PID Guide, version 1.0, 6 August 2014

Primary diseases (PIDs): diagnostic & treatment considerations for patients managed at Red Cross War Memorial Children’s Hospital (RCWMCH)

Lead author: Brian Eley: [email protected]

Contributors: Marc Hendricks, Alan Davidson, Harsha Lochan, Terry Schlaphoff, Shireen Maart

Posted: 7 August 2014

Summary

This guideline describes the management of PIDs at RCWMCH. Diagnosis is constrained by a limited repertoire of tests offered through the National Health Laboratory Services (NHLS). Therapeutic options include haematopoietic stem cell transplantation in patients with severe immunodeficiency such as SCID.

1 | P a g e

PID Guide, version 1.0, 6 August 2014

TABLE OF CONTENTS

I. ABBREVIATIONS Page 3

II. ESTABLISHING A DIAGNOSIS Page 4

A. Clinical considerations Page 4 B. Laboratory considerations Page 8 1. HIV Page 8 2. Undifferentiated recurrent infection Page 8 3. Clinical presentation suggestive of a particular PID category Page 9 4. Chromosomal & genetic testing Page 12 C. Immunological consultation & investigation at RCWMCH Page 12

III. PATIENT RECORDS & PID DATABASE Page 13

IV. PREVENTION OF & TREATMENT OF PIDS Page 14

A. Immunisation practice Page 14 1. Routine immunization of children with PIDs Page 14 2. Household contacts Page 15 B. Antimicrobial prophylaxis Page 15 C. Additional measures to control infections Page 16 D. Immunoglobulin replacement therapy Page 17 1. Management of intravenous IRT Page 17 2. Monthly IVIG infusions at RCWMCH Page 18 E. Haematopoietic stem cell transplantation Page 18 1. Identification of a suitable donor Page 19 2. Tissue typing procedure Page 20 3. Pre-transplantation management Page 21 4. Transplantation & post-transplantation management Page 22 F. Other therapeutic modalities Page 23

V. ONGOING CARE Page 23

VI. REFERENCES Page 25

VII. APPENDICES Page 26

2 | P a g e

PID Guide, version 1.0, 6 August 2014

I. ABBREVIATIONS

BCG Bacillus Calmette Guérin CGD Chronic granulomatous disease CMV Cytomegalovirus CVID Common variable immunodeficiency DTap-IPV/Hib Diphtheria, tetanus, acellular pertussis, inactivated polio vaccine, Haemophilus influenza type B EBV Epstein-Barr virus FMF Familial Mediterranean Fever GvHD Graft versus host disease HBV Hepatitis B vaccine HIV Human immunodeficiency virus HPV Human papillomavirus vaccine HSCT Haematopoietic stem cell transplantation ID Infectious diseases INH Isoniazid IRT Immunoglobulin replacement therapy IVIG Intravenous immunoglobulin LTI Laboratory for Tissue MMR Measles, mumps, rubella vaccine NHLS National Health Laboratory Services OPV Oral polio vaccine PCV Pneumococcal conjugate vaccine PIDs diseases RAG Recombination activation gene RCWMCH Red Cross War Memorial Children’s Hospital RV Rotavirus vaccine SABMR South African Bone Marrow Registry SCID Severe combined immunodeficiency SLE Systemic lupus erythematosus TB Tuberculosis Td Tetanus and reduced amount of diphtheria vaccine TMP-SMX Trimethoprim-sulphamethoxazole VZIG Varicella-zoster immunoglobulin WHIM syndrome Warts, hypogammaglobulinaemia, infections, myelokathexis syndrome XLA X-linked agammaglobulinaemia

3 | P a g e

PID Guide, version 1.0, 6 August 2014

II. ESTABLISHING A DIAGNOSIS

A. Clinical considerations

In the current international classification more than 200 specific PIDs are listed spanning nine major categories. These nine categories and disease examples within each category are included in table 1. For the complete international classification refer to reference 1. Because of limited diagnostic capacity only a small proportion of these PIDs can be diagnosed in South Africa. However, through international collaboration it may possible to confirm the diagnosis of a subset of patients by mutational analysis. This section focuses on the testing capability in our country.

Table 1: The nine major categories of PIDs with specific disease examples

PID category Specific diseases (common examples) Combined (T and ) Severe combined immunodeficiency (SCID) Combined immunodeficiencies with Ataxia-telangiectasia associated or syndromic features DiGeorge syndrome Wiskott-Aldrich syndrome Hyper-IgE syndromes Predominantly deficiencies X-linked agammaglobulinaemia Common variable immunodeficiency Hyper-IgM syndromes IgA deficiency IgG subclass deficiency Specific antibody deficiency with normal immunoglobulin concentrations Transient hypogammaglobulinaemia of infancy Diseases of immune dysregulation Chediak-Higashi syndrome Familial haemophagocytic lymphohistiocytosis Lymphoproliferative syndromes e.g. X-linked lymphoproliferative syndrome Defects of phagocytic number, function, Severe congenital neutropaenia or both Cyclic neutropaenia Leukocyte adhesion deficiency Shwachman-Diamond syndrome Chronic granulomatous disease IL-12p40 deficiency IFN- receptor 1 deficiency GATA2 deficiency Defects in innate immunity IRAK-4 and MyD88 deficiencies WHIM syndrome TLR3 and UNC93B1 deficiencies Chronic mucocutaneous candidiasis Autoinflammatory disorders Familial Mediterranean fever Hyper-IgD syndrome Muckle-Wells syndrome Familial cold autoinflammatory syndrome Complement deficiencies Deficiencies of components of the classic or alternate complement pathway e.g. C5 or C6 deficiency C1 inhibitor deficiency Phenocopies of PID (a)Phenocopies associated with somatic mutations e.g. Autoimmune lymphoproliferative syndrome (ALPS-SFAG) (b) Phenocopies associated with autoantibodies Chronic mucocutaneous candidiasis due to IL-17

4 | P a g e

PID Guide, version 1.0, 6 August 2014

and/or IL-22 autoantibody Adult-onset immunodeficiency due to INF-gamma autoantibody Pulmonary alveolar proteinosis due to GM-CSF autoantibody

A comprehensive clinical history and examination may identify socioeconomic factors, which increase susceptibility to infection, determine the pattern of infections, exclude secondary immunodeficiencies (Table 2), allergies and the crèche syndrome and direct the investigation of non-immunological causes of recurrent infection.

Table 2: Common secondary immunodeficiencies

. Severe acute malnutrition . Vitamin A deficiency . HIV infection . Measles . Immunosuppresive therapy . High-dose glucocorticosteroid administration . Nephrotic syndrome . Exposure to aero-pollutants

Clinical evaluation may assist with the identification of specific PIDs such as combined immunodeficiencies associated with syndromic features and may direct the selection of appropriate laboratory investigations (Table 3).

Table 3: Clinical features that may assist in the diagnosis of PIDs2

Infection history

. Recurrent bacterial infection that is more frequent than expected for the patient’s age . More than one severe infection, e.g. meningitis, osteomyelitis, pneumonia, sepsis . Infections that present atypically, are unusually severe or chronic, or fail regular treatment, especially if intravenous antibiotics are needed . Abscess of an internal organ e.g. liver abscess . Recurrent subcutaneous abscesses in children . Prolonged or recurrent diarrhoea . Any infection caused by an unexpected or opportunistic pathogen e.g. pneumonia caused by pneumocystis jerovecii in an HIV-unexposed child . Severe or long-lasting warts, generalized molluscum contagiosum infection . Extensive candidiasis, recurrent oral thrush in children >1 year . Complications of vaccination such as disseminated BCG or varicella infection, paralytic polio or rotavirus infection . Absence of evidence of secondary immunodeficiencies

Family history

. PID in the family; familial occurrence of similar symptoms or recurrent

5 | P a g e

PID Guide, version 1.0, 6 August 2014

infection (affected males related by the female line, or another clear pattern of inheritance) . Unexplained early infant deaths, deaths due to infection . Consanguinity in the (grand) parents (known or suspected) . or haematological malignancy in several family members . It is essential to construct a detailed family tree may suggest the Mendelian inheritance pattern and direct identification of the genetic defect.

Other clinical features that could point to PID

. Aplasia or hypoplasia of thymus (X-ray) . . Auto-immune disease, especially auto-immune cytopenias, SLE, non-specific arthritides . Bleeding tendency . Congenital cardiac anomalies, mainly conotruncal defects . Chronic diarrhoea, malabsorption, pancreatic insufficiency . Delayed separation of umbilical cord ( >4 weeks) . Delayed shedding of primary teeth . Developmental delay (progressive) . Difficult-to-treat obstructive lung disease . Eczema, dermatitis (severe, atypical) . Failure to thrive . Graft-versus-host reaction after blood transfusion, or mother-to-child (infant) engraftment . Granulomas . Haemolysis . to sunlight . Hypocalcaemic seizures . Inflammatory bowel disease (atypical) . Malignancy (mainly lymphoma) . Non-allergic oedema . Poor wound healing; scarring . Recurrent fever . Rib or other skeletal anomalies (X-ray) . Thymoma . Unexplained bronchiectasis, pneumatoceles, interstitial lung disease . Vasculitis

Physical examination

. Skin and appendages: abnormal hair or teeth; eczema; neonatal erythroderma; (partial) albinism; pale skin; incontinentia pigmenti; nail dystrophy; extensive warts or molluscae; congenital alopecia; vitiligo; petechiae (early onset, chronic); cold abscesses; telangiectasia; absence of sweating

6 | P a g e

PID Guide, version 1.0, 6 August 2014

. Oral cavity: gingivostomatitis (severe); periodontitis; aphthae (recurrent); giant oral ulcers; thrush; dental crowding; conical incisors; enamel hypoplasia; persistent deciduous teeth . Eyes: retinal lesions; telangiectasia . Lymphoid tissue: absence of lymph nodes and tonsils; lymphadenopathy (excessive); asplenia; organomegaly (liver, spleen) . Neurological: ataxia; microcephaly; macrocephaly . Other: angioedema (without urticaria); digital clubbing; dysmorphism; stunted growth or disproportional growth

The PIDs usually manifest with one of eight clinical syndromes, defined in Table 4.

Table 4: Clinical presentations and their associated PIDS (adapted from reference 2)

Recurrent ENT and respiratory tract infection and/or bronchiectasis Most frequent PID category: Predominantly antibody deficiencies Other PID categories associated with this presentation:- - Combined immunodeficiencies - Combined immunodeficiencies with syndromic features - Defects of phagocyte number and/or function - Defects of innate immunity - Complement deficiencies Failure to thrive during infancy associated with intractable diarrhoea and severe eczema Most frequent PID category: Combined immunodeficiencies Other PID categories associated with this presentation:- - Combined immunodeficiencies with syndromic features - Defects of phagocyte number and/or function - Diseases of immune dysregulation - Defects of innate immunity Recurrent pyogenic infection with or without poor wound healing Most frequent PID category: Defects of phagocyte number and/or function Other PID categories associated with this presentation:- - Combined immunodeficiencies with syndromic features - Defects of innate immunity - Complement deficiencies Unusual infections or unusually severe clinical course of infections PID categories associated with this presentation:- - Combined immunodeficiencies - Combined immunodeficiencies with syndromic features - Diseases of immune dysregulation - Defects in innate immunity Recurrent infection caused by the same pathogen PID categories associated with this presentation:- - Combined immunodeficiencies with syndromic features - Defects of phagocyte number and/or function - Defects in innate immunity - Complement deficiencies Autoimmune or chronic inflammatory disease; or lymphoproliferative disorder Most frequent PID category:- diseases of immune dysregulation Other PID categories associated with this presentation:- - Combined immunodeficiencies - Predominantly antibody deficiencies - Combined immunodeficiencies with syndromic features - Defects of phagocyte number and/or function - Autoinflammatory disorders - Complement deficiencies

7 | P a g e

PID Guide, version 1.0, 6 August 2014

Characteristic combinations of clinical features (eponymous syndromes) Specific diseases within most PID categories may be associated with this presentation Angioedema PID associated with this presentation: C1 inhibitor deficiency

Furthermore, the spectrum of infections that a patient experiences may on occasion indicate the most likely PID category

Table 5: spectrum of infections

. Predominantly antibody deficiencies: encapsulated bacteria e.g. S. pneumoniae, H. influenzae, mycoplasma, chronic enterovirus infections . Cellular / combined immunodeficiencies: bacterial e.g. S. pneumoniae, H. influenza, S. aureus, viral e.g. CMV, fungal eg. Candida species, Histoplasmosis sp, and protozoal e.g. Pneumocystis jiroveci infections . Defects of phagocyte number and/or function: catalase positive bacterial infections e.g. S. aureus, Burkholderia cepacia, Serratia marcescens, Nocardia sp. E. coli, Ps. Aeruginosa, M. tuberculosis (particularly in high prevalence settings), Nontuberculous mycobacterial and BCG and fungal infections e.g. Aspergillus fumigatus . Early complement deficiencies: encapsulated bacterial infections . Terminal C deficiencies (C5-C9) & : recurrent neisserial infections

B. Laboratory considerations

1. HIV infection

HIV infection is a significant problem in South Africa and may mimic many of the presentations of PIDs. It should be excluded before requesting other, expensive immunological investigations.

2. Undifferentiated recurrent infection

Some patients with PIDs present with undifferentiated recurrent infection i.e. without a pattern of infections that is suggestive of a particular category of PIDs. These children should be subjected to a general immunological screen. This may be performed in a structured manner, guided by an experienced clinician or clinical immunologist. Table 6 provides a structured approach to the selection of immunological tests in these patients. General immunological screening should assist in the diagnosis of many PIDs (Tables 6 & 7).

Table 6: A structured approach immunological testing in a patient with undifferentiated recurrent infection, adapted from an approach of the Jeffrey Modell Foundation3

Step 1 History, Physical examination, Mass and height Exclude HIV Infection Full blood count and differential count Quantitative immunoglobulin levels (IgA, IgM, IgG, IgE)

Step 2 Specific antibody production (tetanus and/or diphtheria, response to pneumococcal vaccine (pre- and post- titres, production of isohaemagglutinins 8 | P a g e

PID Guide, version 1.0, 6 August 2014

IgG subclass analysis

Step 3 Lymphocyte subsets (CD3, CD4, CD8, CD19, CD16/56) Complement screen (CH50 or CH100) oxidation burst (if indicated) Lymphocyte proliferation studies (using mitogen and antigen stimulation)

Step 4 Advanced testing guided by an experienced clinician e.g. specific complement component screen including mutational screening for common C5 & C6 mutations Blood specimen collection from the index patient and immediate family members for mutational analysis together with completed consent forms for each individual providing specimens (discuss with duty ID senior registrar or consultant to arrange DNA extraction, storage and liaison with an external laboratory)

Table 7: Immunological test may aid the diagnosis of many PIDs

Immunological test PIDs Total lymphocyte count SCID Eosinophil count Neutrophil count Congenital neutropaenias Leukocyte adhesion deficiency () morphology Chediak-Higashi syndrome Platelet count & size Wiskott-Aldrich syndrome (small platelets, thrombocytopaenia) Immunoglobulins (IgM, IgA, IgG) XLA, CVID, IgA deficiency, hyper-IgM syndromes, transient hypogammaglobulinaemia of infancy, combined T and B cell immunodeficiencies IgE Hyper-IgE syndromes, combined T and B cell immunodeficiencies IgG subclasses Isolated IgG subclass deficiency, IgA with IgG subclass deficiency Specific antibody production Specific antibody deficiency with normal immunoglobulin concentrations Lymphocyte subsets XLA, autosomal recessive conditions causing B- hypogammaglobulinaemia, combined T and B cell immunodeficiencies including SCID Lymphocyte proliferative responses Combined T and B cell immunodeficiencies Chronic mucocutaneous candidiasis (absent / reduced proliferation following stimulation with Candida antigen) Complement screen (CH50 or CH100) Deficiencies in the classic complement pathway (C1 to C9) Alternative complement pathway screen Deficiencies of properdin, factor B, factor D, (AH50 or AH100) [available in the private factor I (leading to secondary C3 deficiency), sector] C3 and C5 to C9 Neutrophil oxidative burst test Chronic granulomatous disease

3. Clinical presentation suggestive of a particular PID category4,5

Alternatively, the clinical presentation may suggest a particular PID category or a specific deficiency in which case a more focused investigative approach may be adopted as follows:

9 | P a g e

PID Guide, version 1.0, 6 August 2014

Suspected combined (T and B cell) immunodeficiency: Analysis of white cell count and differential is an important first step in patients with suspected cellular deficiency. Total lymphocyte count in an infant persistently <3.0 x 109/L is predictive of SCID, and therefore an indication for urgent lymphocyte subset analysis. However, using low lymphocyte count will not identify patients with Omenn syndrome who have normal or increased T-cell numbers due to oligoclonal expansion of T-cells. Circulating T-cells may also be identified in patients with SCID because of maternal T-cell engraftment. The next steps in the investigation of suspected cellular deficiency includes measurement of lymphocyte subsets and in vitro functional testing (lymphocyte proliferative studies). Lymphocyte subset analysis in infants suspected of having SCID helps to confirm the diagnosis and identify the potential genetic defect. A simplified classification of SCID based on the presence or absence of T-cells, B-cells and natural killer (NK)-cells is summarised in Table 8.

Table 8: Interpretation of lymphocyte subset results: Classification of severe combined immunodeficiency (SCID)

Immunological phenotype Disease Inheritance T-B+ SCID NK- Common γ-chain deficiency XL Janus kinase-3 deficiency AR

NK+ IL-7 receptor α-chain AR deficiency AR CD45 deficiency AR CD3/CD3/CD3 deficiency AR Coronin-1A deficiency T-B- SCID NK- Adenosine deaminase AR deficiency AR Reticular dysgenesis NK+ RAG-1 / RAG-2 deficiency AR DCLRE1C (Artemis) AR deficiency AR Adenosine deaminase deficiency AR DNA PKcs deficiency AR Cerunnos deficiency

RAG=recombination activation gene, DNA PKcs deficiency = DNA-dependant protein kinase Catalytic subunit deficiency, DCLRE1C deficiency = DNA cross-link repair 1C deficiency

Suspected predominantly antibody deficiencies: Initial screening includes measurement of IgG, IGM and IgA concentrations, and a comparison of these results with normal age-related reference ranges. Quantitation of IgG subclass concentration is most useful in the evaluation of patients with IgA deficiency with recurrent bacterial infection. Measurement of specific antibody production is useful for confirming defective antibody production, and is essential when total immunoglobulin concentrations are only moderately reduced or normal in patients with recurrent bacterial infection. The easiest method is the evaluation of spontaneously produced anti-blood group (isohaemagglutinins) and specific antibodies generated in response to routine childhood immunisation. Isohaemagglutinin titres may be measured at local blood banks. Titres  1 in 8 to A1 and B cells are usually present in normal individuals. Assessing in vivo antibody production involves immunising a patient with protein antigen (e.g. tetanus toxoid or diphtheria toxoid) and

10 | P a g e

PID Guide, version 1.0, 6 August 2014

polysaccharide antigen (23-valent pneumococcal polysaccharide vaccine) and measuring pre-immunisation and 3-4 week post-immunisation antibody titres. In subjects with normal antibody production, a 4-fold increase in antibody concentration occurs. Responses to protein antigens fall within the IgG1 subclass. Response to polysaccharide antigens is located within the IgG2 subclass. Conjugate polysaccharide vaccines are not helpful in the functional evaluation of an IgG2 subclass deficiency or a selective polysaccharide antibody deficiency because the responses to conjugate vaccines fall within the IgG1 and IgG3 subclasses. Determining the presence or absence of B-cells in patients with pan- hypogammaglobulinaemia is important to identify patients with absent B-cells (B- hypogammaglobulinaemia), caused by arrest in early B-cell development. Approximately 85% of patients with this phenotype have XLA (formerly called Bruton agammaglobulinaemia). The remaining patients with this phenotype have one of several autosomal recessive gene defects. Advanced immunological testing requires specialised assays such as the detection of CD40 ligand expression by flow cytometry in patients with an immunoglobulin pattern consistent with CD40 ligand deficiency, and naïve and memory B-cell subset analysis for characterising patients with CVID. Few specialised investigations are currently available in South Africa. Table 9 provides guidance for diagnosing common antibody deficiencies.

Table 9: Interpretation of immunoglobulin results: Major types of predominantly antibody deficiencies

Disease B cells Immunoglobulins X-linked Absent /low Low IgG, IgA, IgM agammaglobulinaemia CVID > 1% Low IgG, IgA Normal / low IgM

Hyper-IgM syndromes Normal Normal / raised IgM Low IgG, IgA

Isolated IgG subclass Normal Normal deficiency

Selective IgA deficiency Normal Low IgA Normal IgG, IgM

Specific antibody deficiency Normal Normal

CVID: Common variable immunodeficiency

Suspected defects of phagocyte number or function: Screening starts with a review of the absolute neutrophil count and morphology. The diagnosis of cyclical neutropaenia requires serial measurement of the neutrophil count, two to three times per week for at least 4-6 weeks. A diagnosis of severe congenital neutropaenia is suggested with neutrophil counts persistently below 0.5 x 109/L. Bone marrow biopsy is required to exclude a malignancy, another cause of neutropaenia or maturational arrest typical of severe congenital neutropaenia. In patients with LAD-1, mild to moderate neutrophilia is usually present in the absence of infection, and during acute infection marked granulocytosis with neutrophil counts reaching above 100 x 109/L may be documented. In LAD-1 the absent expression of the adhesion molecules CD11 and CD18 may be established by flow cytometric assay. An abnormal neutrophil oxidative burst test occurs in CGD. Evaluation of and phagocytosis requires specialised investigations, which are not widely available.

11 | P a g e

PID Guide, version 1.0, 6 August 2014

Suspected complement deficiencies: The best screening test for deficiencies in the classical complement pathway is the total haemolytic complement assay (CH50 or CH100). Deficiencies in the alternative complement pathway can be assessed using the AH50 assay. A decreased AH50 result suggests deficiency of factor B, factor D or properidin. Whereas decrease in both CH50 and AH50 results suggest a deficiency of one of the shared complement components, C3 to C9. A C1 esterase inhibitor assay is used to screen for enzyme deficiency, and the CH50 assay will be abnormal in C2 deficiency.

Hyper-IgE and Hyper-IgD syndromes: A consistent feature of Job Syndrome is an elevated IgE concentration, typically >2000 IU/mL. Hyper-IgD Syndrome is caused by mutations in the mevalonate kinase gene. Approximately 80% of these patients have elevated plasma IgD concentrations. In the remaining 20% of patients, the IgD level is normal. However, other autoinflammatory disorders, such as FMF may be associated with an elevated IgD concentration.

4. Chromosomal and genetic testing

Chromosomal analysis is important in children with suspected DiGeorge syndrome to determine the presence of 22q11.2 deletion and can be arranged through the Department of Human Genetics, UCT.

Genetic analysis is required for a definitive diagnosis of most PIDs, but is not routinely available in South Africa. Genetic testing may occasionally be arranged through an externally based laboratory. Before genetic analysis can be arranged EDTA-preserved specimens are required, usually form the index patient and the parents. DNA extraction is currently performed in the 4th floor ICH laboratory by special arrangement, consult with Professor Eley. A consent form (Appendix 1) must be completed on every individual submitting a specimen for genetic analysis. The original forms should be given to Professor Eley for safe-keeping.

C. Immunological consultation & investigations at RCWMCH

Several sub-specialist services contribute to the management of conditions listed in the latest international classification of PIDs.1 For example, the haematology/oncology service (Professor A Davidson, Dr M Hendricks, Dr A van Eyssen) manages children with neutropaenic syndromes, some children with Wiskott-Aldrich syndrome, familial haematophagocytic lymphohistiocytosis, children with PIDs manifesting with oncological diseases and children with PIDs following HSCT. The rheumatology service directed by Dr C Scott manages all patients with autoinflammatory diseases. The Neurology service (Professor J Wilmshurst, Dr A Ndondo) manages children with Ataxia-telangiectasia. The cardiology service (Dr R de Dekker) manages children with cardiogenetic conditions such as DiGeorge syndrome. The pulmonology service (Dr M Zampoli, Dr A Vanker) manages children with pulmonary alveolar proteinosis. The Allergy service (Professor M Levin) manages children with C1 inhibitor deficiency and Professor Potter based in the Lung Institute, University of Cape Town currently manages terminal complement deficiencies. Adult patients with primary immunodeficiencies are managed by Professor Stanley Ress / Dr Jonny Peter at Groote Schuur Hospital.

12 | P a g e

PID Guide, version 1.0, 6 August 2014

Role of the infectious diseases service: The infectious diseases service traditionally manages many children with PIDs who manifest with infectious disease problems as described in table 4. The duty senior registrar and consultant for the infectious diseases service (contact through the RCWMCH telephone exchange: 021-6585111) share clinical responsibilities for immunology. Appointments for outpatient consultation can be arranged with the duty senior registrar or consultant – immunology patients are seen on Monday mornings in A11, refer section V. Ongoing care on page 22 for more details about outpatient activities.

Immunological investigation: Immune function tests are expensive. Therefore, requests for immunological investigation must be discussed with the duty senior registrar or Prof Eley. Thereafter, specific tests may be arranged with the NHLS chemical pathology service on extension: 5226. Specimens should ideally be sent to Specimen Reception before 09h30. The specimens are transported to Tygerberg Hospital where the tests are performed. Results are obtainable from the Chemical Pathology laboratory, RCWMCH or through the NHLS DISA website.

Table 10: Specimen requirements

Test name Specimen requirements Immunoglobulin concentrations 2-3 ml clotted tube IgG subclass concentrations 2-3 ml clotted tube Specific IgG titres 2-3 ml clotted blood Lymphocyte subset analysis 2-3 ml EDTA blood Oxidative burst test 2-3 ml heparinised blood CH50 (Total haemolytic complement) 2-3 ml clotted blood on ice Lymphocyte proliferative tests 2-3 ml EDTA blood Leucocyte adhesion markers 2-3 ml EDTA blood

DNA extraction for mutational analysis: Generally 2-5 ml EDTA-preserved blood per subject is sufficient for DNA extraction. Blood specimens should be collected from the index patient and immediate family members together with completed consent forms for each individual providing a specimen. Discuss with ID senior registrar or consultant to arrange DNA extraction and storage, and liaison with an external laboratory.

(2) Role of the Haematology / Oncology service: Some of the conditions listed in the latest international PID classification are managed with haematopoeitic stem cell transplant (HSCT). Once the need for HSCT has been established blood for HLA tissue typing should be sent from the patient and potential donors as a matter of urgency. Please discuss with the haematology/oncology consultant on call and the South African Bone Marrow Registry (see page 19).

III. PATIENT RECORDS & PID DATABASE

The ID service maintains a database of all children diagnosed or managed with PIDs at RCWMCH. Once a patient has been diagnosed with a PID, a datasheet (appendix 2) should be completed, usually by the ID senior registrar in collaboration with the referring doctor. The completed data sheets should be reviewed at the ad hoc Tuesday lunchtime Immunology meeting. Once the initial entry is complete, the data sheets should be given to Spasina King (extension number: 5821) who will enter the data into the electronic database. Datasheets may be accessed by clinicians

13 | P a g e

PID Guide, version 1.0, 6 August 2014

attending to PID patients. Furthermore, therapeutic updates, complications and a change to the outcome should be entered on the data sheet when necessary.

IV. PREVENTION OF INFECTION &TREATMENT OF PIDs

Several strategies of preventing infection and treating children with PIDs including routine immunization practice, antimicrobial prophylaxis, immunoglobulin replacement therapy (IRT) and haematopoietic stem cell transplantation (HSCT).

A. Immunisation practice

1. Routine immunisation of children with PIDs

Currently, The South African EPI schedule recommends that the following vaccines be administered to all children.

Table 11: South African EPI schedule

Age of child Recommended vaccines At birth BCG, OPV (0) 6 weeks OPV (1), PCV (1), HBV (1), DTaP-IPV/Hib (1), RV (1) 10 weeks HBV (2), DTaP-IPV/Hib (1), RV (2) 14 weeks PCV (2), HBV (3), DTaP-IPV/Hib (3), RV (2) 9 months PCV (3), Measles vaccine (1) 18 months DTaP-IPV/Hib (4), Measles vaccine (2) 6 years Td (1) 9-12 years HPV x 2 doses, 6 months apart 12 years Td (2)

The EPI programme also makes provision for the administration of annual inactivated influenza vaccine to high-risk patient groups.6 In the private sector, MMR, Varicella, Hepatitis A vaccines and additional doses of inactivated pertussis-containing vaccine is recommended in older children.

Immunisation with live viral or bacterial vaccines is known concern for individuals with serious PIDs of T-cell, B-cell and phagocytic cell origin. Recommendations for immunizing patients with PIDs have been formulated by the American Academy of Pediatrics and are summarized in table 12.

Table 12: Immunisation of children with PIDs7,8

Category/conditions Vaccines Comments contraindicated Severe antibody OPV, smallpox, Effectiveness of any vaccine is uncertain if it deficiencies e.g. XLA & LAIV, Yellow fever, depends only on humoral immunity e.g. PPSV23, CVID most live bacterial MPSV4; IVIG interferes with measles and possibly vaccines; consider varicella responses; Efficacy of pneumococcal measles; there is no vaccination is not documented in severe antibody data for varicella or deficiency; Consider measles & varicella vaccines rotavirus vaccines Little vaccine-related viral infection is seen with CVID Less severe antibody OPV, other live All vaccines probably effective; immune responses deficiencies e.g. IgA or vaccines appear to may be attenuated IgG subclass deficiency be safe Complete, cell-mediated All live vaccines All vaccines are probably ineffective; immune deficiencies e.g. SCID, response might be attenuated; pneumococcal and complete DiGeorge Hib vaccines are recommended syndrome

14 | P a g e

PID Guide, version 1.0, 6 August 2014

Partial defects e.g. most Selected live Effectiveness of any vaccine depends on degree of patients with DiGeorge vaccines: BCG, immune suppression; pneumococcal, Hib and syndrome, Wiskott- Ty21a S. typhi meningococcal vaccines are recommended; Aldrich syndrome, Ataxia vaccine, LAIV, consider Hib vaccine if not administered in infancy telangiectasia MMR, varicella, NB: Weight of evidence does not support strict herpes zoster, OPV, avoidance of all live vaccines, particularly when the yellow fever, CD4 count is >500 cells/mm3 smallpox & rotavirus Complement deficiencies None All routine vaccines are probably effective; pneumococcal and meningococcal vaccines recommended Phagocytic disorders Live-bacterial All inactivated vaccines and probably all live-virus e.g. CGD, leukocyte vaccines i.e. BCG & vaccines are safe & effective adhesion defects, Ty21a S. typhi myeloperoxidase vaccine deficiency Defects affecting the Live mycobacterial There are no reported live attenuated viral vaccine- interferon-gamma – IL- vaccines, currently induced infection, but caution is urged 12 pathway only BCG There is very few data on live vaccines other than BCG

Re-immunisation following HSCT is essential and should be directed by the transplant centre. This is addressed in section E, Haematopoietic stem cell transplantation and Appendix 3.

2. Household contacts7,8

. Immunocompetent close contacts of patients with compromised immunity should not receive smallpox vaccine or OPV because these vaccine viruses may be transmitted to immunocompromised individuals.

. Close contacts can receive other standard vaccines including MMR, rotavirus vaccine, varicella vaccine and measles vaccine because these pose little risk of infection to the immunocompromised individual.

. Varicella vaccine is recommended for susceptible contacts of immunocompromised patients because transmission of vaccine virus from healthy individuals is rare. No post-immunisation precautions are required unless the vaccine recipient develops a vesicular exanthema – in this situation isolation of the immunocompromised patient and administration of VZIG is recommended, and the household contact should be treated with acyclovir.

. Annual immunization with inactivated influenza vaccine is recommended for all household contacts from 6 months of age onwards

. Scheduled periodic immunization of household contacts with pertussis vaccine (Tdap), pneumococcal vaccine, measles, mumps and rubella vaccine should proceed.

B. Antimicrobial prophylaxis: Several PIDs may benefit from antimicrobial prophylaxis, including those with concomitant bronchiectasis (Table 13). In milder conditions (e.g. IgA deficiency and transient hypogammaglobulinaemia of infancy) recurrent infections may occasionally require short courses of prophylactic antibiotics. Antimicrobial prophylaxis may be added in patients with severe antibody

15 | P a g e

PID Guide, version 1.0, 6 August 2014

deficiencies, if IRT is unable to adequately control the frequency of recurrent infections.

Table 13: Oral antimicrobial prophylaxis for children with PIDs9

Disease Regimen Severe combined TMP-SMX 5mg/kg (of TMP) once daily immunodeficiency Fluconazole 3mg/kg once daily Acyclovir 20mg/kg/dose, four times daily

Common variable TMP-SMX 5mg/kg (of TMP) once daily, 3 days/week immunodeficiency where IRT is inadequate X-linked agammaglobulinaemia TMP-SMX 5mg/kg (of TMP) once daily, 3 days/week where IRT is inadequate DiGeorge syndrome TMP-SMX 5mg/kg (of TMP) once daily, 3 days/week Fluconazole 3mg/kg once daily

Wiskott-Aldrich syndrome TMP-SMX 5mg/kg (of TMP) once daily, 3 days/week Fluconazole 3mg/kg once daily Acyclovir 20mg/kg/dose, four times daily Phenoxymethylpenicillin if splenectomy i.e. 125 mg twice daily (<5 years) or 250mg twice daily (>5 years)

Ataxia-telangiectasia Azithromycin 10 mg/kg/day, 3 days/week

Job syndrome TMP-SMX 5mg/kg (of TMP) once daily or Flucloxacillin 125-250mg twice daily If pneumatocoeles are present add Itraconazole 100mg daily (< 13 years or < 50kg) or 200mg daily (≥13 years or ≥50kg)

Chronic granulomatous disease TMP-SMX 5mg/kg (of TMP) once daily Itraconazole 100mg daily (< 13 years or < 50kg) or 200mg daily (≥13 years or ≥50kg)

Terminal complement (e.g. C5 or Phenoxymethylpenicillin125 mg twice daily (<5 years) or C6) deficiencies 250mg twice daily (>5 years)

TMP/SMX = Trimethoprim-sulphamethoxazole

C. Additional measures to control infections

General advice aimed at reducing the risk of infection will benefit many immunodeficient patients and should include advice on personal, hand and dental hygiene, avoidance of exposure to individuals with active infections and the provision of annual influenza immunisation for household contacts.

Furthermore, antimicrobial prophylaxis is often administered with other interventions to reduce the risk of infection.

. In SCID, prior to a HSCT, the patient should ideally be managed in a laminar flow facility, with full contact precautions, and receive sterile feeds. In the immediate post-HSCT period similar management interventions apply.

16 | P a g e

PID Guide, version 1.0, 6 August 2014

. In CGD, life-long interferon-γ 50 μg/m3 three times per week is recommended in combination with cotrimoxazole and itraconazole prophylaxis. However, high cost limits its use in South Africa.

. Job Syndrome, colonization and infection by Staphylocccus aureus may be prevented or controlled by regular bleach or chlorhexidine soaks i.e. bleach 120 ml per bath tub or chlorhexidine soaks three times per week and intermittent muporocin applications i.e. twice daily nasal applications for 5-10 days, repeat this prescription intermittently

. In addition to penicillin prophylaxis, meningococcal immunisation is recommended for patients with terminal complement deficiencies.

D. Immunoglobulin Replacement Therapy (IRT)10-12: Many antibody deficiencies and combined deficiencies, in which quantitative or qualitative IgG deficiency occurs may benefit from IRT (Table 11).

Table 14: Immunodeficiencies that may benefit from IRT

Predominantly antibody deficiencies X-linked agammaglobulinaemia Common variable immunodeficiency Hyper-IgM syndromes IgG subclass deficiency with/without IgA deficiency Specific antibody deficiency with normal immunoglobulin concentration (selected cases) Transient hypogammaglobulinaemia of infancy (selected cases)

Combined T and B cell immunodeficiencies Severe combined immunodeficiency Wiskott-Aldrich syndrome Ataxia-telangiecatia with IgG or IgG subclass deficiency X-linked lymphoproliferative syndrome

1. Management of intravenous IRT

. Lifelong IRT is required for severe, genetically confirmed deficiencies, including conditions in which arrest in early B-cell development is demonstrated. . Incompletely defined predominantly antibody deficiencies characterised by the presence of circulating B-cells and subnormal IgG concentration may require IRT for 1-5 years followed by re-evaluation of serum immunoglobulin concentrations since hypogammaglobulinaemia may be transient and resolve spontaneously. If IgG deficiency persists IRT should be restarted and may have to be continued indefinitely if it fails to resolve. . IRT is not indicated for IgA Deficiency. . Treatment with intravenous immunoglobulin (IVIG) is most widely used. Subcutaneous immunoglobulin administration is less frequently used but equally effective. . Intravenous immunoglobulin is administered at 3-4 week intervals at the usual starting dose of 400 mg per kg for patients without chronic lung disease and 600 mg per kg for those with bronchiectasis.

17 | P a g e

PID Guide, version 1.0, 6 August 2014

. The mean half-life of IgG is 18-25 days. After achieving steady state, an IgG trough concentration of >5g/L reduces the infection rate and trough levels up to 10g/L will progressively reduce the risk of pneumonia. Progressive adjustment of the total infusion dose is required to optimise the IgG trough concentration. . In some children, IRT does not adequately control the underlying infectious disease risk and routine cotrimoxazole prophylaxis may be added (refer below, section B: antibiotic prophylaxis) . When commencing an IVIG infusion, start at a rate of 0.01-0.02 ml/kg/minute and if this is tolerated, gradually increase the rate to 0.1 ml/kg/minute. Infusions should be given under the same supervision and monitoring conditions that are required for blood transfusions. . Adverse events: Anaphylactic reactions are extremely rare but have been described in IgA-deficient patients with circulating anti-IgA antibodies. Most IVIG reactions are non-anaphylactoid, typically manifesting with nausea, chills, headache, backache, malaise, fever, pruritis, and/or tingling. Reactions occur in 5-15% of patients receiving IVIG and are mainly related to the rate of infusion. Slowing or stopping the infusion for 15-30 minutes will reverse most reactions. Recurring side effects can be modified or prevented by administering aspirin (15 mg/kg/dose, orally) ibuprofen (5 mg/kg/dose, orally) or hydrocortisone (6 mg/kg/dose, IV, maximum 100 mg) one hour before an IVIG infusion. There is no risk of HIV or hepatitis B transmission. Some immunoglobulin preparations have been associated with outbreaks of non-A, non-B hepatitis, including hepatitis C. . In children receiving long-term IVIG, alanine aminotransferase (ALT) levels should be monitored at 3-6 monthly intervals to detect subclinical transmitted hepatitis.

2. Monthly IVIG infusions at RCWMCH: Intravenous immunoglobulin infusions are administered in the 'Day room' in ward E2, usually on Friday mornings. To make a booking contact the ward sister, extension: 5053 or 5153. All children managed through the immunology service should be discussed with a senior registrar or immunology consultant to ensure continuity of care.

E. Haematopoietic stem cell transplantation (HSCT)

HSCT is the treatment of choice of severe immunodeficiencies (Table 13).

Table 13: Indications for HSCT12

Severe combined immunodeficiency (SCID): refer table 8 for the spectrum of SCID diseases

Other combined immunodeficiencies (CID) including Omenn’s syndrome, Adenosine deaminase deficiency, purine nucleoside phosphorylase deficiency, MCH class II deficiency, CD40 ligand deficiency & CD40 deficiency

Combined immunodeficiencies with associated or syndromic features such as Wiskott-Aldrich syndrome & DiGeorge syndrome

Phagocytic disorders including Kostmann , severe congenital neutropaenia, chronic granulomatous disease, leukocyte adhesion deficiency type I,

18 | P a g e

PID Guide, version 1.0, 6 August 2014

& IFN-gamma receptor deficiency type I

Diseases of immune dysregulation such as X-linked lymphoproliferative syndrome, familial haemophagocytic lymphohistiocytosis, Chediak-Hugashi syndrome & Griselli syndrome

1. Identification of a suitable donor, e.g. sibling, haplo-identical parent / sibling, other related donor e.g. cousin, matched unrelated donor:

Patients who may benefit from HSCT should be discussed with immunology service or haematology/oncology service at the earliest opportunity. Once there is agreement that a child may benefit from HSCT i.e. agreement by the haematology/oncology service at RCWMCH, the referral doctor and the parents, a suitable donor should be sought.

Please note: Tissue typing should be arranged in consultation with the haematology/oncology service at RCWMCH. This is done through NHLS Laboratory for Tissue Immunology (LTI) in Cape Town.

First tissue type all siblings to determine whether a matched related donor can be identified. The chance of finding a suitable HLA-identical donor among the siblings ranges from 15-20%.

If there are no siblings or if there is no suitable sibling donor blood specimens may be requested from the parents to determine whether they would be suitable HLA- nonidentical donors. Testing of the extended family to identify a potential cousin donor could also be considered.

If a matched unrelated donor (MUD) is considered, the South African Bone Marrow Registry (SABMR) can be approached to search for a suitable donor. To request a MUD search, the Preliminary Search Request Form (Appendix 4) must be submitted to the SABMR together with the HLA typing of the patient and any family members who were HLA typed. Please note: The SABMR and LTI are separate entities. The SABMR has a purely administrative role but works very closely with LTI as the laboratory of choice for HLA typing for the matched unrelated donor programme. This is because of its international accreditation.

The patient must have been HLA-A, -B and -DRB1 DNA typed for a basic MUD search report to be generated.

Based on the preliminary search results, it is the treating physician’s decision whether the search proceeds to active testing of donors (“search activation”).

Before search activation, the patient should ideally be HLA typed at High Resolution at the HLA-A, -B, -C, -DRB1 and -DQB1 loci.

The SABMR will, at any stage of the search, guide the treating physician / transplant centre regarding the next steps, including additional testing of patient and potential donors.

The search for a suitable donor could be extended to International Bone Marrow registries if private funding or medical aid support is available.

19 | P a g e

PID Guide, version 1.0, 6 August 2014

Extent of the search and timelines

Ms Terry Schlaphoff or Ms Veronica Borrill at the SABMR can be contacted to arrange the donor search.

Tissue typing of unrelated donors may take at least 3-4 weeks to be completed. If the search for a suitable donor is extended beyond South Africa, more time may be required to find a suitable donor.

From preliminary search to transplantation with an unrelated donor, may take from 2 – 6 months.

South African Bone Marrow Registry Physical Address: Postal Address: J57 Old Outpatients Building SABMR Groote Schuur Hospital PO Box 13353 Observatory Mowbray Cape Town Cape Town 7925 7705

2. Tissue typing procedure

The following blood specimens are required from the patient: and potential donors for HLA typing:

. 1 x 10ml Clotted (Red Top) . 7 x 5ml EDTA (Purple Top)

. For paediatric patients, the volume of blood required will depend on the patient’s total nucleated cell count (TNC). Usually 5-10 ml of EDTA- preserved blood is required from a young child and about 15 ml of EDTA- preserved blood from the siblings. However, the TNC is used to determine the required blood volume to ensure that enough DNA is obtained from the blood specimen. Please contact the laboratory to discuss the optimal blood volumes.

Specimens together with the completed request form (Appendix 5) should be sent to:

NHLS Laboratory for Tissue Immunology C17 Specimen Reception Groote Schuur Hospital Laboratory contact number: 021-404 4502

All specimens must reach the laboratory within 24 hours of having been taken.

Specimens that are transported to the laboratory using a courier service must be properly packaged to avoid breakage while in transit.

Please store and transport the specimens at room temperature. (22°C)

Ensure that all tubes are correctly labelled and that a request form (Appendix 5) has been properly completed.

20 | P a g e

PID Guide, version 1.0, 6 August 2014

Please inform the laboratory telephonically when specimens are expected to arrive in the tissue immunology laboratory.

3. Pre-transplantation management12,13

Please note: The following recommendations focus on the pre-transplantation management of children with SCID and other severe combined immunodeficiencies. Children with other PIDs may require a different set of pre-transplantation requirements. This should be discussed with an immunologist or haematologist.

. Careful pre-transplantation management is required to optimize the success of HSCT.

. Avoid live vaccines, including BCG vaccine where possible.

. If the child has received neonatal BCG vaccination, then rule out disseminated BCG infection. Recent evidence suggests that 51% of children with SCID experiences BCG-related complications, 34% disseminated and 17% localized BCG complications.14

. Also rule out tuberculosis, and active CMV or Ebstein-Barr virus infection [CMV and EBV viral load tests]. Active CMV infection should be managed with intravenous gancyclovir / oral valgancyclovir followed by secondary valgancyclovir prophylaxis.

. If the patient develops lower respiratory tract infection screen the nasopharyngeal aspirate for respiratory viruses and PCP, and screen the patient for active CMV infection.

. Where possible treat all active infections aggressively with appropriate antimicrobial agents.

. Strict infection control practice is absolutely essential

o Ideally the patient should be isolated in a laminar flow facility (positive pressure ventilation). o Visitors should be restricted to the parents. If parents have active infections they should avoid patient contact. o Implement strict contact precautions and where possible use dedicated nursing and medical personnel on each shift. o Ensure that that medical & nursing personnel assigned to the patient do not have active infections. o Where possible institute a low germ diet (use sterile feeds)

. Commence prophylactic medication as soon as possible:

o TMP-SMX, fluconazole & acyclovir prophylaxis guidelines for SCID are described in table 12 o IVIG for children with SCID: use 1g/kg/week o TB post-exposure prophylaxis: INH prophylaxis according to standard dosing guidelines (10 mg/kg/day); Note that pre-exposure prophylaxis is not indicated

21 | P a g e

PID Guide, version 1.0, 6 August 2014

. Nutritional support: Occasionally, patients with severe failure to thrive may require total parenteral nutrition to improve their nutritional status prior to the HSCT

. Blood products, if required, should always be irradiated to prevent graft- versus-host disease, and to minimise CMV transmission blood products should be CMV-negative.

. The ID and haematology services at RCWMCH are available throughout the pre-transplantation period to support attending doctors

. Prior to transplantation the haematology service at RCWMCH would ideally counsel the parents regarding the HSCT and the post-transplantation course.

. Transfer to RCWMCH: Because of constant bed pressures patients from outside Cape Town and who are in the process of being worked-up for HSCT should not be transferred to RCWMCH unless a suitable donor has been identified. Thus the referring clinician must take responsibility for the early stages of pre-transplantation management using the above guidance.

. Pre-transplantation conditioning (myeloablative chemotherapy) may be indicated when there is residual T-lymphocyte immunity to prevent graft rejection and in unrelated transplants. Conditioning is usually not required for patients undergoing a matched sibling donor HSCT. However, myeloablative chemotherapy may be required prior to an unrelated HSCT.

4. Transplantation and post-transplantation care12, 13, 15

. Transplantation and post-HSCT management, including post-hospitalisation follow-up, are co-ordinated through the haematology service, RCWMCH.

. Prior to the transplantation central venous access is mandatory.

. Apart from umbilical cord HSCTs, donor stem cells are usually sourced from peripheral blood. Occasionally, stem cells are harvested from the bone marrow compartment by direct aspiration under anaesthetic.

. After processing the donor stem cells are infused through the central line into the peripheral blood compartment of the recipient.

. Haematopoietic engraftment usually occurs 14 to 28 days after the stem cell infusion. Consequently T-cell and B-cell immune reconstitution occurs. Post- transplantation graft-versus-host disease (GvHD) prophylactic cyclosporine and/or immunodepletion of stem cells may delay lymphocyte recovery by 3-6 months. Failure of B-cell reconstitution may occur resulting in the need for long-term post-transplantation IRT.

. Most patients require GvHD prophylaxis (cyclosporine) for at least 3 months post-HSCT. During this period ongoing antimicrobial prophylaxis plus IRT (or stabilized human serum infusions) are required. Many centres continue prophylaxis until CD4 counts are greater than 200 cells per µL and PHA proliferative responses of greater than 50% of normal control values are achieved.15

22 | P a g e

PID Guide, version 1.0, 6 August 2014

. Several post-HSCT problems may occur including:

o Immediate (within the 1st 30 days): neutropaenia and breakdown of mucocutaneous barrier (e.g. mucositis) may increase the risk for bacterial infection o Intermediate phase (30-100 days): immunosuppression, GvHD, fungal infections may develop due to colonization of invasive lines (Candida spp.) or inhalation of spores (Aspergillus spp.), the risk for PJP persists, there is a risk of nosocomial infection including RSV, influenza, parainfluenzae virus infection, there is a risk of reactivation of CMV or herpesvirus infection and post-HSCT lymphoproliferative disease (EBV-triggered). o Late-phase (>100 days): persistent humoral immune deficiencies may predispose to infection by encapsulated bacteria and there is an increased risk of Varicella-zoster infection o Non-infectious problems that may arise include chronic GvHD, incomplete immune reconstitution, hepatic veno-occlusive disease, endocrine effects including growth delay & thyroid dysfunction and neurodevelopmental delay

. Re-immunisation is delayed for 12 months in patients who’ve received an HLA-identical HSCT and for 18 months in patients who have received other HSCT types. Re-immunisation protocols are centre-specific and depend on the available vaccines. Appendix 3 describes the re-immunisation schedule currently employed at RCWMCH. In addition, household contacts should be immunized against influenza

F. Other therapies include cytokines such as myeloid haematopoietic growth factors for congenital and cyclical neutropaenia, enzyme replacement for adenosine deaminase deficiency and C1 esterase inhibitor deficiency, colchicine in FMF, and IL-1 blocking strategies in autoinflammatory disorders including patients with colchicine-resistant FMF.

V. ONGOING CARE

. Follow-up of patients managed by services other than the infectious Diseases service (refer section: C. Immunological consultation & investigation at RCWMCH, page 12) should be arranged through those services, contactable through the RCWMCH telephone exchange: 021-6595111.

. Immunology patients managed through the infectious diseases service are reviewed at an outpatient clinic conducted every Monday morning in A11. Appointments are made through the duty ID senior registrar or consultant by contacting either through the RCWMCH telephone exchange: 021-6585111. To re-book missed appointments contact the receptionist in A11: 021- 6585812.

. Most patients requiring regular follow-up and who live outside Cape Town are managed by their referring doctors with telephonic and/or email support from the duty senior registrar / consultant affiliated to the ID service at RCWMCH

23 | P a g e

PID Guide, version 1.0, 6 August 2014

. Regular intravenous immunoglobulin administration: For patients receiving regular IVIG infusions at RCWMCH, these infusions are administered in ward E2 day-care cubicle on Friday mornings. Bookings are arranged through the duty ID senior registrar.

24 | P a g e

PID Guide, version 1.0, 6 August 2014

VI. REFERENCES

1. Al-Herz W, Bousfiha A, Casanova JL, 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 Apr 22;5:162. doi: 10.3389/fimmu.2014.00162. 2. de Vries E; European Society for Immunodeficiencies (ESID) members. Patient- centred screening for primary immunodeficiency, a multi-stage diagnostic protocol designed for non-immunologists: 2011 update. Clin Exp Immunol. 2012 Jan;167(1):108-19. doi: 10.1111/j.1365-2249.2011.04461.x. 3. Jeffrey Modell Foundation. Four stages of PI testing. URL: http://www.info4pi.org/aboutPI/index.cfm?section=aboutPI&content=algorithm&CFID=4622460 &CFTOKEN=24c84ee4b884b5c5-DD5B5C15-9F85-5583-009F502D8518D735 4. Oliveira JB, Fleisher TA. Laboratory evaluation of primary immunodeficiencies. J Allergy Clin Immunol 2010 Feb;125(2 Suppl 2):S297-30;. doi: 10.1016/j.jaci.2009.08.043. 5. Bousfiha AA, Jeddane L, Ailal F, et al. A phenotypic approach for IUIS PID classification and diagnosis: guidelines for clinicians at the bedside. J Clin Immunol 2013 Aug;33(6):1078-87; doi: 10.1007/s10875-013-9901-6. 6. Walaza S. Recommendations pertaining to the use of viral vaccines: Influenza 2014. S Afr J Med 2014;104: 166-7. 7. American Academy of Pediatrics. Red Book, 29th Edition: 2012 Report of the Committee on Infectious Diseases 2012;74-81. 8. Shearer WT, Fleisher TA, et al. Recommendations for live viral and bacterial vaccines in immunodeficienct patients and their close relatives. J Allergy Clin Immunol 2014;133:961-6. 9. Papadopoulou-Alataki E1, Hassan A, Davies EG. Prevention of infection in children and adolescents with primary immunodeficiency disorders. Asian Pac J Allergy Immunol. 2012 Dec;30(4):249-58. 10. Orange JS1, 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 Oct;137(1):21-30. doi: 10.1016/j.clim.2010.06.012. 11. Wood P, Stanworth S, Burton J, Jones A, Peckham DG, Green T, Hyde C, Chapel H; UK Primary Immunodeficiency Network. Recognition, clinical diagnosis and management of patients with primary antibody deficiencies: a systematic review. Clin Exp Immunol. 2007 Sep;149(3):410-23. 12. García JM, Español T, Gurbindo MD, Casas C C. Update on the treatment of primary immunodeficiencies. Allergol Immunopathol (Madr). 2007 Sep-Oct;35(5):184- 92. 13. Davidson A, Novitzky N. Stem cell transplantation for primary immunodeficiency. Curr Allergy Clin Immunol 2012;25(4):205-8. 14. Marciano BE, Huang C-Y, Joshi G, et al. BCG vaccination in patients with severe combined immunodeficiency: complications, risks, and vaccination policies. J Allergy Clin Immunol 2014;133:1134-41. 15. Griffith LM, Cowan MJ, Notarangelo LD, et al. Improving cellular therapy for primary immune deficiency diseases: recognition, diagnosis, and management. J Allergy Clin Immunol 2009;124(6):1152-60.

25 | P a g e

PID Guide, version 1.0, 6 August 2014

Appendix 1: Refer separate pdf file for UCT request form for molecular studies, including consent for DNA analysis

26 | P a g e

PID Guide, version 1.0, 6 August 2014

Appendix 2: Immunology Data Collection Form (Cross War Memorial Children’s Hospital)

Biographical data (If available, use sticker to complete some of the data fields)

Name: Folder number:

Date of birth(Y-M-D): Date of testing:

Age (months):

Address:

Referral source

 State  Private  Outpatient  Inpatient, specify ward:

 Cape Town  Western Cape  Another province, specify:

Referring doctor:______Contact number:______

Indication(s) for investigation  Recurrent infection  Failure to thrive  Family history  Reaction to live- attenuated vaccines, specify:  Dysmorphic features, specify:  Other, specify: History of chief complaint (e.g. list major infections & dates, spectrum of infections, frequency of infections, associated hospitalisation):

Family History  Consanguity (Y / N)  Primary immunodeficiency, specify:

27 | P a g e

PID Guide, version 1.0, 6 August 2014

 Early childhood deaths, specify:

Family tree

Investigations Previous investigations

Date Results HIV rapid HIV DNA PCR White cell count (x 109/L) Neutrophil count (x 109/L) Lymphocyte count (x 109/L) Total protein, Albumin Mantoux Sweat test Allergy testing Other tests

Immunoglobulins (g/L) Results Reference range IgG IgA IgM IgE IgG1 IgG2 IgG3 IgG4 Measles antibody titre Tetanus antibody titre Pneumococcal antibody titre Other (specify)

28 | P a g e

PID Guide, version 1.0, 6 August 2014

Lymphocyte subsets (x 109/L) Percentage Absolute count Reference count range (absolute count) CD3+ CD3+ CD4+ (Helper cells) CD3+CD8+ (Suppressor cells) CD19/20+ (B-cells) CD16/56+ (NK cells) DR+ (activated lymphocytes)

Lymphocyte stimulation tests Patient results Phycohaemaglutinin (PHA) Stimulation index Concanavalin A (Con A) Stimulation index Pokeweed (PWM) Stimulation index Additional antigen (specify): Stimulation index Control results Phycohaemaglutinin (PHA) Stimulation index Concanavalin A (Con A) Stimulation index Pokeweed (PWM) Stimulation index Additional antigen (specify): Stimulation index

Complement tests Results Reference range Total haemolytic complement Component assay (specify):

Oxidative burst test Patient results Oxidising : resting Oxidising neutrophils: stimulated Control results Oxidising neutrophils: resting Oxidising neutrophils: stimulated

Other tests (specify):

29 | P a g e

PID Guide, version 1.0, 6 August 2014

Sample bank

 serum  DNA  PBMCs  EBV-transformed cells Other (specify):

Comment (results)

Diagnosis

Associated problems (at diagnosis)

Treatment  Antibiotic prophylaxis  IVIG  BMT  Other (specify): Treatment details (e.g. duration IVIG, BMT type/induction):

Complications & outcome (e.g. complications after diagnosis, hospitalisation, cancers, death, lost-to-follow-up; include dates)

30 | P a g e

PID Guide, version 1.0, 6 August 2014

Appendix 3: Post-HSCT immunization schedule, RCWMCH (2012)

. Sibling Allo / Auto HSCT recipients: must be 12 months post-HSCT, off all immunosuppression for 6 months (12 months for live vaccines), off IVIG for 3 months and have no evidence of active GvHD before re-immunisation schedule is commenced.

. All other HSCT recipients: must be 18 months post-HSCT, off all immunosuppression for 6 months, (12 months for live vaccines), off IVIG for 3 months, have no evidence of active GvHD, and have had immune reconstitution documented before re- immunisation schedule is commenced.

. For patients with chronic GvHD and not on IVIG, consider non-live vaccines

Sibling donor & Other HSCT Number of doses auto HSCT recipients recommended Vaccine recipients (time from HSCT) (time from HSCT)

1. Tetanus toxoid 12 months 18 months 3 (DTaP-IPV / HiB 2. Diphtheria toxoid combined vaccine) at 3. acellular Pertussis monthly intervals 4. inactivated Polio 5. Haem influenzae B 6. Hepatitis B 3 Hep B 6. Pneumococcus 15 months 21 months 2 (conjugate vaccine) 24 months 30 months 1 (polysaccharide)

7. Measles 18 + 24 months 24 + 30 months 2 (as MMR) NB 12 (attenuated) months off all 8. Mumps immunosuppressive (attenuated) therapy 9. Rubella (attenuated)

10. Varicella >24 months >24 months 1 (if recommended) (attenuated)

11. Influenza 12 months 18 months 1 (annually in winter months)

Adapted from BCH BMT Guidelines 2003 & EPI (SA) Revised Childhood Immunisation Schedule, April 2009

. Immunisation of siblings and close contacts:

o Close contacts including sublings may receive standard vaccines with exception of smallpox and OPV, refer page 15 for more detail o The recipient should be isolated from children vaccinated with OPV for at 4-6 weeks. o These precautions apply until patient is at least 12 months post-HSCT, off all immunosuppression for 12 months and no evidence of active chronic GvHD.

. Avoid BCG unless clear case of need and good evidence of immune recovery

Significant contact with measles or with VZV infection requires passive immunisation (IVIG or acyclovir). This recommendation is applicable until 12 months post sibling/auto HSCT and 18 months post other HSCT and 12 months off all immunosuppression.

31 | P a g e

PID Guide, version 1.0, 6 August 2014

Appendix 4: Refer separate pdf file entitled: SABMR preliminary search request form August 2014

32 | P a g e

PID Guide, version 1.0, 6 August 2014

Appendix 5: Refer separate pdf file entitled: Tissue typing request form July 2014

33 | P a g e