Journal of Infection (2014) xx, 1e4

www.elsevierhealth.com/journals/jinf

Recent advances in chronic granulomatous disease David Goldblatt Institute of Child Health, University College London and The Great Ormond Street Hospital for Sick Children NHS Foundation Trust, 30 Guilford Street, London WC1N 1EH, UK Accepted 18 July 2014 Available online - - -

KEYWORDS Primary immunodeficiency; Immunocompromise; Haematopoietic stem cell transplant; Gene therapy

Summary Chronic Granulomatous Disease (CGD) is a primary immunodeficiency caused by abnormities in the NADPH Oxidase that is involved in the respiratory burst responsible for initiating the killing of microbes ingested by phagocytic cells. The hallmark of CGD is recurrent infection but the inflammatory complications can prove difficult to treat. New insights into the mechanisms responsible for the inflammatory complications have led to new therapies. The treatment of CGD colitis with an anti-tumour necrosis alpha agent has been shown to be successful but associated with significant infectious complications. Haematopoietic stem cell transplants offer the possibility of cure for those with ether a matched or unrelated donor transplant, with results of the latter improving significantly over recent years. Gene Therapy offers the promise of cure without the need for a transplant but better vectors are required. ª 2014 The British Infection Association. Published by Elsevier Ltd. All rights reserved.

Introduction CGD is a primary immunodeficiency disorder with a recent reported incidence in Europe of approximately 1/250,0001 but as low as 1/120,000 in the United Kingdom and Ireland.2 CGD is caused by an inherited defect in the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase enzyme complex present in a variety of cells including phagocytes. The NADPH oxidase enzyme complex consists of two membrane-spanning subunits, gp91phox and p22phox, as well as three cytosolic components p47phox, p67phox, and

p40phox. (Reviewed by Segal3) Rac1 and Rac2, lowmolecular-weight guanosine triphosphate-binding proteins, also associate with the complex and are involved in the regulation of the NADPH oxidase.4,5 Approximately, 66% of all CGD cases result from mutations within the X-linked gp91phox gene (CYBB), followed by the autosomal recessive forms of CGD, with defects in the gene coding for p47phox (NCF-1) accounting for 30% of all CGD cases. Only 5% of the cases are due to mutations in CYBA, NCF-2 or NCF-4 which encode for p22phox, p67phox and p40phox respectively.6,7

E-mail address: [email protected] http://dx.doi.org/10.1016/j.jinf.2014.07.013 0163-4453/ª 2014 The British Infection Association. Published by Elsevier Ltd. All rights reserved. Please cite this article in press as: Goldblatt D, Recent advances in chronic granulomatous disease, J Infect (2014), http://dx.doi.org/ 10.1016/j.jinf.2014.07.013

2

Molecular basis of CGD NADPH oxidase is required for the ‘respiratory burst’ and has a critical role in microbial killing. It reduces molecular oxygen to superoxide, which subsequently reacts to form reactive oxygen species (ROS) such as hydrogen peroxide, hypochlorous acid, and hydroxyl radicals. Superoxides and their derivatives were initially thought to be the effector molecules responsible for microbial killing but new insights suggest that that this may be over simplistic.8 For example recent work exploring the mechanisms underlying NADPH oxidase function suggest that the influx of cations into and pH changes within the phagocytic vacuoles activate neutrophil proteases (neutrophil elastase, cathepsin G) that are critical for the intracellular antimicrobial activity of phagocytes.9 Activated neutrophils also kill by extracellular mechanisms, including neutrophil extracellular traps (NETs), which are composed of chromatin decorated with granular proteins.10 NETs bind bacteria and fungi and expose antimicrobial molecules. Generation of NETs is NADPH dependent and thus impaired in CGD.11 Sufferers of CGD therefor have impaired phagocytic function and are susceptible to recurrent bacterial and fungal infections. Patients are particularly susceptible to fungal infection, typically from Aspergillus species, but also catalase positive bacteria including Staphylococcus aureus and Burkholderia cepacia.12 Most patients present with infections, typically lymph node abscesses, but also recurrent respiratory infection, deep-seated abscesses and septicaemia.13 A major clinical complication of CGD is however due to uncontrolled inflammation in various organs including the GI tract, bladder, skin and the eye.14 The molecular basis for this inflammation and its associated trigger(s) remain poorly understood but a source of considerable interest with the focus on pathways that disrupt normal T cell regulatory control of inflammatory mechanisms.15,16 The consequences are the predominant cause of poor quality of life for those living with CGD.

Diagnosing CGD Making the diagnosis of CGD is not technically difficult, and historically is based on the use of the “gold standard” nitroblue tetrazolium assay.17 More recently the description and subsequently wide availability of a flow cytometric based assay18 has facilitated the diagnosis of CGD in nonspecialist laboratories. This assay is based on the reduction of dihydrorhodamine-123 (DHR) by phorbal myristate acetate stimulated phagocytic cells and is particularly useful as it can demonstrate two populations of cells in carriers. However, care must be taken in the interpretation of a negative test as acutely ill patients may show abnormal stimulation indices which normalise on recovery.19 Once abnormal NADPH oxidase has been discovered a variety of techniques can be used to assess the presence of absence of individual NADPH proteins.20 More difficult is to know whom to test for CGD as some of the clinical manifestations are not uncommon. For example, The National Laboratory for Leukocyte Functions in Israel evaluated 1296 patients over a period of 24 years, referred because of recurrent, severe pyogenic infections.21 CGD was diagnosed in 51

D. Goldblatt patients (3.9%) illustrating that even one of the defining clinical sign of CGD, recurrent bacterial infection, is not a sensitive or specific sign for CGD. As a result of the nonspecificity of the clinical symptoms late diagnosis is not uncommon and is not restricted to those with milder disease due to residual NADPH oxidase.22

Colitis Colitis is an important gastrointestinal manifestation of CGD, typically seen more commonly, but not exclusively in the X-linked form of the disease.23 In a recent survey 15/94 patients in UK and Ireland had colitic symptoms prior to diagnosis and on follow up as many as 40% of patients2,12,13 manifest with colitis. The clinical presentation of colitis is similar to that of inflammatory bowel disease, either Ulcerative Colitis (UC) or Crohns disease,24,25 and includes diarrhoea, abdominal pain and rectal bleeding. As occasionally the diagnosis of CGD is made by gastroenterologists who have previously labelled a patient as having a primary diagnosis of inflammatory bowel disease (IBD), Jaggi and colleagues screened 120 individuals aged 12 months to 21 years with a diagnosis of Chrohn’s disease, Ulcerative colitis or indeterminate IBD for CGD.26 Thirty percent of the patients had also reported a history of infection, yet using DHR not a single patient was shown to have CGD. The mainstay of therapy to date for the colitic complications of CGD has been T cell modulating agents such as azathioprine, sulfasalazine and thalidomide combined with or replacing the use of steroids.27,28 More recently, following the success of anti-tumour necrosis factor alpha (TNF-a) treatment in IBD, antibodies to TNF-a have been used in CGD. Recent experience from the NIH indicates that while the treatment is effective and can induce remission of the colitis, the immunosuppressive properties associated with antagonising TNF on a background of immune deficiency can prove problematic.29 Uzel and colleagues describe 5 CGD patients (3 XL, 2AR) who developed infectious complications while on Infliximab therapy. Three of the patients developed Burkholderia infections, one a candida infection and one multiple liver abscesses presumed to be due to Staphylococcus aureus. Neutralising TNF-a thus appears to enhance susceptibility of CGD patients to infections suggesting that TNF-a has an important role in host immunity where NADPH oxidase is absent.

Haematopoietic stem cell transplant in CGD The only curative procedure for CGD currently is a haematopoietic stem cell transplant (HSCT). The uncertain long term outcomes for CGD led the Working Party Inborn Errors of the European Blood and Marrow Transplantation (EBMT) group toward a pragmatic clinical approach to indications for HSCT in CGD.30 The group considered standard risk patients to be those in good health but with a history of one life threatening infection in the past or severe granulomatous disease and/or the non-availability of specialist care and/or non-compliance with antibiotic prophylaxis. In this clinical setting patients generally have an excellent outcome after HSCT if a HLA matched donor sibling (or matched unrelated donor (MUD), see below) is available.

Please cite this article in press as: Goldblatt D, Recent advances in chronic granulomatous disease, J Infect (2014), http://dx.doi.org/ 10.1016/j.jinf.2014.07.013

Recent advances in chronic granulomatous disease Experience in Europe has shown excellent results with HLA identical sibling HSCT’s with survival in excess of 90% and the reversal of complications such as colitis.30 Recently a multinational prospective study of reduced intensity conditioning (RIC) and HLA matched HSCT was published.31 21 HLA matched related donors and 35 HLA MUD’s were recruited to the study. Overall 2 year probability of survival was 96% with graft failure in only 3/56 patients similar numbers with Graft versus host disease. The overall survival for MUD’s was similar with an overall survival of 91%, a significant improvement on earlier studies and outcomes. In centres with experience, HSCT’s should be considered the treatment of choice for a child of standard risk with any genetic form of CGD. Post HSCT children have improved clinical outcomes including growth32 and demonstrate improved psychological outcomes.33

3

3. 4.

5.

6. 7.

8. 9.

Gene therapy for CGD 10.

The first human stem cell (HSC) gene therapy trial for CGD disease was initiated in the US without chemotherapy, but only achieved short-term engraftment with no long-lasting benefit for patients.34 This suggested that it was probably necessary to condition patients prior to gene therapy, as CGD-corrected cells are not likely to have any selective growth or survival advantage when re-infused. More recently, five early phase clinical studies, involving a total of 12 patients have been conducted in several centres worldwide incorporating conditioning with low intensity alkylating agent.35 The experience of Grez and colleagues demonstrated biologically restoration of NADPH-oxidase activity in circulating neutrophils for a short time after engraftment, and some clinical benefit allowing the majority of patients to clear pre-existing infections. However, clinical benefit was usually only transient, and gene marking rapidly decreased with only few patients having significant marking after three months, indicating an absence of long-term engraftment.36 Two patients treated with a specific gammaretroviral vector developed myelodysplasia with monosomy 7 as a result of insertional transactivation of myeloproliferative genes.37 One subject died 27 months after gene therapy from overwhelming sepsis while the other underwent a successful allogeneic HSC transplant. To improve the safety and efficacy of gene therapy for CGD, a new lentiviral vector has been designed that directs transgene expression primarily in myeloid cells38 and holds out the promise for safer gene therapy and long term cure of CGD.

11.

12.

13.

14.

15.

16.

17.

18.

19.

Conflict of interest The author has no conflict of interest to report.

20.

References

21.

1. van den Berg JM, van Koppen E, Ahlin A, Belohradsky BH, Bernatowska E, Corbeel L, et al. Chronic granulomatous disease: the European experience. PLoS One 2009;4(4):e5234. 2. Jones LB, McGrogan P, Flood TJ, Gennery AR, Morton L, Thrasher A, et al. Special article: chronic granulomatous disease in the United Kingdom and Ireland: a comprehensive

22.

23.

national patient-based registry. Clin Exp Immunol 2008; 152(2):211e8. Segal AW. The NADPH oxidase and chronic granulomatous disease. Mol Med Today 1996;2(3):129e35. Abo A, Pick E, Hall A, Totty N, Teahan CG, Segal AW. Activation of the NADPH oxidase involves the small GTP-binding protein p21rac1. Nature 1991;353(6345):668e70. Knaus UG, Heyworth PG, Evans T, Curnutte JT, Bokoch GM. Regulation of phagocyte oxygen radical production by the GTP-binding protein Rac 2. Science 1991;254(5037):1512e5. Meischl C, Roos D. The molecular basis of chronic granulomatous disease. Springer Semin Immunopathol 1998;19(4):417e34. Segal BH, Leto TL, Gallin JI, Malech HL, Holland SM. Genetic, biochemical and clinical features of chronic granulomatous disease. Medicine 2000;79:170e200. Nauseef WM. How human neutrophils kill and degrade microbes: an integrated view. Immunol Rev 2007;219:88e102. Ahluwalia J, Tinker A, Clapp LH, Duchen MR, Abramov AY, Pope S, et al. The large-conductance Ca2þ-activated Kþ channel is essential for innate immunity. Nature 2004;427:853e8. Brinkmann V, Zychlinsky A. Beneficial suicide: why neutrophils die to make NETs. Nat Rev Microbiol 2007;5(8):577e82. Bianchi M, Niemiec MJ, Siler U, Urban CF, Reichenbach J. Restoration of anti-Aspergillus defense by neutrophil extracellular traps in human chronic granulomatous disease after gene therapy is calprotectin-dependent. J Allergy Clin Immunol 2011;127(5). 1243e52 e7. Winkelstein JA, Marino MC, Johnston Jr RB, Boyle J, Curnutte J, Gallin JI, et al. Chronic granulomatous disease. Report on a national registry of 368 patients. Medicine (Baltimore) 2000;79(3):155e69. Cale CM, Jones AM, Goldblatt D. Follow up of patients with chronic granulomatous disease diagnosed since 1990. Clin Exp Immunol 2000;120(2):351e5. Rosenzweig SD. Inflammatory manifestations in chronic granulomatous disease (CGD). J Clin Immunol 2008;28(Suppl. 1): S67e72. Bylund J, MacDonald KL, Brown KL, Mydel P, Collins LV, Hancock RE, et al. Enhanced inflammatory responses of chronic granulomatous disease leukocytes involve ROSindependent activation of NF-kappa B. Eur J Immunol 2007; 37(4):1087e96. Rieber N, Hector A, Kuijpers T, Roos D, Hartl D. Current concepts of hyperinflammation in chronic granulomatous disease. Clin Dev Immunol 2012;2012:252460. Levinsky RJ, Harvey BA, Rodeck CH, Soothill JF. Phorbol myristate acetate stimulated NBT test: a simple method suitable for antenatal diagnosis of chronic granulomatous disease. Clin Exp Immunol 1983;54(2):595e8. Roesler J, Hecht M, Freihorst J, Lohmann-Matthes ML, Emmendorffer A. Diagnosis of chronic granulomatous disease and of its mode of inheritance by dihydrorhodamine 123 and flow microcytofluorometry. Eur J Pediatr 1991;150(3):161e5. Ang EY, Soh JY, Liew WK, Chan KW, Thoon KC, Chong CY, et al. Reliability of acute illness dihydrorhodamine-123 testing for chronic granulomatous disease. Clin Lab 2013; 59(1e2):203e6. Roos D, de Boer M. Molecular diagnosis of chronic granulomatous disease. Clin Exp Immunol 2014;175(2):139e49. Ben-Ari J, Wolach O, Gavrieli R, Wolach B. Infections associated with chronic granulomatous disease: linking genetics to phenotypic expression. Expert Rev Anti Infect Ther 2012; 10(8):881e94. Liese JG, Jendrossek V, Jansson A, Petropoulou T, Kloos S, Belohradsky BH, et al. Chronic granulomatous disease in adults. Lancet 1996;347:220e3. Marciano BE, Rosenzweig SD, Kleiner DE, Anderson VL, Darnell DN, Anaya-O’Brien S, et al. Gastrointestinal

Please cite this article in press as: Goldblatt D, Recent advances in chronic granulomatous disease, J Infect (2014), http://dx.doi.org/ 10.1016/j.jinf.2014.07.013

4

24.

25.

26.

27.

28. 29.

30.

31.

32.

D. Goldblatt involvement in chronic granulomatous disease. Pediatrics 2004;114(2):462e8. Schappi MG, Smith VV, Goldblatt D, Lindley KJ, Milla PJ. Colitis in chronic granulomatous disease. Arch Dis Child 2001;84(2): 147e51. Levine S, Smith VV, Malone M, Sebire NJ. Histopathological features of chronic granulomatous disease (CGD) in childhood. Histopathology 2005;47(5):508e16. Jaggi P, Scherzer R, Knieper R, Mousa H, Prasad V. Utility of screening for chronic granulomatous disease in patients with inflammatory bowel disease. J Clin Immunol 2012;32(1): 78e81. Agarwal S, Mayer L. Diagnosis and treatment of gastrointestinal disorders in patients with primary immunodeficiency. Clin Gastroenterol Hepatol Off Clin Pract J Am Gastroenterol Assoc 2013;11(9):1050e63. Holland SM. Chronic granulomatous disease. Clin Rev Allergy Immunol 2010;38(1):3e10. Uzel G, Orange JS, Poliak N, Marciano BE, Heller T, Holland SM. Complications of tumor necrosis factor-alpha blockade in chronic granulomatous disease-related colitis. Clin Infect Dis 2010;51(12):1429e34. Seger RA. Hematopoietic stem cell transplantation for chronic granulomatous disease. Immunol Allergy Clin N Am 2010;30(2): 195e208. Gungor T, Teira P, Slatter M, Stussi G, Stepensky P, Moshous D, et al. Reduced-intensity conditioning and HLA-matched haemopoietic stem-cell transplantation in patients with chronic granulomatous disease: a prospective multicentre study. Lancet 2014;383(9915):436e48. Cole T, Pearce MS, Cant AJ, Cale CM, Goldblatt D, Gennery AR. Clinical outcome in children with chronic granulomatous

33.

34.

35.

36.

37.

38.

disease managed conservatively or with hematopoietic stem cell transplantation. J Allergy Clin Immunol 2013;132(5): 1150e5. Cole T, McKendrick F, Titman P, Cant AJ, Pearce MS, Cale CM, et al. Health related quality of life and emotional health in children with chronic granulomatous disease: a comparison of those managed conservatively with those that have undergone haematopoietic stem cell transplant. J Clin Immunol 2013; 33(1):8e13. Malech HL, Maples PB, Whiting-Theobald N, Linton GF, Sekhsaria S, Vowells SJ, et al. Prolonged production of NADPH oxidase-corrected granulocytes after gene therapy of chronic granulomatous disease. Proc Natl Acad Sci U S A 1997;94(22): 12133e8. Grez M, Reichenbach J, Schwable J, Seger R, Dinauer MC, Thrasher AJ. Gene therapy of chronic granulomatous disease: the engraftment dilemma. Mol Ther 2011;19(1):28e35. Kang HJ, Bartholomae CC, Paruzynski A, Arens A, Kim S, Yu SS, et al. Retroviral gene therapy for X-linked chronic granulomatous disease: results from phase I/II trial. Mol Ther 2011; 19(11):2092e101. Stein S, Ott MG, Schultze-Strasser S, Jauch A, Burwinkel B, Kinner A, et al. Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease. Nat Med 2010;16(2): 198e204. Santilli G, Almarza E, Brendel C, Choi U, Beilin C, Blundell MP, et al. Biochemical correction of X-CGD by a novel chimeric promoter regulating high levels of transgene expression in myeloid cells. Mol Ther 2011;19(1):122e32.

Please cite this article in press as: Goldblatt D, Recent advances in chronic granulomatous disease, J Infect (2014), http://dx.doi.org/ 10.1016/j.jinf.2014.07.013

Recent advances in chronic granulomatous disease.

Chronic Granulomatous Disease (CGD) is a primary immunodeficiency caused by abnormities in the NADPH Oxidase that is involved in the respiratory burst...
268KB Sizes 0 Downloads 10 Views