lobectomy
Immunomodulation:
- Corticosteroids for hyperinflammatory complications (colitis, obstructive granulomas)
- Dose: Prednisone 0.5-1 mg/kg daily, taper based on response
- Risk-benefit analysis essential given infection risk
- Anakinra (IL-1 receptor antagonist) emerging as alternative
Key References:
- Marciano BE, et al. Common severe infections in chronic granulomatous disease. Clin Infect Dis. 2015;60(8):1176-1183.
- Leiding JW, et al. Chronic Granulomatous Disease. GeneReviews. University of Washington, Seattle; Updated 2020.
- Kuhns DB, et al. Residual NADPH oxidase and survival in chronic granulomatous disease. N Engl J Med.2010;363(27):2600-2610.
Hyper-IgE Syndrome (Job's Syndrome): The Triad of Abscesses, Pneumatoceles, and Eczema
Genetic Subtypes and Pathophysiology
Classical autosomal dominant Hyper-IgE Syndrome (AD-HIES) results from dominant-negative STAT3 mutations. STAT3 is crucial for IL-6, IL-10, IL-11, IL-21, and IL-23 signaling, affecting Th17 cell differentiation, B-cell class switching, and tissue remodeling. Autosomal recessive forms involve DOCK8 (dedicator of cytokinesis 8) mutations, causing a distinct phenotype with severe viral infections, food allergies, and malignancy susceptibility.
The IgE elevation (typically >2000 IU/mL, often >10,000) results from impaired IL-10 signaling and dysregulated B-cell function. Paradoxically, eosinophilia is common despite elevated IgE, and anaphylaxis is rare.
Pearl #3: The "Job" Eponym
The syndrome's alternative name references the biblical figure Job, who was "smote with sore boils from the sole of his foot unto his crown." This colorfully describes the recurrent cold staphylococcal abscesses characteristic of the disease. The "cold" designation reflects minimal surrounding inflammation despite purulent collections.
Clinical Phenotype
Pathognomonic Triad (Classic AD-HIES):
- Recurrent staphylococcal abscesses: Skin, lungs, joints; characteristically "cold" with minimal erythema or warmth
- Pneumatoceles: Persistent air-filled cysts following pneumonia, prone to superinfection with Aspergillus or Pseudomonas
- Severe eczema: Early-onset, persistent, often confused with atopic dermatitis
Connective Tissue Abnormalities:
- Characteristic facial features: Prominent forehead, deep-set eyes, broad nasal bridge, facial asymmetry (develop with age)
- Scoliosis (>60% of patients)
- Bone fractures from minimal trauma (reduced bone density despite normal DEXA)
- Retained primary teeth (failure of physiologic root resorption)
- Hyperextensible joints
Vascular Abnormalities:
- Coronary artery aneurysms and ectasia (major cause of mortality)
- Intracranial aneurysms
- Aortic dilation
- Increased stroke risk from aneurysm rupture or thrombosis
Diagnostic Criteria
NIH Scoring System: Weighted score ≥40 suggests HIES (maximum 360 points):
- Highest IgE level: 10 IU/mL (0 pts) to >2000 IU/mL (10 pts)
- Eczema severity: 0-4 (0-5 pts)
- Upper respiratory infections: 0-1 per year (0 pts) to >2 per year (4 pts)
- Pneumonia episodes: 0 (0 pts) to ≥3 (10 pts)
- Parenchymal lung abnormalities: None (0 pts) to bronchiectasis or pneumatoceles (5 pts)
- Retained primary teeth: >3 after age 13 (8 pts)
- Scoliosis: Maximum curvature >10° (5 pts)
- Fractures: 0 (0 pts) to >3 with minimal trauma (5 pts)
- Characteristic face: 0-5 (0-5 pts)
- High palate: 0-5 (0-5 pts)
Genetic testing of STAT3 confirms diagnosis and distinguishes from recessive forms (DOCK8, TYK2, ZNF341).
Critical Care Complications
Invasive Fungal Infections: Pneumatoceles become colonized with Aspergillus fumigatus, forming fungus balls (aspergillomas). Invasive pulmonary aspergillosis may follow, particularly during corticosteroid therapy or viral infections. Mucormycosis, Cryptococcus, and endemic fungi also occur.
Management:
- Voriconazole or isavuconazole first-line
- CT-guided aspiration of aspergillomas may reduce fungal burden
- Surgical resection if medical therapy fails (high morbidity given abnormal healing)
- Long-term suppressive therapy often required
Pneumonia with Pneumatocele Formation: S. aureus pneumonia causes necrotizing inflammation with pneumatocele formation in 75% of cases. Pneumatoceles persist lifelong, creating a substrate for chronic infections.
Acute management:
- Anti-staphylococcal therapy: Vancomycin for MRSA, nafcillin/oxacillin for MSSA
- Chest tube placement controversial (may promote bronchopleural fistula)
- Avoid positive pressure ventilation when possible (increases pneumatocele tension)
Hemoptysis: Major or massive hemoptysis may result from:
- Aspergilloma erosion into pulmonary vessels
- Bronchiectasis with friable mucosa
- Pseudomonas or Mycobacterium abscessus superinfection
Management algorithm:
- Stabilize: Large-bore IV access, type and cross, reverse coagulopathy
- Localize: CT angiography (avoid if renal dysfunction; use interventional radiology consultation)
- Bronchoscopy: Identify bleeding source, consider topical hemostatic agents
- Bronchial artery embolization: First-line intervention for ongoing bleeding
- Surgical resection: For refractory bleeding or destroyed lung segments
- Tranexamic acid: May reduce bleeding (1g IV TID)
Oyster #3: Corticosteroids in HIES—A Double-Edged Sword
Eczema in HIES may be severe and refractory, tempting clinicians to use systemic corticosteroids. However, steroids dramatically increase infection risk in these already immunocompromised patients. Aspergillus pneumonia, disseminated herpes simplex, and bacterial sepsis complicate steroid use.
Approach:
- Optimize topical therapy: High-potency corticosteroids, calcineurin inhibitors (tacrolimus, pimecrolimus)
- Dupilumab (IL-4/IL-13 inhibitor): Emerging data suggest efficacy for eczema in HIES with acceptable safety
- If systemic steroids unavoidable: Lowest dose, shortest duration, ensure antimicrobial prophylaxis, high vigilance for infections
Management Principles
Antimicrobial Prophylaxis:
- Trimethoprim-sulfamethoxazole: Daily dosing reduces staphylococcal infections
- Antifungal prophylaxis: Itraconazole or posaconazole for patients with pneumatoceles
- Some centers use continuous or pulsed penicillinase-resistant penicillins
Hack #2: The Bleach Bath Protocol
Dilute bleach baths (0.005% sodium hypochlorite, approximately ½ cup household bleach per full bathtub) twice weekly reduce skin colonization with S. aureus and decrease infection frequency. Mechanism: Direct antimicrobial effect plus anti-inflammatory properties. Simple, inexpensive, effective.
Immunoglobulin Replacement: Not routinely indicated (specific antibody responses typically intact), but consider in patients with documented antibody deficiency or recurrent severe infections despite prophylaxis.
Interferon-Gamma: Some patients benefit from 50 μg/m² SC three times weekly, though evidence is limited compared to CGD.
Hematopoietic Stem Cell Transplantation: The only curative therapy. Indicated for:
- DOCK8 deficiency (excellent outcomes with HLA-matched donors)
- Severe AD-HIES with life-threatening infections (more controversial, outcomes mixed)
- Best performed before extensive lung disease, severe infections, or malignancy develops
Monitoring:
- Annual echocardiography (coronary aneurysms)
- Brain MRI/MRA baseline and if neurologic symptoms (aneurysms, lacunar infarcts)
- Bone density screening
- Chest CT every 2-3 years (assess pneumatoceles, bronchiectasis)
Key References:
- Freeman AF, et al. The hyper-IgE syndromes. Immunol Allergy Clin North Am. 2008;28(2):277-291.
- Grimbacher B, et al. Hyper-IgE syndrome with recurrent infections--an autosomal dominant multisystem disorder. N Engl J Med. 1999;340(9):692-702.
- Szczawinska-Poplonyk A, et al. Hyper-IgE syndromes--clinical manifestation diversity in primary immune deficiency. Orphanet J Rare Dis. 2011;6:76.
When to Suspect a PID in an Adult: The 10 Warning Signs
The Jeffrey Modell Foundation and American Red Cross established 10 warning signs suggesting PID. While designed for pediatric screening, these apply to adults with modifications:
The 10 Warning Signs (Adult-Adapted):
Four or more new ear infections within one year
- Adult consideration: Chronic otitis media or sinusitis in adults warrants investigation
Two or more serious sinus infections within one year
- Adult consideration: Recurrent bacterial sinusitis requiring multiple antibiotic courses, especially requiring IV therapy or hospitalization
Two or more months on antibiotics with little effect
- Critical care pearl: Patients requiring months of continuous antimicrobials for persistent infections or slow resolution
Two or more pneumonias within one year
- Red flag: Recurrent bacterial pneumonia, especially with same organism or encapsulated bacteria
- High risk: Pneumonia with unusual organisms (Pneumocystis jirovecii, Aspergillus, Burkholderia)
Failure of an infant to gain weight or grow normally
- Adult equivalent: Chronic diarrhea with malabsorption, failure to thrive, or unexplained weight loss
Recurrent, deep skin or organ abscesses
- Classic for: CGD (catalase-positive organisms), HIES (cold staphylococcal abscesses)
- Adult consideration: Hepatosplenic or perianal abscesses, particularly if recurrent
Persistent thrush or fungal infection on skin
- Adult manifestation: Chronic mucocutaneous candidiasis, recurrent esophageal candidiasis (without HIV)
- Consider: AIRE deficiency (autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy)
Need for intravenous antibiotics to clear infections
- Critical care relevance: Infections requiring prolonged IV therapy, slow response to appropriate antimicrobials
Two or more deep-seated infections including septicemia
- High-risk organisms: Unusual pathogens (Serratia, Burkholderia, Chromobacterium), fungal sepsis
- Pattern recognition: Recurrent episodes of sepsis, particularly with different organisms
A family history of PID
- Inheritance patterns: X-linked (CGD, X-SCID, XLA), autosomal recessive (many PIDs), autosomal dominant (STAT3)
- Take detailed pedigree: Infant deaths, recurrent infections, consanguinity, autoimmunity, lymphoma
Pearl #4: Additional "Red Flags" for Adult-Onset PID
Beyond the classic 10 warning signs, intensivists should consider PID when encountering:
- Autoimmune manifestations: Cytopenias, inflammatory arthritis, vasculitis, granulomas (CVID, CTLA4 haploinsufficiency, STAT3 GOF)
- Bronchiectasis in young adults: Particularly non-CF bronchiectasis with recurrent exacerbations
- Unexplained granulomatous disease: Lung, liver, spleen, lymph nodes (CVID, CGD, STAT1 GOF)
- Lymphoproliferative disease or lymphoma at young age: Especially in context of recurrent infections
- Severe or disseminated viral infections: Varicella, herpes zoster, EBV, CMV (T-cell defects, DOCK8)
- Opportunistic infections without HIV: PCP, Cryptococcus, disseminated MAC, Aspergillus
- Invasive fungal infections in non-neutropenic hosts: Think CGD or CARD9 deficiency
- BCG dissemination after vaccination: Mendelian susceptibility to mycobacterial disease (MSMD)
- Adverse vaccine reactions: Disseminated BCG or vaccine-strain viral infections
- Recurrent septic shock with common organisms: May indicate subtle antibody or complement deficiencies
Diagnostic Approach in the ICU
When PID is suspected, initiate workup while managing acute illness:
First-Tier Investigations:
Complete blood count with differential
- Lymphopenia: <1500/μL suggests T-cell or combined immunodeficiency
- Neutropenia or neutrophilia
- Thrombocytopenia (autoimmune or consumptive)
- Eosinophilia (Hyper-IgE syndrome, DOCK8 deficiency)
Immunoglobulin quantification
- IgG, IgA, IgM, IgE
- Interpret with age-appropriate reference ranges
- Low IgG + low IgA/IgM suggests CVID
- Isolated IgA deficiency (most common PID, usually asymptomatic)
- Markedly elevated IgE (>2000 IU/mL): Hyper-IgE syndrome, DOCK8 deficiency, Omenn syndrome
Vaccine-specific antibodies
- Tetanus, diphtheria, pneumococcal serotypes (pre- and post-vaccination)
- Protective responses: Tetanus ≥0.15 IU/mL, ≥70% pneumococcal serotypes with ≥1.3 μg/mL
- Absent responses indicate functional antibody deficiency despite normal Ig levels
Lymphocyte subset enumeration (flow cytometry)
- CD3+ (total T cells): 700-2100/μL
- CD4+ (helper T cells): 300-1400/μL
- CD8+ (cytotoxic T cells): 200-900/μL
- CD19+ or CD20+ (B cells): 80-616/μL
- CD16+56+ (NK cells): 90-600/μL
Second-Tier Investigations (Immunology consultation-guided):
Complement assessment
- CH50 (classical pathway): Detects C1-C9 deficiencies
- AH50 (alternative pathway): Detects factor B, D, properdin deficiencies
- C3, C4 levels
- Consider for recurrent Neisseria infections
Neutrophil function
- DHR flow cytometry for CGD
- NBT test if DHR unavailable
- Myeloperoxidase deficiency screening (if suggestive history)
T-cell function
- Proliferation assays to mitogens (PHA, ConA) and antigens (tetanus, candida)
- Delayed hypersensitivity skin testing (candida, tetanus, mumps)
Genetic testing
- Targeted gene panel for suspected diagnosis (STAT3, CYBB, NCF1, etc.)
- Whole exome sequencing for undefined presentations
- Essential for definitive diagnosis, genetic counseling, and HSCT planning
Hack #3: The "Sepsis Workup Plus PID Screen"
For critically ill patients with suggestive features, order the PID screening panel concurrently with sepsis workup. Obtain blood samples BEFORE administering blood products or IVIG, as these confound immunoglobulin interpretation. A simple panel ordered at admission (CBC with diff, Ig levels, lymphocyte subsets) costs relatively little but may provide crucial diagnostic information.
Special Considerations for Specific Presentations
Recurrent pneumonia algorithm:
- Bronchoscopy with BAL (cultures, galactomannan, PCR panels)
- High-resolution chest CT (bronchiectasis, pneumatoceles, nodules)
- PFTs with DLCO (restrictive patterns in GLILD)
- Consider CVID (check Ig levels, vaccine responses), HIES (IgE, eosinophils), or CGD (DHR)
Unusual organism algorithm:
- Burkholderia cepacia: CGD (check DHR)
- Serratia marcescens: CGD
- Aspergillus in non-neutropenic host: CGD or HIES (check DHR, IgE)
- Pneumocystis jirovecii without HIV: CD4+ T-cell deficiency (check lymphocyte subsets, CD4 count)
- Disseminated NTM or BCG: MSMD (check IFN-γ pathway: IL-12, IL-12R, IFN-γR, STAT1)
- Neisseria meningitidis (recurrent): Complement deficiency (CH50, AH50), properdin deficiency
- Chronic mucocutaneous candidiasis: AIRE deficiency, STAT1 GOF, IL-17 pathway defects
Key References:
- Bonilla FA, et al. Practice parameter for the diagnosis and management of primary immunodeficiency. J Allergy Clin Immunol. 2015;136(5):1186-1205.
- Bousfiha A, et al. Human inborn errors of immunity: 2019 update on the classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol. 2020;40(1):24-64.
The Role of IVIG and Hematopoietic Stem Cell Transplant
Intravenous Immunoglobulin (IVIG) Therapy
Mechanism of Action
IVIG provides passive humoral immunity through multiple mechanisms:
- Antibody replacement: Provides opsonizing antibodies against encapsulated bacteria and other pathogens
- Immunomodulation: Fc receptor blockade, complement modulation, anti-idiotypic antibody networks
- Anti-inflammatory effects: Modulation of cytokine production, T-cell and B-cell regulation
- Neutralization: Bacterial toxins, viral particles, autoantibodies
Indications in Primary Immunodeficiency
Established indications:
Antibody deficiency disorders:
- CVID
- X-linked agammaglobulinemia (XLA)
- Specific antibody deficiency with recurrent infections
- Hyper-IgM syndrome
- Good syndrome (thymoma with immunodeficiency)
Combined immunodeficiencies with antibody deficiency:
- Wiskott-Aldrich syndrome
- Ataxia-telangiectasia
- DiGeorge syndrome (if antibody deficient)
Adjunctive therapy:
- Autoimmune complications of PID (cytopenias, inflammatory disease)
- Severe infections in PID patients already on replacement therapy
Controversial/investigational:
- CGD (antibody responses usually intact; use for specific indications)
- HIES (specific antibody responses typically normal; reserve for documented deficiency)
Dosing Strategies
Replacement therapy (antibody deficiency):
- Standard dosing: 400-600 mg/kg IV every 3-4 weeks
- Target trough: IgG >500-600 mg/dL (some patients require >800 mg/dL)
- Individualization: Dose and interval adjusted based on:
- Trough IgG levels
- Infection frequency and severity
- Bronchiectasis presence
- Patient weight and volume distribution
- IgG catabolism rate (varies between individuals)
High-dose therapy (immunomodulation):
- Loading dose: 1-2 g/kg over 2-5 days
- Indications: Severe infections with sepsis, autoimmune cytopenias, severe inflammatory complications
Subcutaneous immunoglobulin (SCIG):
- Dose: Weekly dosing at ~1.5× monthly IVIG dose (divided by 4)
- Advantages: Steady-state IgG levels, fewer systemic reactions, home administration, improved quality of life
- Disadvantages: Injection site reactions, patient burden, not suitable for acute/critical illness
- Conversion: IVIG 400 mg/kg monthly ≈ SCIG 150 mg/kg weekly
Administration in Critical Care
Premedication considerations:
- Acetaminophen 650-1000 mg PO/IV
- Diphenhydramine 25-50 mg PO/IV
- Consider corticosteroids (hydrocortisone 50-100 mg IV) for patients with prior severe reactions
Infusion protocols:
- Start slowly: 0.5-1 mg/kg/min for first 30 minutes
- Gradually increase: Double rate every 15-30 minutes if tolerated
- Maximum rate: 4-8 mg/kg/min (varies by product)
- Monitor: Vital signs every 15 minutes during infusion
Pearl #5: Product Selection Matters
IVIG products differ in IgA content, osmolality, sugar content, and stabilizers. For critically ill patients:
- IgA-deficient patients with anti-IgA antibodies: Use IgA-depleted products (Gammaked, Gammagard Liquid, Bivigam)
- Renal dysfunction: Avoid sucrose-containing products (increased osmotic nephrosis risk); prefer L-proline stabilized products
- Diabetes: Avoid maltose-containing products (interfere with glucose monitoring)
- Thrombosis risk: Consider lower osmolality products, ensure adequate hydration
Adverse Effects and Management
Immediate reactions (during/within hours):
- Infusion-related reactions: Headache, chills, myalgias, fever, nausea
- Management: Slow infusion rate, premedication, switch products
- Anaphylaxis: Rare, associated with IgA deficiency and anti-IgA antibodies
- Management: Stop infusion, epinephrine, supportive care, use IgA-depleted product subsequently
- Hypotension: Volume shifts, cytokine release
- Management: Slow infusion, volume support
Delayed reactions (days to weeks):
Aseptic meningitis: Severe headache, photophobia, meningismus, CSF pleocytosis
- Risk factors: High-dose IVIG, migraine history, prior episodes
- Management: Supportive care, NSAIDs, avoid/reduce dose in future
- Mechanism: Meningeal inflammation from immune complex deposition
Hemolytic anemia: Anti-A/anti-B isoagglutinins in IVIG attack recipient RBCs
- Risk factors: Non-O blood type, high-dose IVIG, repeated doses
- Monitoring: CBC, haptoglobin, LDH, indirect bilirubin, DAT
- Management: Transfusion if severe, switch products with lower isoagglutinin titers
Thromboembolism: DVT, PE, stroke, MI
- Risk factors: Age >65, obesity, immobility, hypercoagulable states, high-dose IVIG
- Incidence: 1-5% (higher in critically ill)
- Mechanism: Increased viscosity, platelet activation, factor XI activation
- Prevention: Hydration, mobilization, consider prophylactic anticoagulation in high-risk patients
Acute kidney injury: Osmotic nephrosis, ATN
- Risk factors: Pre-existing renal disease, diabetes, age >65, volume depletion, sucrose-containing products
- Prevention: Avoid sucrose products, ensure euvolemia, monitor creatinine
- Management: Supportive care, may require dialysis
Transfusion-related acute lung injury (TRALI): Rare, respiratory distress within 6 hours
- Management: Supportive care, mechanical ventilation if needed
Oyster #4: The "Pseudohyponatremia" of IVIG
High-dose IVIG increases serum protein and viscosity, causing falsely low sodium measurements by indirect ion-selective electrode methods (most common lab assays). True serum osmolality and direct sodium measurement remain normal. Clinicians may inappropriately treat this pseudohyponatremia, causing true hypernatremia. Always consider IVIG timing when interpreting electrolytes post-infusion.
Monitoring During IVIG Therapy
Pre-infusion:
- IgG trough level (before each dose)
- Baseline CBC, creatinine, LFTs
- Urinalysis (for proteinuria)
Post-infusion (especially high-dose):
- CBC at 48-72 hours (hemolysis screening)
- Creatinine at 24-48 hours
- Clinical assessment for thrombosis (if symptomatic, obtain appropriate imaging)
Long-term monitoring:
- IgG troughs every 3-6 months once stable
- Annual comprehensive metabolic panel
- Infection diary (frequency, severity, antibiotic requirements)
- Pulmonary function tests annually (if bronchiectasis or lung disease)
Hematopoietic Stem Cell Transplantation (HSCT)
Indications
HSCT offers curative potential for many PIDs by replacing defective immune cells. Timing and donor selection critically impact outcomes.
Strong indications (high mortality without HSCT):
- Severe combined immunodeficiency (SCID)
- Wiskott-Aldrich syndrome
- X-linked lymphoproliferative disease (XLP)
- Hemophagocytic lymphohistiocytosis (HLH)
- DOCK8 deficiency
- X-linked chronic granulomatous disease (debated, especially if recurrent life-threatening infections)
- Hyper-IgM syndrome (CD40L deficiency)
Conditional indications (case-by-case basis):
- CVID with severe complications (refractory cytopenias, GLILD, malignancy)
- Autosomal recessive CGD with severe phenotype
- AD-HIES with life-threatening infections (controversial)
- CTLA4 haploinsufficiency with severe disease
- STAT1 gain-of-function with severe infections
Generally not indicated:
- Well-controlled antibody deficiencies on IVIG
- Mild CGD phenotypes responsive to prophylaxis
- Complement deficiencies (no stem cell defect)
Donor Selection and Conditioning
Donor hierarchy:
- HLA-matched sibling donor (MSD): Best outcomes, lowest GVHD
- Matched unrelated donor (MUD) 10/10 or 9/10: Excellent outcomes with modern immunosuppression
- Haploidentical donor: Parents or siblings, requires T-cell depletion or post-transplant cyclophosphamide
- Umbilical cord blood: Option when no matched donor available
Conditioning intensity:
- Myeloablative conditioning (MAC): Busulfan-based, cyclophosphamide
- Higher toxicity but better engraftment and immune reconstitution
- Preferred for SCID, WAS, XLP, HLH
- Reduced-intensity conditioning (RIC): Fludarabine-based, low-dose busulfan or treosulfan
- Lower toxicity, reduced late effects
- Preferred for older patients, organ dysfunction, some PIDs where mixed chimerism acceptable
- Non-myeloablative conditioning: Minimal conditioning
- Experimental in PIDs
Special considerations:
- Pre-transplant infections: Aggressively treat and achieve source control before HSCT
- Organ dysfunction: Particularly lung disease (CGD, HIES, CVID with bronchiectasis) increases transplant-related mortality
- HLA antibodies: Screen in multiply-transfused patients; may require desensitization
- Age: Outcomes better when performed in childhood, but adults can undergo HSCT successfully
Outcomes and Complications
Success rates (vary by disease and era):
- SCID with MSD: >95% survival
- SCID with MUD: 70-90% survival
- DOCK8 deficiency: 70-85% survival (excellent outcomes)
- CGD: 60-80% survival (variable by center and era)
- WAS: 85-90% survival
Early complications (<100 days):
- Infections: Bacterial, viral (CMV, adenovirus, HHV-6), fungal (Aspergillus, Candida, Pneumocystis)
- GVHD (acute): Skin, GI tract, liver
- Engraftment failure: Primary or secondary graft rejection
- Veno-occlusive disease (VOD/SOS): Hepatic sinusoidal injury
- Transplant-associated thrombotic microangiopathy (TA-TMA)
Late complications (>100 days):
- Chronic GVHD: Multi-organ involvement, may require prolonged immunosuppression
- Immune reconstitution: T cells recover first (3-6 months), B cells later (6-12 months)
- Infectious complications: Persistent viral infections (CMV, EBV), PCP until immune recovery
- Endocrine dysfunction: Thyroid disease, growth hormone deficiency, gonadal failure
- Secondary malignancies: EBV-associated PTLD, skin cancers, others
Hack #4: Gene Therapy—The Emerging Alternative
Gene therapy has achieved remarkable success for X-linked SCID, ADA-SCID, and X-linked CGD in clinical trials. Autologous CD34+ cells are transduced with lentiviral vectors carrying functional genes, then reinfused after conditioning. Advantages include no GVHD risk and no donor requirement. Limitations include cost, availability (few centers), and potential insertional mutagenesis (mitigated by modern vectors). Several gene therapy products have received regulatory approval internationally, with more in development.
Current FDA-approved gene therapies for PIDs:
- None yet approved in U.S. (as of early 2025), but several in late-stage trials
- European Medicines Agency has approved therapies for ADA-SCID
Emerging targets:
- X-linked CGD (early clinical trial data promising)
- Wiskott-Aldrich syndrome (clinical trials ongoing)
- X-linked hyper-IgM syndrome
Post-HSCT Management in ICU
Infection prophylaxis (continue until immune reconstitution):
- Antibacterial: Fluoroquinolone or trimethoprim-sulfamethoxazole
- Antifungal: Fluconazole or posaconazole (Aspergillus coverage if high risk)
- Antiviral: Acyclovir for HSV/VZV
- PCP prophylaxis: Trimethoprim-sulfamethoxazole or atovaquone/pentamidine
CMV management:
- Weekly PCR surveillance
- Pre-emptive therapy (valganciclovir or ganciclovir) if viremia detected
- CMV disease requires IV ganciclovir ± foscarnet, IVIG
GVHD treatment:
- First-line: High-dose corticosteroids (methylprednisolone 2 mg/kg/day)
- Steroid-refractory: Ruxolitinib, extracorporeal photopheresis, anti-TNF agents, others
Immune reconstitution monitoring:
- Lymphocyte subsets monthly initially
- Mitogen responses, immunoglobulin levels
- Vaccine-specific antibodies (after reconstitution)
Key References:
- Perez EE, et al. Update on the use of immunoglobulin in human disease: A review of evidence. J Allergy Clin Immunol. 2017;139(3S):S1-S46.
- Gennery AR. Hematopoietic stem cell transplantation in primary immunodeficiencies. Hematol Oncol Clin North Am. 2011;25(1):127-149.
- Kohn DB, et al. Autologous ex vivo lentiviral gene therapy for adenosine deaminase deficiency. N Engl J Med.2021;384(21):2002-2013.
Critical Care Pearls: Practical Approach to PID Patients
Pre-ICU Optimization (When Possible)
For elective procedures or anticipated ICU admission:
- Optimize IVIG dosing: Ensure trough >600-800 mg/dL
- Update antimicrobial prophylaxis
- Ensure vaccinations current (inactivated vaccines only)
- Pulmonary optimization: Bronchodilators, airway clearance, treat bronchiectasis exacerbations
- Nutritional assessment and support
- Screen for latent infections: Fungal, mycobacterial, viral
Infection Management Principles
1. Early, aggressive antimicrobial therapy:
- Broader spectrum than typical empiric regimens
- Cover unusual organisms based on PID type
- Prolonged courses (often weeks to months)
- Therapeutic drug monitoring for antimicrobials and antifungals
2. Source control:
- Early surgical consultation for abscesses, necrotic tissue
- CGD abscesses often require drainage despite antimicrobials
- Consider interventional radiology for deep-seated collections
3. Adjunctive therapies:
- Optimize IVIG levels during acute infection
- Consider granulocyte transfusions for CGD with life-threatening fungal infections (controversial, limited availability)
- Interferon-gamma for CGD and severe mycobacterial infections
- G-CSF for neutropenic complications (limited role in most PIDs)
Respiratory Failure Management
Ventilation strategies:
- Lung-protective ventilation (tidal volume 6-8 mL/kg IBW, plateau pressure <30 cm H₂O)
- HIES patients: Minimize positive pressure (risk of pneumatocele rupture)
- Consider early tracheostomy if prolonged ventilation anticipated (many PID patients have difficult airways due to structural abnormalities)
ARDS in PID:
- May result from infection, TRALI, or inflammatory complications
- Standard ARDSNet protocols apply
- Treat underlying cause aggressively
- Prone positioning, ECMO per usual indications
Bronchoscopy:
- Essential for diagnosis in new infiltrates
- BAL: Cultures (bacterial, fungal, mycobacterial, viral), galactomannan, β-D-glucan, PCR panels
- Transbronchial biopsy if safe (assess for GLILD, lymphoma, organizing pneumonia)
Hemodynamic Management
Septic shock:
- Standard resuscitation principles apply
- Early appropriate antimicrobials critical
- Consider high-dose IVIG (1-2 g/kg) in antibody-deficient patients with sepsis
- Vasopressor choice: No specific PID-related modifications
- Corticosteroids: Hydrocortisone per septic shock protocols, but be vigilant for fungal superinfection
Cardiomyopathy:
- Viral myocarditis more common (enterovirus in CVID, coxsackievirus)
- Coronary aneurysms in HIES may cause ACS
- Standard heart failure management
Hematologic Complications
Autoimmune cytopenias:
- High-dose IVIG (1-2 g/kg)
- Corticosteroids: Prednisone 1 mg/kg or equivalent
- Rituximab: 375 mg/m² weekly × 4 (for ITP, AIHA, or Evans syndrome)
- Refractory cases: Consider thrombopoietin receptor agonists (ITP), splenectomy (last resort)
Thrombosis:
- Higher incidence in PID (inflammation, IVIG, infections)
- Standard anticoagulation unless contraindicated
- Prophylactic anticoagulation for high-risk patients receiving IVIG
Nutritional Support
- Early enteral nutrition when feasible
- Many PID patients have baseline malabsorption (CVID enteropathy, chronic diarrhea)
- Ensure adequate protein (1.5-2 g/kg) for immune function and healing
- Micronutrient supplementation: Vitamin D (many PIDs have deficiency), zinc, selenium
- Consider parenteral nutrition if enteral feeding not tolerated
Family Communication and Goals of Care
Prognostic discussions:
- PID patients often survive severe illnesses with aggressive care
- Discuss realistic goals: Some complications (e.g., refractory GLILD, progressive CGD lung disease) may be irreversible
- HSCT remains curative option for many, even after severe ICU illness
Genetic counseling:
- Offer to family members when diagnosis made
- Discuss inheritance patterns, carrier testing, prenatal diagnosis
- Sibling screening may identify affected individuals pre-symptomatically
Future Directions and Research Horizons
Novel Therapeutics
1. Targeted biologics:
- JAK inhibitors (tofacitinib, ruxolitinib): STAT1 GOF, CTLA4 haploinsufficiency, steroid-refractory GVHD
- IL-1 blockade (anakinra): CGD hyperinflammation, autoinflammatory complications
- Dupilumab: HIES eczema, atopic manifestations of DOCK8 deficiency
- BTK inhibitors: Autoimmune complications of CVID
2. Advanced gene therapies:
- CRISPR/Cas9 gene editing: Precise correction of mutations in patient cells
- In vivo gene therapy: Delivery of therapeutic genes directly to patients without ex vivo manipulation
- Base editing and prime editing: Next-generation precise gene correction
3. Newborn screening expansion:
- T-cell receptor excision circles (TRECs): Identifies SCID and severe T-cell lymphopenia
- Kappa-deleting recombination excision circles (KRECs): Identifies B-cell lymphopenia
- Implementation in many countries enables pre-symptomatic diagnosis and improved outcomes
Precision Medicine Approaches
Pharmacogenomics:
- Individualized conditioning regimens for HSCT based on genetic variants
- Antimicrobial dosing optimization using patient-specific pharmacokinetics
Biomarkers:
- Predicting GLILD development in CVID (cytokine profiles, B-cell subsets)
- Identifying patients at high risk for lymphoma (clonal expansions, EBV load)
- CGD hyperinflammation markers (IL-18, S100 proteins)
Registry Studies and Real-World Evidence
International registries (USIDNET, ESID registry) collecting longitudinal data enable:
- Natural history studies
- Genotype-phenotype correlations
- Treatment outcome assessments
- Rare complication identification
Conclusion
Primary immunodeficiencies in adults present unique challenges in critical care settings. The intensivist must maintain diagnostic vigilance for these "zebras," as delayed recognition increases morbidity and mortality. Key principles include:
- Suspect PID in patients with recurrent infections, unusual organisms, paradoxical inflammatory complications, or autoimmunity
- Obtain appropriate screening tests early (CBC, Ig levels, lymphocyte subsets) before blood products confound results
- Provide aggressive antimicrobial therapy tailored to the specific PID, covering unusual pathogens
- Optimize immunoglobulin replacement in antibody-deficient patients
- Balance infection risk with immunomodulation when treating inflammatory complications
- Pursue source control aggressively for abscesses and deep-seated infections
- Involve multidisciplinary team (immunology, infectious diseases, surgery, HSCT) early
- Consider curative therapies (HSCT, gene therapy) even after severe ICU illness
The expanding field of clinical immunology continues to elucidate genetic mechanisms, enabling targeted therapies and curative interventions. As gene therapy and novel biologics evolve, outcomes for PID patients will continue to improve. The critical care physician plays a vital role in managing life-threatening complications, bridging patients to definitive therapy, and improving survival in these complex disorders.
Summary: Clinical Pearls and Oysters
Pearls:
- CVID phenotyping predicts complications: "infection-only" phenotype has best prognosis; GLILD and enteropathy significantly increase mortality
- Catalase-positive organisms (S. aureus, Burkholderia, Serratia, Aspergillus, Nocardia) specifically target CGD patients
- "Job" syndrome describes recurrent "cold" staphylococcal abscesses with minimal inflammation
- Additional red flags beyond the 10 warning signs: autoimmunity, bronchiectasis, granulomas, lymphoma, severe viral infections, opportunistic infections in non-HIV patients
- IVIG product selection matters: IgA-depleted for anti-IgA antibodies, avoid sucrose in renal disease, consider osmolality in thrombosis risk
Oysters (Diagnostic Dilemmas):
- GLILD vs. infection in CVID: Requires aggressive diagnostic workup, often both coexist; empiric antibiotics while pursuing diagnosis
- "Sterile" granulomas in CGD: Often represent incompletely cleared infection; treat empirically for fungi, consider immunomodulation cautiously
- Corticosteroids in HIES: Tempting for severe eczema but dramatically increase infection risk; optimize topicals, consider dupilumab
- IVIG pseudohyponatremia: Falsely low sodium from increased protein/viscosity; avoid inappropriate treatment
Hacks:
- IVIG in CVID septic shock: Consider aggressive loading (1-2 g/kg) alongside standard sepsis management
- Bleach bath protocol: 0.005% sodium hypochlorite twice weekly reduces S. aureus colonization in HIES
- "Sepsis workup plus PID screen": Order CBC with diff, Ig levels, lymphocyte subsets at admission BEFORE blood products
- Gene therapy: Emerging curative alternative to HSCT without GVHD risk; approved for some PIDs internationally
Recommended Reading
Comprehensive Reviews:
- Tangye SG, et al. Human inborn errors of immunity: 2022 update on the classification from the International Union of Immunological Societies Expert Committee. J Clin Immunol. 2022;42(7):1473-1507.
- Notarangelo LD. Primary immunodeficiencies. J Allergy Clin Immunol. 2010;125(2 Suppl 2):S182-194.
Guidelines:
- Bonilla FA, et al. Practice parameter for the diagnosis and management of primary immunodeficiency. J Allergy Clin Immunol. 2015;136(5):1186-1205.
- Seidel MG, et al. The European Society for Immunodeficiencies (ESID) Registry Working Definitions for the Clinical Diagnosis of Inborn Errors of Immunity. J Allergy Clin Immunol Pract. 2019;7(6):1763-1770.
Online Resources:
- International Union of Immunological Societies Expert Committee: https://iuis.org
- Jeffrey Modell Foundation: https://www.info4pi.org
- Immune Deficiency Foundation: https://primaryimmune.org
- European Society for Immunodeficiencies Registry: https://esid.org
Acknowledgments: The author acknowledges the contributions of immunologists, intensivists, and infectious disease specialists whose clinical insights inform this review.
Conflicts of Interest: None declared.
Funding: None.
No comments:
Post a Comment