Acessibilidade / Reportar erro

Importance of immune response genes in hemophilia A

Abstract

Hemophilia A is a disease caused by a deficiency of coagulation factor VIII resulting from genetic inheritance linked to chromosome X. One treatment option is the administration of plasma or recombinant FVIII. However, some patients develop inhibitors or antibodies against this factor. Inhibitors are alloantibodies that bind to the epitope of factor VIII causing it to be recognized by the immune system as a foreign peptide. This is the most serious complication in hemophilia patients in respect to replacement therapy. Some studies have suggested that genetic factors influence the development of factor VIII inhibitors such as ethnicity, family history, mutations in the factor VIII gene and in genes of the immune system. The aim of this study was to conduct a literature review to assess the influence of genetic factors of immune response genes, especially genes of the major histocompatibility complex and cytokines, which may be related to the development of factor VIII inhibitors in hemophilia A patients. Understanding these risk factors will help to determine future differential treatment in the control and prevention of the development of inhibitors.

Hemophilia A; Blood coagulation; Factor VIII; Major histocompatibility complex; Cytokines; Human Leukocyte Antigen


Introduction

Hemophilia A is a hereditary bleeding disorder caused by the deficiency or abnormality of factor VIII (FVIII) coagulant activity(11. Bolton-Maggs PH, Pasi KJ. Haemophilias A and B. Lancet. 2003;361(9371):1801-9.). The deficiency or dysfunction of FVIII, a glycoprotein that operates as a cofactor in the activation of factor X (FX) via activated factor IX (FIX), does not allow the formation of a normal clot at the site of an injury(11. Bolton-Maggs PH, Pasi KJ. Haemophilias A and B. Lancet. 2003;361(9371):1801-9.

2. Shen BW, Spiegel PC, Chang CH, Huh JW, Lee JS, Kim J, et al. The tertiary structure and domain organization of coagulation factor VIII. Blood. 2008;111(3):1240-7.
-33. Giangrande PL. The molecular basis of hemophilia. In: Provan D, Gribben JG, editors. Molecular Hematology. 3rd ed. Oxford, UK: Wiley-Blackwell; 2008. p.184-92.). In 2010, according to the register of the World Federation of Hemophilia, there were approximately 10,065 hemophilia patients in Brazil with 80% being hemophilia A(44. World Federation of Hemophilia. Report on the Annual Global Survey 2010 [Internet[. Montreal, Canada: WFH; 2011 [cited 2013 Jan 21]. Available from: http://www1.wfh.org/publications/files/pdf-1427.pdf
http://www1.wfh.org/publications/files/p...
).

The treatment of hemophilia A involves FVIII replacement therapy. However, some patients develop inhibitors, that are polyclonal antibodies against the administered FVIII.(55. Brasil. Ministério da Saúde. Secretaria de Atenção à Saúde. Departamento de Atenção Especializada. Manual de reabilitação na hemofilia [Internet]. Brasília: MS; 2011 [cited 2012 Jul 27]. Available from: http://portal.saude.gov.br/portal/arquivos/pdf/manual_de_reabilitacao_consulta_publica.pdf
http://portal.saude.gov.br/portal/arquiv...
) This reduces the effectiveness of replacement therapy and is one of the main complications in the treatment of patients(55. Brasil. Ministério da Saúde. Secretaria de Atenção à Saúde. Departamento de Atenção Especializada. Manual de reabilitação na hemofilia [Internet]. Brasília: MS; 2011 [cited 2012 Jul 27]. Available from: http://portal.saude.gov.br/portal/arquivos/pdf/manual_de_reabilitacao_consulta_publica.pdf
http://portal.saude.gov.br/portal/arquiv...
,66. Brown TM, Lee WC, Joshi AV, Pashos CL. Health-related quality of life and productivity impact in haemophilia patients with inhibitors. Haemophilia. 2009;15(4):911-7.). Various mechanisms are involved in the formation of inhibitors, for example, ethnicity, family history, FVIII gene mutations and processes that involve immune system genes. The study of genetic factors is essential to elucidate mechanisms that may influence the development of inhibitors as in the absence of a genetic predisposing factor there is much lower risk of developing inhibitors. On the other hand, the combined action of genetic and non-genetic factors can significantly contribute to the development of inhibitors. These factors may activate or inhibit the immune response depending on changes in immunological regulators and cytokine profiles. By predicting these factors in patients, it may be possible to avoid the formation of FVIII inhibitors and provide more effective treatment(77. Astermark J. Prevention and prediction of inhibitor risk. Haemophilia. 2012;18(Suppl 4):38-42.).

As the identification of factors related to the mechanisms involved in the development of inhibitors in hemophilia A is of paramount importance, the aim of this study was to conduct a review of the literature in order to assess the influence of genetic factors and immune system genes on the development of FVIII inhibitors in hemophilia A patients. This study may contribute to a greater understanding of the mechanisms involved in the susceptibility that some people have in producing alloantibodies against FVIII, as well as to develop new, more effective therapies for hemophilia patients with inhibitors.

Development of factor VIII inhibitors

Antibodies, also called inhibitors, are high-affinity immunoglobulin G (IgG) that are directed against infused FVIII; this reduces the effectiveness of treatment(88. Fulcher CA, de Graaf Mahoney S, Zimmerman TS. FVIII inhibitor IgG subclass and FVIII polypeptide specificity determined by immunoblotting. Blood. 1987;69(5):1475-80.). These inhibitors make the adequate correction of bleeding diathesis more difficult in cases of bleeding and in surgery, and also make regular prophylaxis with FVIII impossible(99. Gouw SC, van den Berg HM, Oldenburg J, Astermark J, de Groot PG, Margaglione M, et al. F8 gene mutation type and inhibitor development in patients with severe hemophilia A: systematic review and meta-analysis. Blood. 2012;119(12):2922-34.). Studies have shown that the overall prevalence of inhibitors in unselected hemophiliac populations is 5-7% and the prevalence amongst severe hemophilia patients is substantially greater, at between 12% and 13%(1010. Wight J, Paisley S. The epidemiology of inhibitors in haemophilia A: a systematic review. Haemophilia. 2003;9(4):418-35. Comment in: Haemophilia. 2004;10(2):197-8.).

The FVIII inhibitors are classified as either high- or low-responding depending on how an individual's immune system is stimulated upon repeated exposure to FVIII. If the immune system reacts briskly and strongly, the amount of inhibitor directed against FVIII can rise quickly to very high levels with titers of at least 5 Bethesda units. This type of inhibitor is generally characterized as high-responding. On the other hand, the immune system may be stimulated in such a way that its response to exposure to FVIII is slower and weaker, and the inhibitor titer remains low, usually under 5 Bethesda units. When these characteristics are present, the inhibitor is generally termed low-responding(1111. DiMichele DM. Inhibitors in haemophilia: a primer. Haemophilia. 2000;6(Suppl 1):38-40.).

It is noteworthy that during the lifetime of a person with hemophilia, the risk of developing an inhibitor is variable(1111. DiMichele DM. Inhibitors in haemophilia: a primer. Haemophilia. 2000;6(Suppl 1):38-40.). There are reports that the risk is at its highest soon after the first exposure to FVIII, with the cumulative risk of inhibitor development leveling off after 20 exposure days or at 6-10 years of age(1010. Wight J, Paisley S. The epidemiology of inhibitors in haemophilia A: a systematic review. Haemophilia. 2003;9(4):418-35. Comment in: Haemophilia. 2004;10(2):197-8.).

The treatment of hemophilia patients with inhibitors is a major challenge, and is based on the use of agents that generate thrombin, independent of the actions of FVIII or FIX, the so-called bypassing agents. The two most widely used bypassing agents are activated prothrombin complex concentrate (APCC) and recombinant activated factor VII (rFVIIa). Despite their efficacy in the control of most bleeding episodes, treatment with these agents present several limitations compared to the use of FVIII in non-inhibitor patients such as a less predictable hemostatic response, lack of laboratory monitoring tests and a higher cost. Furthermore, because of the lower efficacy and shorter half-life, regular prophylaxis is not feasible with these agents(1111. DiMichele DM. Inhibitors in haemophilia: a primer. Haemophilia. 2000;6(Suppl 1):38-40.).

Mechanism of the formation of factor VIII inhibitors

Several studies indicate that the immune response triggered by the presence of exogenous FVIII is a T helper cell-mediated event that depends on antigen-presenting cells (CAAs), such as macrophages, dendritic cells and B cells(1212. André S, Meslier Y, Dimitrov JD, Repessé Y, Kaveri SV, Lacroix-Desmazes S, et al. A cellular viewpoint of anti-FVIII immune response in hemophilia A. Clin Rev Allergy Immunol. 2009;37(2):105-13.,1313. Astermark J. Inhibitor development: patient-determined risk factors. Haemophilia. 2010;16(102):66-70.).

For the synthesis of antibodies against FVIII to occur, part of the FVIII administered to the hemophilia patient must be internalized by CAAs, degraded and presented to a class II Major Histocompatibility Complex (MHC) molecule(1212. André S, Meslier Y, Dimitrov JD, Repessé Y, Kaveri SV, Lacroix-Desmazes S, et al. A cellular viewpoint of anti-FVIII immune response in hemophilia A. Clin Rev Allergy Immunol. 2009;37(2):105-13.,1414. White GC 2nd, Kempton CL, Grimsley A, Nielsen B, Roberts HR. Cellular immune responses in hemophilia: why do inhibitors develop in some, but not all hemophiliacs? J Thromb Haemost. 2005;3(8):1676-81.). A plasma membrane complex of CAAs, formed when FVIII peptides bind to MHC molecules, is then made available for recognition by CD4+ T cells(1515. Ghosh K, Shetty S. Immune response to FVIII in hemophilia A: an overview of risk factors. Clin Rev Allergy Immunol. 2009;37(2):58-66.). In addition to this, intracellular peptide fragments of FVIII, synthesized in small quantities by the patient, are presented via MHC Class I molecules to CD8+ T cells(1616. Chaves DG, Rodrigues CV. Development of factor VIII inhibitors in hemophilia A. Rev Bras Hematol Hemoter. 2009;31(5):384-90.).

The peptide-MHC complex on the surface of CAAs is recognized by antigen T cell receptors (TCRs). For the presentation of antigens to the TCRs to be efficient, a second signal occurs between the CAAs and T cells; the co-stimulating CD80/86 molecules expressed in CAAs bind to CD28 expressed in T cells(1010. Wight J, Paisley S. The epidemiology of inhibitors in haemophilia A: a systematic review. Haemophilia. 2003;9(4):418-35. Comment in: Haemophilia. 2004;10(2):197-8.,1717. Hoyer LW. The incidence of factor VIII inhibitors in patients with severe hemophilia A. Adv Exp Med Biol. 1995;386:35-45.). Activation of T cells occurs when both signals are present; this can be either type-1 helper T cell (Th1) activation, responsible for the secretion of cytokines such as interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α) and interleukin-2 (IL-2), which are important in the cellular immunity system, or type-2 helper T cell (Th2) activation which secretes interleukin-4 (IL-4), interleukin-5 (IL-5), Interleukin-6 (IL-6) and interleukin-10 (IL-10), among others that are important in humoral immunity. Moreover, the expression of CD2, CD30, CD40L and CD28 is increased on the CD4+ T cell surface(1212. André S, Meslier Y, Dimitrov JD, Repessé Y, Kaveri SV, Lacroix-Desmazes S, et al. A cellular viewpoint of anti-FVIII immune response in hemophilia A. Clin Rev Allergy Immunol. 2009;37(2):105-13.,1515. Ghosh K, Shetty S. Immune response to FVIII in hemophilia A: an overview of risk factors. Clin Rev Allergy Immunol. 2009;37(2):58-66.,1818. Oldenburg J, Pavlova A. Genetic risk factors for inhibitors to factors VIII and IX. Haemophilia. 2006;12(Suppl 6):15-22.,1919. Pratt KP, Thompson AR. B-cell and T-cell epitopes in anti-factor VIII immune responses. Clin Rev Allergy Immunol. 2009;37(2):80-95.). When secreted by Th1 or Th2, these cytokines stimulate the differentiation of B cells, which change the isotype of immunoglobulin and produce specific antibodies against plasma FVIII. In addition, the B cells secrete the cytokine interleukin-12 (IL-12) that in turn stimulates the Th1-mediated production of IFN-γ(1818. Oldenburg J, Pavlova A. Genetic risk factors for inhibitors to factors VIII and IX. Haemophilia. 2006;12(Suppl 6):15-22.). The cytokines from Th1 stimulate the development of immunoglobulin G1 (IgG1) and immunoglobulin G2 (IgG2) subclasses, whereas Th2 cells stimulate the development of immunoglobulin G4 (IgG4). Studies in patients with hemophilia A have shown that the titers of high-response inhibitors are correlated to IgG4 levels, which suggests that the Th2-mediated immune response is strongly related to the synthesis of anti-FVIII antibodies(1919. Pratt KP, Thompson AR. B-cell and T-cell epitopes in anti-factor VIII immune responses. Clin Rev Allergy Immunol. 2009;37(2):80-95.).

Factors predisposing patients to the development of factor VIII inhibitors

Ethnicity and family history of factor VIII inhibitors

Ethnicity and family history have been associated with predisposition for the development of FVIII inhibitors. One study showed that Afro-Americans had a higher risk of developing inhibitors(2020. Gill JC. The role of genetics in inhibitor formation. Thromb Haemost. 1999;82(2):500-4.). This association and family history of the development of inhibitors were also observed in The Malmö International Brother Study (MIBS). The results of this study show that the incidence of inhibitors is high in the subgroup of people of African descent when compared to Caucasians (55.6% vs. 27.4%)(2121. Astermark J, Berntorp E, White GC, Kroner BL; MIBS Study Group. The Malmö International Brother Study (MIBS): further support for genetic predisposition to inhibitor development in hemophilia patients. Haemophilia. 2001;7(3):267-72.). It is believed that the racial component is mainly based on genetic variants in immune response determinants, because the FVIII mutation spectrum does not differ between races(1818. Oldenburg J, Pavlova A. Genetic risk factors for inhibitors to factors VIII and IX. Haemophilia. 2006;12(Suppl 6):15-22.).

Additionally, it was observed that the risk for the formation of inhibitors increases significantly in patients with a family history of inhibitors; the absolute risk for the development of inhibitors in patients with a family history of inhibitor development was 48% (95% confidence interval - 95% CI: 35-62%), while the risk in patients with no family history was 15% (95% CI: 11-21%)(2121. Astermark J, Berntorp E, White GC, Kroner BL; MIBS Study Group. The Malmö International Brother Study (MIBS): further support for genetic predisposition to inhibitor development in hemophilia patients. Haemophilia. 2001;7(3):267-72.).

Another study by the same group assessed the possibility of genetic mutations influencing the formation of inhibitors in individuals of the same family. One hundred and thirteen mainly Caucasian families with two or more brothers with severe hemophilia were analyzed. All brothers in 59 of the families developed inhibitors and 25 (42.4%) of these had a family history of inhibitor development. It was found that the most common type of mutation in the FVIII gene was inversion of intron 22. Inhibitors were identified in 45 of 74 families (60.8%) with this type of mutation; and in 18 (40%) of these 45 families, all the brothers developed inhibitors(1818. Oldenburg J, Pavlova A. Genetic risk factors for inhibitors to factors VIII and IX. Haemophilia. 2006;12(Suppl 6):15-22.).

In light of these data, the influence of genetic factors in the development of inhibitors is evident(2121. Astermark J, Berntorp E, White GC, Kroner BL; MIBS Study Group. The Malmö International Brother Study (MIBS): further support for genetic predisposition to inhibitor development in hemophilia patients. Haemophilia. 2001;7(3):267-72.). Non-genetic factors also appear to influence the immune response and consequently change the risk of developing inhibitors in each family. However, it is unlikely that these factors alone can explain the similarities reported. These observations suggest that there must be changes in the immune response that may be based on both genetic markers and non-genetic factors(1818. Oldenburg J, Pavlova A. Genetic risk factors for inhibitors to factors VIII and IX. Haemophilia. 2006;12(Suppl 6):15-22.).

Mutations in the factor VIII gene

The FVIII gene is located on the end of the long arm of chromosome X (Xq28). It comprises 186,000 base pairs distributed between 26 exons and 25 introns. The product of this gene is a polypeptide of 2332 amino acids (inactivated circulating pro-cofactor) and the activated polypeptide is formed of six arranged domains(22. Shen BW, Spiegel PC, Chang CH, Huh JW, Lee JS, Kim J, et al. The tertiary structure and domain organization of coagulation factor VIII. Blood. 2008;111(3):1240-7.,2222. Gitschier J, Wood WI, Goralka TM, Wion KL, Chen EY, Eaton DH, et al. Characterization of the human factor VIII gene. Nature. 1984;312(5992):326-30.,2323. Castaldo G, D'Argenio V, Nardiello P, Zarrilli F, Sanna V, Rocino A, et al. Haemophilia A: molecular insights. Clin Chem Lab Med. 2007;45(4):450-61.). The A2, A3 and C2 domains are the regions in which anti-FVIII antibodies can react and impair the coagulation cascade(2424. Scandella D, Mattingly M, de Graaf S, Fulcher CA. Localization of epitopes for human factor VIII inhibitor antibodies by immunoblotting and antibody neutralization. Blood. 1989;74(5):1618-26.,2525. Scandella D, Kessler C, Esmon P, Hurst D, Courter S, Gomperts E, et al. Epitope specificity and functional characterization of factor VIII inhibitors. Adv Exp Med Biol. 1995;386:47-63.). Studies have shown that in patients with missense mutations clustered in the A2 and C2 domains, the risk of inhibitor formation is fourfold greater than in patients with mutations outside this region. This indicates that any changes in the three-dimensional structure of this part of the FVIII molecule may affect its immunogenicity(1818. Oldenburg J, Pavlova A. Genetic risk factors for inhibitors to factors VIII and IX. Haemophilia. 2006;12(Suppl 6):15-22.). The anti-A2 antibodies and in some cases, anti-C2 antibodies, for example, can interact with the A2 domain and C2 domain, respectively, and neutralize the procoagulant activity of FVIII(2525. Scandella D, Kessler C, Esmon P, Hurst D, Courter S, Gomperts E, et al. Epitope specificity and functional characterization of factor VIII inhibitors. Adv Exp Med Biol. 1995;386:47-63.). The C2 domain however is affected by anti-C2 antibodies which prevent FVIII binding to phospholipids and von Willebrand factor(2626. Arai M, Scandella D, Hoyer LW. Molecular basis of factor VIII inhibition by human antibodies. Antibodies that bind to the factor VIII light chain prevent the interaction of factor VIII with phospholipid. J Clin Invest. 1989;83(6):1978-84.), and anti-A3 antibodies target the A3 domain to prevent the interaction of FIX with activated FVIII(2727. Lenting PJ, van de Loo JW, Donath MJ, van Mourik JA, Mertens K. The sequence Glu1811-Lys1818 of human blood coagulation factor VIII comprises a binding site for activated factor IX. J Biol Chem. 1996;271(4):1935-40.).

By the year 2012, 5243 types of mutations associated with this disease had been reported according to the HAMSTeRS (Haemophilia A Mutation Test and Resource Search Site) electronic database(2828. HAMSTeRS. The Haemophilia A Mutation, Structure, Test and Resource Site. [Internet]. London: University College London and Royal Free Hospital London; 2013 [cited 2013 April 16]. Available from: http://hadb.org.uk/
http://hadb.org.uk/...
). The group with the highest risk of producing anti-FVIII antibodies is the one with the greatest changes in the gene(1818. Oldenburg J, Pavlova A. Genetic risk factors for inhibitors to factors VIII and IX. Haemophilia. 2006;12(Suppl 6):15-22.,2929. Astermark J, Oldenburg J, Escobar M, White GC 2nd, Berntorp E; Malmö International Brother Study group. The Malmo International Brother Study (MIBS). Genetic defects and inhibitor development in siblings with severe hemophilia A. Haematologica. 2005;90(7):924-31. Comment in: Haematologica. 2005;90(7):869A.). One meta-analysis observed that the risk of inhibitor development in patients with large deletions and nonsense mutations is higher than in patients with intron 22 inversions (pooled OR = 3.6 and OR = 1.4, respectively). The risk of patients with intron 1 inversions and splice-site mutations is virtually equal (pooled OR = 0.9 and OR = 1.0, respectively), and the risk of patients with small deletions and insertions and missense mutations are lower (pooled OR = 0.5 and OR = 0.3, respectively)(99. Gouw SC, van den Berg HM, Oldenburg J, Astermark J, de Groot PG, Margaglione M, et al. F8 gene mutation type and inhibitor development in patients with severe hemophilia A: systematic review and meta-analysis. Blood. 2012;119(12):2922-34.). Inversions in intron 22 (30-50%) and intron 1 (0-5%) are usually associated with the severe hemophilia A phenotype and an intermediary risk for forming inhibitors(1515. Ghosh K, Shetty S. Immune response to FVIII in hemophilia A: an overview of risk factors. Clin Rev Allergy Immunol. 2009;37(2):58-66.,3030. Albánez S, Ruiz-Sáez A, Boadas A, de Bosch N, Porco A. Identification of factor VIII gene mutations in patients with severe haemophilia A in Venezuela: identification of seven novel mutations. Haemophilia. 2011;17(5):e913-8.,3131. Zimmermann MA, Oldenburg J, Müller CR, Rost S. Unusual genomic rearrangements in introns 1 and 22 of the F8 gene. Hamostaseologie. 2011;31(Suppl 1):S69-73.).

In Brazil, a study of 86 Caucasian patients, investigated the occurrence of inversion mutations in 47 patients. Of these, 33 (70%) had severe hemophilia and 14 (30%) moderate or mild hemophilia. An increase in the frequency of inversion mutations (13/33; 39.4%) was observed in the subgroup classified as severe with the majority (11/13; 86.4%) being mutations in intron 22(3232. Soares RP, Chamone DA, Bydlowski SP. Factor VIII gene inversions and polymorphisms in Brazilian patients with haemophilia A: carrier detection and prenatal diagnosis. Haemophilia. 2001;7(3):299-305.).

Class I and II human leukocyte antigen molecules

In humans, MHC genes, located on the short arm of chromosome 6, are named human leukocyte antigen (HLA) genes. The HLA system occupies a highly polymorphic DNA region of approximately 3600 kilobases (Kb). These loci contain most of the genetic information necessary for the development of antigen presentation activity(3333. Howell WM, Carter V, Clark B. The HLA system: immunobiology, HLA typing, antibody screening and crossmatching techniques. J Clin Pathol. 2010;63(5):387-90.).

There are two main types of HLA gene products, class I and class II molecules, which recognize different types of protein antigens, intracellular (cytosolic antigens) and extracellular. CD4 T cells recognize antigens presented by class II molecules, while CD8 cells recognize antigens that are together with class I molecules(3434. Magalhães PS, Böhlke M, Neubarth F. The Major Histocompatibility Complex (MHC): genetic codification, structural bases and clinical implications. Rev Med UCPEL. 2004;2(1):54-9.,3535. Donadi EA. How to understand the nomenclature and the mechanisms involved on the association between histocompatibility antigens and alleles with disease. Medicina (Ribeirão Preto). 2000;33(1):7-18.).

The class III region is found between the groups of class I and II genes, where genes that code for various components of the complement system and structurally related cytokines such as TNF-α, lymphotoxin-α and lymphotoxin-β are located(3636. Meyer D, Thomson G. How selection shapes variation of the human major histocompatibility complex: a review. Ann Hum Genet. 2001;65(Pt 1):1-26.).

The generation of antibodies against peptide antigens bound to FVIII involves cell cooperation that results in the presentation of antigens to T and B cells. The start of this process involves the processing of proteins by antigen-presenting cells and subsequent association of these peptides to HLA molecules in these cells. For extracellular proteins, such as the exogenous FVIII administered to hemophilia A patients, it is the HLA class II molecules that mediate the processing of antigenic peptides(1414. White GC 2nd, Kempton CL, Grimsley A, Nielsen B, Roberts HR. Cellular immune responses in hemophilia: why do inhibitors develop in some, but not all hemophiliacs? J Thromb Haemost. 2005;3(8):1676-81.).

Recently, DNA systematic sequence analyses between haplotypes have yielded information on polymorphisms across the complete MHC. This information provides pools of sequence variants for disease association analysis(3737. Traherne JA. Human MHC architecture and evolution: implications for disease association studies. Int J Immunogenet. 2008;35(3):179-92.). Several studies are committed to identifying an association between haplotypes of HLA class I and II molecules and the risk for the development of inhibitors. Hay et al. studied 176 hemophilia patients in England and found a higher frequency of the HLA-DRB1*15:01/DQB1*06:02/DQA1*01:02 haplotype in patients with inhibitors, but only the frequency of the HLA-DQA1*01:02 allele was considered statically significant (OR = 2.7; 95% CI: 1.2-5.9). The HLA-DRB1*01, HLA-DQB1*05:01 and DRB1*01:01 alleles were also higher in the group of patients with inhibitors in the presence of the intron 22 inversion mutation, but the values were not statistically significant(3838. Hay CR, Ollier W, Pepper L, Cumming A, Keeney S, Goodeve AC, et al. HLA class II profile: a weak determinant of factor VIII inhibitor development in severe haemophilia A. UKHCDO Inhibitor Working Party. Thromb Haemost. 1997;77(2):234-7.). The HLA-DQA1*01:02, HLA-DQB1*06:02 and HLA-DRB1*15 alleles were found at higher frequencies in patients with inhibitors by Oldenburg et al. in Germany. Moreover, other HLA class I alleles (HLA-A*03, HLA-B*07 and HLA-C*07) were identified at higher frequencies(3939. Oldenburg J, Picard JK, Schwaab R, Brackmann HH, Tuddenham EG, Simpson E. HLA genotype of patients with severe haemophilia A due to intron 22 inversion with and without inhibitors of factor VIII. Thromb Haemost. 1997;77(2):238-242.).

Despite the weak haplotype association in these studies, another more recent investigation demonstrated a significant association for class II HLA molecules. This case-control study found a positive association of the DRB1*15:01/DQB1*06:02 haplotype with the formation of inhibitors in 260 severe hemophilia patients from Germany (p-value = 0.0423; OR = 1.9; 95% CI: 1.01-3.57)(4040. Pavlova A, Delev D, Lacroix-Desmazes S, Schwaab R, Mende M, Fimmers R, et al. Impact of polymorphisms of the major histocompatibility complex class II, interleukin-10, tumor necrosis factor-alpha and cytotoxic T-lymphocyte antigen-4 genes on inhibitor development in severe hemophilia A. J Thromb Haemost. 2009;7(12):2006-15.).

The studies that had the most statistically significant results were those that tested for associations of alleles of HLA genes in respect to susceptibility or resistance of patients in developing inhibitors. The main associations have been found for HLA class II molecules, perhaps because, from the point of view of immunity, these alleles are more important in presenting the peptides of FVIII to T cells, and so studies have focused more on genotyping these alleles. One study of 57 hemophilia patients (with and without inhibitors) and 36 blood donors without the disease from Thailand found a higher frequency of the DRB1*15 allele in patients with inhibitors (30.6%) than in patients without inhibitors (19.2%). However, statistical significance was obtained only when the frequency of this allele was compared between patients with inhibitors and controls (30.6% vs. 13.9%; p-value = 0.021; OR = 0.021; 95% CI: 1.16-6.47)(4141. Nathalang O, Sriwanitchrak P, Sasanakul W, Chuansumrit A. Association of HLA class II alleles and the occurrence of factor VIII inhibitor in Thai patients with hemophilia A. Turk J Hematol. 2012;29(1):34-9.).

In another study performed in Germany, the frequencies of the HLA-DRB1*16 (0.122 vs. 0.014; p-value = 0.0001) and DQB1*05:02 alleles (0.112 vs. 0.058; p-value = 0.0149) were higher in 57 patients with acquired hemophilia enrolled in the study compared to the normal European population. On the other hand, the HLA-DRB1*15 (0.087 vs. 0.172; p-value = 0.0260) and DQB1*06:02 alleles (0.078 vs. 0.142; p-value = 0.0149) were less frequent in the patients with acquired hemophilia. However, on comparing patients with acquired hemophilia but without inhibitors with those with inhibitors, the DRB1*16 and DQB1*05:02 alleles were correlated to lower risk (OR = 1.1 and 1.5, respectively) and the DRB1*15 and DQB1*06:02 alleles were considered high risk (OR = 2.2 and 3.7, respectively)(4242. Pavlova A, Zeitler H, Scharrer I, Brackmann HH, Oldenburg J. HLA genotype in patients with acquired haemophilia A. Haemophilia. 2010;16(102):107-12.).

Recently, an investigation conducted in southern Brazil evaluated the influence of HLA class I and class II alleles on the development of severe hemophilia in a group of 171 patients (50 patients with inhibitors and 131 without inhibitors). This study showed a high frequency of HLA-C*16 and HLA-DRB1*14 alleles in patients with inhibitors compared to those without inhibitors indicating a probable association between these alleles and the development of inhibitors. However, the difference in the frequency of the HLA-C*16 allele was not statistically significant compared to the healthy population in the region. The HLA-DRB1*14 allele is more common in white Brazilians than in Caucasians in general; this may be due to the great miscegenation of the population(4343. De Barros MF, Herrero JC, Sell AM, De Melo FC, Braga MA, Pelissari CB, et al. Influence of class I and II HLA alleles on inhibitor development in severe haemophilia A patients from the south of Brazil. Haemophilia. 2012;18(3):e236-40.).

Most studies have investigated alleles in respect to susceptibility to develop inhibitors. However, resistance alleles are also of great importance in association studies. For example, the DQA1*01:03, DQB1*06:03 and HLA-C*02 alleles have been reported as having a role in resistance to the development of inhibitors in hemophilia patients(3838. Hay CR, Ollier W, Pepper L, Cumming A, Keeney S, Goodeve AC, et al. HLA class II profile: a weak determinant of factor VIII inhibitor development in severe haemophilia A. UKHCDO Inhibitor Working Party. Thromb Haemost. 1997;77(2):234-7.,3939. Oldenburg J, Picard JK, Schwaab R, Brackmann HH, Tuddenham EG, Simpson E. HLA genotype of patients with severe haemophilia A due to intron 22 inversion with and without inhibitors of factor VIII. Thromb Haemost. 1997;77(2):238-242.).

HLA-C*05 is another allele that may provide protection against the formation of inhibitors. This was observed in an American population of 44 hemophilia patients, 28 of whom had no inhibitors and 16 were positive. HLA-C*05 was identified in 11 (39.29%) patients without inhibitors, whereas none in the group with inhibitors had this allele. Therefore, the presence of the HLA-C*05 acted as a protective factor in the development of inhibitors (p-value < 0.02)(4444. Aly AM, Aledort LM, Lee TD, Hoyer LW. Histocompatibility antigen patterns in haemophilic patients with factor VIII antibodies. Br J Haematol. 1990;76(2):238-41.).

Furthermore, Ohta et al. demonstrated a protective role of the HLA-A*24 allele. The HLA-A, B and C antigens and HLA-DQA1, DQB1, DRB1 and DPB1 alleles were analyzed in 20 Japanese hemophilia A patients with inhibitors. The results showed that the absence of the HLA-A*24 allele is a risk factor for the formation of inhibitors compared to its presence (36.8% vs. 82.6%; p-value = 0.003; OR = 0.123). Additionally, according to this study, the HLA-DRB1*04, DQB1*04 and DQA*03:01 alleles may be associated with the development of inhibitors(4545. Ohta H, Takahashi I, Kojima T, Takamatsu J, Shima M, Yoshioka A, et al. Histocompatibility antigens and alleles in Japanese haemophilia A patients with or without factor VIII antibodies. Tissue Antigens. 1999;54(1):91-7.).

Although some studies have shown that the HLA system, in particular HLA class II molecules, may be of greater importance in the development of inhibitors, the association between HLA and the formation of FVIII inhibitors varies between different ethnic groups and depends on the geographic region. These data may be useful in the recognition of groups at high risk for the formation of inhibitors in different populations. Table 1 shows some association studies of HLA class I and class II alleles for risks or protection to the development of inhibitors in hemophilia patients.

Table 1
Analysis of HLA alleles class I and II associated to the development of inhibitors

Polymorphisms of cytokine genes

Cytokines are a group of soluble proteins produced by different cells in response to antigens, which act regulating the innate and adaptive immune reactions. About 180 genes encode these proteins(4646. Abbas AK, Lichtman AH, Pillai S. Cellular and molecular immunology. 7th ed. Rio de Janeiro: Elsevier; 2012.).

Single nucleotide polymorphisms (SNPs) in the regulatory regions of cytokine genes, common in many populations, can affect the transcription and influence the production of cytokines and consequently, change the profile of the immune response(4747. Visentainer JE, Sell AM, da Silva GC, Cavichioli AD, Franceschi DS, Lieber SR, et al. TNF, IFNG, IL6, IL10 and TGFB1 gene polymorphisms in South and Southeast Brazil. Int J Immunogenet. 2008;35(4-5):287-93.). Thus, any difference in the gene frequency of these cytokines in different populations may have clinical relevance and be important to obtain more specific genetic markers for diagnosis and prognosis(4747. Visentainer JE, Sell AM, da Silva GC, Cavichioli AD, Franceschi DS, Lieber SR, et al. TNF, IFNG, IL6, IL10 and TGFB1 gene polymorphisms in South and Southeast Brazil. Int J Immunogenet. 2008;35(4-5):287-93.,4848. Franceschi DA, Viel DO, Sell AM, Tsuneto LT, Visentainer JE. Otimização de metodologia PCR-SSP para identificação de polimorfismos genéticos de TNF e IL2. Rev Bras Hematol Hemoter. 2009;31(4):241-46.).

The SNPs in cytokine genes are known due to their association in several diseases(4949. Hollegaard MV, Bidwell JL. Cytokine gene polymorphism in human disease: on-line databases, Supplement 3. Genes Immun. 2006;7(4):269-76.,5050. Scheller J, Ohnesorge N, Rose-John S. Interleukin-6 trans-signalling in chronic inflammation and cancer. Scand J Immunol. 2006;63(5):321-9.), as well as their influence on the production of antibodies in autoimmune diseases(5151. Visentainer JE, Sell AM, Franceschi DA, Lieber SR, Souza CA. Importância de polimorfismos de genes reguladores de citocinas em transplantes de células progenitoras hematopoiéticas. Braz J Pharm Sci. 2008;44(4):739-48.). Most of the SNPs studied are in promoter regions and in the exon itself, or in microsatellites of intron regions(4848. Franceschi DA, Viel DO, Sell AM, Tsuneto LT, Visentainer JE. Otimização de metodologia PCR-SSP para identificação de polimorfismos genéticos de TNF e IL2. Rev Bras Hematol Hemoter. 2009;31(4):241-46.).

Several studies have reported associations of these SNPs with the development of inhibitors in hemophilia A patients (Table 2).

Table 2
Analysis of Single nucleotide polymorphisms in cytokine genes associated with the development of inhibitors in Hemophilia A

In a recent study of Chinese hemophilia patients, the -819T and -592A alleles of the IL10 gene were more frequently found in individuals with FVIII inhibitors. In addition, some haplotypes of this gene (TA at -819 position and CA and CC at position -592) indicate predisposition of hemophilia patients for developing inhibitors(5252. Lu Y, Ding Q, Dai J, Wang H, Wang X. Impact of polymorphisms in genes involved in autoimmune disease on inhibitor development in Chinese patients with haemophilia A. Thromb Haemost. 2012;107(1):30-6.).

Another cytokine, which also plays an important role in immune modulation in hemophilia patients, is the TNF. This cytokine has a potent pro-inflammatory action. The analysis of polymorphisms in four alleles of the TNF gene (-827C>T, -308G>A, -238A>G and 670A>G) of 164 hemophilia patients (124 severe, 26 moderate and 14 mild) identified an association between the -308A/A genotype and the formation of inhibitors. The -308A allele was identified in 46 (59.7%) of 77 patients with inhibitors and in 40 (46.0%) of 87 patients without inhibitors (p-value = 0.87; OR = 1.7). The association between the -308A/A genotype and the formation of inhibitors was also evident in the subgroup of patients (n = 124) with severe hemophilia (p-value < 0.001; OR = 19.2)(5353. Astermark J, Oldenburg J, Carlson J, Pavlova A, Kavakli K, Berntorp E, et al. Polymorphisms in the TNFA gene and the risk of inhibitor development in patients with hemophilia A. Blood. 2006;108(12):3739-45.).

These findings were also observed in other patient groups. The polymorphism in the -308 region of the TNF gene was correlated with the development of inhibitors. Individuals homozygous for the allele A present a higher risk of developing inhibitors compared to heterozygotes (OR = 7519; 95% CI: 3168-17.844). This relationship is also valid on analyzing severe hemophilia patients (OR = 8163; 95% CI: 2521-26.434)(5454. Zhang LL, Yu ZQ, Zhang W, Cao LJ, Su J, Bai X, et al. [Relationship between factor VIII inhibitor development and polymorphisms of TNFalpha and CTLA-4 gene in Chinese Han patients with hemophilia A]. Zhonghua Xue Ye Xue Za Zhi. 2011;32(3):168-72. Chinese).

Pavlova et al. also confirmed higher frequencies of the -308G>A polymorphism in the TNF gene of patients in Germany (0.22 vs. 0.13; OR = 1.80). The homozygous A/A genotype (OR = 4.7) was more pronounced in severe hemophilia patients with FVIII inhibitors. The same group of researchers found that the 1082G allele of the IL10 gene was more common in these patients (0.55 vs. 0.43; p-value = 0.008)(4040. Pavlova A, Delev D, Lacroix-Desmazes S, Schwaab R, Mende M, Fimmers R, et al. Impact of polymorphisms of the major histocompatibility complex class II, interleukin-10, tumor necrosis factor-alpha and cytotoxic T-lymphocyte antigen-4 genes on inhibitor development in severe hemophilia A. J Thromb Haemost. 2009;7(12):2006-15.).

These and other association studies using genetic targets have focused on finding new markers to try to offer better treatment options to patients and avoid complications. Polymorphisms that influence the Th1/Th2 response may be instrumental to genotypically classify patients and check the risk of developing inhibitors(5555. Lozier JN, Rosenberg PS, Goedert JJ, Menashe I. A case-control study reveals immunoregulatory gene haplotypes that influence inhibitor risk in severe haemophilia A. Haemophilia. 2011;17(4):641-9.). Hence, it is evident that polymorphisms in the TNF and IL10 genes are best characterized in respect to the development of inhibitors in hemophilia patients, and may be future candidate genes.

Conclusions

During the last few years, progress in the identification of determinants in the development of anti-FVIII alloantibodies has occurred. After analysis of different studies, several factors related to the appearance of hemophilia A were found that highlight immune response genes.

This review identifies several important points: the incidence of inhibitors is greater in individuals of African descendancy and in patients with a family history of the inhibitor. Furthermore, the risks of inhibitor development in patients with large deletions and nonsense mutations are higher than in patients with intron 22 inversions; these inversions are related to a higher risk of the development of inhibitors than in patients with intron 1 inversions and splice-site mutations. In addition, the risk of patients with small deletions and insertions and missense mutations is lower.

The majority of associations of immune response genes to the production of inhibitors in hemophilia patients are related to HLA class II alleles: HLA-DRB1*14, DRB1*15, HLA-DQB1*06:02, DQB1*06:03, HLA-DQA1*01:02 and DQA1*01:03 and the DRB1*15:01/DQB1*06:02 haplotype. Associations have also been found for some HLA class I alleles: HLA-A*24 and C*05.

In relation to cytokine genes, the IL10 -1082G, -819T, -592A alleles are related to increased risk for the production of inhibitors in hemophilia patients. TNF is another cytokine gene associated with the formation of inhibitors, specifically the genotype -308A/A.

This review intends to assist in the development of more targeted genetic association studies of hemophilia patients and immune system genes, and also to assist in the understanding of the participation of these genes in the formation of inhibitors.

The authors thank all the employees who participated in the review of the study. The manuscript was linguistically revised by Tania Mara de Oliveira.

References

  • 1
    Bolton-Maggs PH, Pasi KJ. Haemophilias A and B. Lancet. 2003;361(9371):1801-9.
  • 2
    Shen BW, Spiegel PC, Chang CH, Huh JW, Lee JS, Kim J, et al. The tertiary structure and domain organization of coagulation factor VIII. Blood. 2008;111(3):1240-7.
  • 3
    Giangrande PL. The molecular basis of hemophilia. In: Provan D, Gribben JG, editors. Molecular Hematology. 3rd ed. Oxford, UK: Wiley-Blackwell; 2008. p.184-92.
  • 4
    World Federation of Hemophilia. Report on the Annual Global Survey 2010 [Internet[. Montreal, Canada: WFH; 2011 [cited 2013 Jan 21]. Available from: http://www1.wfh.org/publications/files/pdf-1427.pdf
    » http://www1.wfh.org/publications/files/pdf-1427.pdf
  • 5
    Brasil. Ministério da Saúde. Secretaria de Atenção à Saúde. Departamento de Atenção Especializada. Manual de reabilitação na hemofilia [Internet]. Brasília: MS; 2011 [cited 2012 Jul 27]. Available from: http://portal.saude.gov.br/portal/arquivos/pdf/manual_de_reabilitacao_consulta_publica.pdf
    » http://portal.saude.gov.br/portal/arquivos/pdf/manual_de_reabilitacao_consulta_publica.pdf
  • 6
    Brown TM, Lee WC, Joshi AV, Pashos CL. Health-related quality of life and productivity impact in haemophilia patients with inhibitors. Haemophilia. 2009;15(4):911-7.
  • 7
    Astermark J. Prevention and prediction of inhibitor risk. Haemophilia. 2012;18(Suppl 4):38-42.
  • 8
    Fulcher CA, de Graaf Mahoney S, Zimmerman TS. FVIII inhibitor IgG subclass and FVIII polypeptide specificity determined by immunoblotting. Blood. 1987;69(5):1475-80.
  • 9
    Gouw SC, van den Berg HM, Oldenburg J, Astermark J, de Groot PG, Margaglione M, et al. F8 gene mutation type and inhibitor development in patients with severe hemophilia A: systematic review and meta-analysis. Blood. 2012;119(12):2922-34.
  • 10
    Wight J, Paisley S. The epidemiology of inhibitors in haemophilia A: a systematic review. Haemophilia. 2003;9(4):418-35. Comment in: Haemophilia. 2004;10(2):197-8.
  • 11
    DiMichele DM. Inhibitors in haemophilia: a primer. Haemophilia. 2000;6(Suppl 1):38-40.
  • 12
    André S, Meslier Y, Dimitrov JD, Repessé Y, Kaveri SV, Lacroix-Desmazes S, et al. A cellular viewpoint of anti-FVIII immune response in hemophilia A. Clin Rev Allergy Immunol. 2009;37(2):105-13.
  • 13
    Astermark J. Inhibitor development: patient-determined risk factors. Haemophilia. 2010;16(102):66-70.
  • 14
    White GC 2nd, Kempton CL, Grimsley A, Nielsen B, Roberts HR. Cellular immune responses in hemophilia: why do inhibitors develop in some, but not all hemophiliacs? J Thromb Haemost. 2005;3(8):1676-81.
  • 15
    Ghosh K, Shetty S. Immune response to FVIII in hemophilia A: an overview of risk factors. Clin Rev Allergy Immunol. 2009;37(2):58-66.
  • 16
    Chaves DG, Rodrigues CV. Development of factor VIII inhibitors in hemophilia A. Rev Bras Hematol Hemoter. 2009;31(5):384-90.
  • 17
    Hoyer LW. The incidence of factor VIII inhibitors in patients with severe hemophilia A. Adv Exp Med Biol. 1995;386:35-45.
  • 18
    Oldenburg J, Pavlova A. Genetic risk factors for inhibitors to factors VIII and IX. Haemophilia. 2006;12(Suppl 6):15-22.
  • 19
    Pratt KP, Thompson AR. B-cell and T-cell epitopes in anti-factor VIII immune responses. Clin Rev Allergy Immunol. 2009;37(2):80-95.
  • 20
    Gill JC. The role of genetics in inhibitor formation. Thromb Haemost. 1999;82(2):500-4.
  • 21
    Astermark J, Berntorp E, White GC, Kroner BL; MIBS Study Group. The Malmö International Brother Study (MIBS): further support for genetic predisposition to inhibitor development in hemophilia patients. Haemophilia. 2001;7(3):267-72.
  • 22
    Gitschier J, Wood WI, Goralka TM, Wion KL, Chen EY, Eaton DH, et al. Characterization of the human factor VIII gene. Nature. 1984;312(5992):326-30.
  • 23
    Castaldo G, D'Argenio V, Nardiello P, Zarrilli F, Sanna V, Rocino A, et al. Haemophilia A: molecular insights. Clin Chem Lab Med. 2007;45(4):450-61.
  • 24
    Scandella D, Mattingly M, de Graaf S, Fulcher CA. Localization of epitopes for human factor VIII inhibitor antibodies by immunoblotting and antibody neutralization. Blood. 1989;74(5):1618-26.
  • 25
    Scandella D, Kessler C, Esmon P, Hurst D, Courter S, Gomperts E, et al. Epitope specificity and functional characterization of factor VIII inhibitors. Adv Exp Med Biol. 1995;386:47-63.
  • 26
    Arai M, Scandella D, Hoyer LW. Molecular basis of factor VIII inhibition by human antibodies. Antibodies that bind to the factor VIII light chain prevent the interaction of factor VIII with phospholipid. J Clin Invest. 1989;83(6):1978-84.
  • 27
    Lenting PJ, van de Loo JW, Donath MJ, van Mourik JA, Mertens K. The sequence Glu1811-Lys1818 of human blood coagulation factor VIII comprises a binding site for activated factor IX. J Biol Chem. 1996;271(4):1935-40.
  • 28
    HAMSTeRS. The Haemophilia A Mutation, Structure, Test and Resource Site. [Internet]. London: University College London and Royal Free Hospital London; 2013 [cited 2013 April 16]. Available from: http://hadb.org.uk/
    » http://hadb.org.uk/
  • 29
    Astermark J, Oldenburg J, Escobar M, White GC 2nd, Berntorp E; Malmö International Brother Study group. The Malmo International Brother Study (MIBS). Genetic defects and inhibitor development in siblings with severe hemophilia A. Haematologica. 2005;90(7):924-31. Comment in: Haematologica. 2005;90(7):869A.
  • 30
    Albánez S, Ruiz-Sáez A, Boadas A, de Bosch N, Porco A. Identification of factor VIII gene mutations in patients with severe haemophilia A in Venezuela: identification of seven novel mutations. Haemophilia. 2011;17(5):e913-8.
  • 31
    Zimmermann MA, Oldenburg J, Müller CR, Rost S. Unusual genomic rearrangements in introns 1 and 22 of the F8 gene. Hamostaseologie. 2011;31(Suppl 1):S69-73.
  • 32
    Soares RP, Chamone DA, Bydlowski SP. Factor VIII gene inversions and polymorphisms in Brazilian patients with haemophilia A: carrier detection and prenatal diagnosis. Haemophilia. 2001;7(3):299-305.
  • 33
    Howell WM, Carter V, Clark B. The HLA system: immunobiology, HLA typing, antibody screening and crossmatching techniques. J Clin Pathol. 2010;63(5):387-90.
  • 34
    Magalhães PS, Böhlke M, Neubarth F. The Major Histocompatibility Complex (MHC): genetic codification, structural bases and clinical implications. Rev Med UCPEL. 2004;2(1):54-9.
  • 35
    Donadi EA. How to understand the nomenclature and the mechanisms involved on the association between histocompatibility antigens and alleles with disease. Medicina (Ribeirão Preto). 2000;33(1):7-18.
  • 36
    Meyer D, Thomson G. How selection shapes variation of the human major histocompatibility complex: a review. Ann Hum Genet. 2001;65(Pt 1):1-26.
  • 37
    Traherne JA. Human MHC architecture and evolution: implications for disease association studies. Int J Immunogenet. 2008;35(3):179-92.
  • 38
    Hay CR, Ollier W, Pepper L, Cumming A, Keeney S, Goodeve AC, et al. HLA class II profile: a weak determinant of factor VIII inhibitor development in severe haemophilia A. UKHCDO Inhibitor Working Party. Thromb Haemost. 1997;77(2):234-7.
  • 39
    Oldenburg J, Picard JK, Schwaab R, Brackmann HH, Tuddenham EG, Simpson E. HLA genotype of patients with severe haemophilia A due to intron 22 inversion with and without inhibitors of factor VIII. Thromb Haemost. 1997;77(2):238-242.
  • 40
    Pavlova A, Delev D, Lacroix-Desmazes S, Schwaab R, Mende M, Fimmers R, et al. Impact of polymorphisms of the major histocompatibility complex class II, interleukin-10, tumor necrosis factor-alpha and cytotoxic T-lymphocyte antigen-4 genes on inhibitor development in severe hemophilia A. J Thromb Haemost. 2009;7(12):2006-15.
  • 41
    Nathalang O, Sriwanitchrak P, Sasanakul W, Chuansumrit A. Association of HLA class II alleles and the occurrence of factor VIII inhibitor in Thai patients with hemophilia A. Turk J Hematol. 2012;29(1):34-9.
  • 42
    Pavlova A, Zeitler H, Scharrer I, Brackmann HH, Oldenburg J. HLA genotype in patients with acquired haemophilia A. Haemophilia. 2010;16(102):107-12.
  • 43
    De Barros MF, Herrero JC, Sell AM, De Melo FC, Braga MA, Pelissari CB, et al. Influence of class I and II HLA alleles on inhibitor development in severe haemophilia A patients from the south of Brazil. Haemophilia. 2012;18(3):e236-40.
  • 44
    Aly AM, Aledort LM, Lee TD, Hoyer LW. Histocompatibility antigen patterns in haemophilic patients with factor VIII antibodies. Br J Haematol. 1990;76(2):238-41.
  • 45
    Ohta H, Takahashi I, Kojima T, Takamatsu J, Shima M, Yoshioka A, et al. Histocompatibility antigens and alleles in Japanese haemophilia A patients with or without factor VIII antibodies. Tissue Antigens. 1999;54(1):91-7.
  • 46
    Abbas AK, Lichtman AH, Pillai S. Cellular and molecular immunology. 7th ed. Rio de Janeiro: Elsevier; 2012.
  • 47
    Visentainer JE, Sell AM, da Silva GC, Cavichioli AD, Franceschi DS, Lieber SR, et al. TNF, IFNG, IL6, IL10 and TGFB1 gene polymorphisms in South and Southeast Brazil. Int J Immunogenet. 2008;35(4-5):287-93.
  • 48
    Franceschi DA, Viel DO, Sell AM, Tsuneto LT, Visentainer JE. Otimização de metodologia PCR-SSP para identificação de polimorfismos genéticos de TNF e IL2. Rev Bras Hematol Hemoter. 2009;31(4):241-46.
  • 49
    Hollegaard MV, Bidwell JL. Cytokine gene polymorphism in human disease: on-line databases, Supplement 3. Genes Immun. 2006;7(4):269-76.
  • 50
    Scheller J, Ohnesorge N, Rose-John S. Interleukin-6 trans-signalling in chronic inflammation and cancer. Scand J Immunol. 2006;63(5):321-9.
  • 51
    Visentainer JE, Sell AM, Franceschi DA, Lieber SR, Souza CA. Importância de polimorfismos de genes reguladores de citocinas em transplantes de células progenitoras hematopoiéticas. Braz J Pharm Sci. 2008;44(4):739-48.
  • 52
    Lu Y, Ding Q, Dai J, Wang H, Wang X. Impact of polymorphisms in genes involved in autoimmune disease on inhibitor development in Chinese patients with haemophilia A. Thromb Haemost. 2012;107(1):30-6.
  • 53
    Astermark J, Oldenburg J, Carlson J, Pavlova A, Kavakli K, Berntorp E, et al. Polymorphisms in the TNFA gene and the risk of inhibitor development in patients with hemophilia A. Blood. 2006;108(12):3739-45.
  • 54
    Zhang LL, Yu ZQ, Zhang W, Cao LJ, Su J, Bai X, et al. [Relationship between factor VIII inhibitor development and polymorphisms of TNFalpha and CTLA-4 gene in Chinese Han patients with hemophilia A]. Zhonghua Xue Ye Xue Za Zhi. 2011;32(3):168-72. Chinese
  • 55
    Lozier JN, Rosenberg PS, Goedert JJ, Menashe I. A case-control study reveals immunoregulatory gene haplotypes that influence inhibitor risk in severe haemophilia A. Haemophilia. 2011;17(4):641-9.
  • 56
    Chaves D, Belisário A, Castro G, Santoro M, Rodrigues C. Analysis of cytokine genes polymorphism as markers for inhibitor development in haemophilia A. Int J Immunogenet. 2010;37(2):79-82.
  • 57
    Pinto P, Ghosh K, Shetty S. Immune regulatory gene polymorphisms as predisposing risk factors for the development of factor VIII inhibitors in Indian severe haemophilia A patients. Haemophilia. 2012;18(5):794-7.

Publication Dates

  • Publication in this collection
    2013

History

  • Received
    16 Apr 2013
  • Accepted
    05 July 2013
Associação Brasileira de Hematologia e Hemoterapia e Terapia Celular R. Dr. Diogo de Faria, 775 cj 114, 04037-002 São Paulo/SP/Brasil, Tel. (55 11) 2369-7767/2338-6764 - São Paulo - SP - Brazil
E-mail: secretaria@rbhh.org