21
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Atypical HUS caused by anti-complement factor H antibody: a hematologist's perspective

      letter

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          TO THE EDITOR: Hemolytic uremic syndrome (HUS) is characterized by a triad of non-immune hemolytic micro-angiopathic anemia, thrombocytopenia, and acute renal failure [1]. The majority of cases are "typical HUS," caused by an enteric infection of Shiga toxin-producing Escherichia coli. Rarely, however, HUS is caused by dysregulation of the alternative pathway of the complement system and is known as atypical HUS (aHUS). One rare subtype is caused by anti-complement factor H (CFH) antibodies. Such patients are commonly treated by nephrologists; however, they will usually present to a hematologist for variable cytopenias. We report here one such patient who presented to us primarily for cytopenias. CASE A 9-year-old girl presented to our hematology clinic complaining of vomiting for 15 days and generalized weakness and back and neck pain of similar duration. She also complained of vague abdominal pain. She had no fever or diarrhea immediately preceding the episode. There was no history of significant past illness. Her growth and development was normal for her age. She was born out of non-consanguineous marriage with no perinatal complications. She has one older brother who is apparently normal. On examination, she had significant pallor and icterus, without cyanosis or edema. On admission, her blood pressure was 118/78 mmHg, pulse 98 beats/min, respiratory rate 26 breaths/min. Abdomen was soft, nontender with normal bowel sounds. Liver was 3 cm palpable; there was no splenomegaly, lymphadenopathy, or skin rash. A summary of her laboratory parameters at admission is shown in Table 1. A peripheral smear examination showed 20 NRBCs/100 WBCs, polychromasia, anisopoikilocytosis and microspherocytes with presence of fragmented cells (Fig. 1). Differential count was normal with no atypical cells found. She was diagnosed with a case of thrombotic microangiopathy. Plasma exchange therapy was started. The laboratory parameters started improving after 1 day (Fig. 2). She responded to the therapy and her vomiting, abdominal pain, and icterus improved. Based on the clinical features, normal stool culture, and low C3 level, diagnosis of aHUS was considered and a genetic screen was performed for common mutations associated with aHUS. Results of the genetic screen are shown in Table 2. She was started on immunosuppressants with pulse cyclophosphamide and daily prednisolone. The patient is currently on a tapering dose of prednisolone. She has been doing well for the last 14 months, with no evidence of recurrence to date. DISCUSSION The patient presented with the classic triad of HUS. Although the most common cause of HUS in childhood is infection with a Shiga toxin-containing strain of E. coli, such an etiology was unlikely for this patient because of the absence of any suggestive history and a negative stool culture. We therefore considered aHUS, a genetic disorder in which 50-60% of cases are due to mutations in the alternate complement regulatory pathway. These mutations lead to endothelial damage and uncontrolled complement activation [2]. The disease is severe, and associated with poor prognosis and increased mortality. The most common mutation identified is in the gene for CFH (13%), a protein important in down-regulating the alternative complement pathway. Mutations in a variety of other genes for proteins that either regulate or are components of the alternative pathway may also cause aHUS, such as complement factor I (10%), membrane cofactor protein (11%), and thrombomodulin gene (4%) proteins. Importantly, in a minority of patients (5-10%), aHUS may develop due to auto-antibodies to CFH [3]. These antibodies interfere with binding of CFH to C3 convertase C3bBb, resulting in dysregulated complement activation. Anti-CFH-associated aHUS behaves differently clinically than the other aHUS subtypes, with the former frequently occurring in children less than 2 years of age, and the latter being more common in teenagers. Our patient was 9 years old at presentation. Additionally, clinical presentation of anti-CFH-associated aHUS is more aggressive, with a higher frequency of abdominal pain, vomiting, extra-renal complications, seizures, pancreatitis, and hepatitis [3, 4]. Our patient presented with abdominal pain and transaminasemia suggestive of hepatitis. Frequency of relapses is also higher with anti-CFH-associated aHUS when compared to most other types [5, 6]. Plasma therapy has been the cornerstone of aHUS therapy and was essentially the only therapy available until recently. Plasma therapy is effective due to its ability to deliver normal levels of complement factors and, when an exchange is done, to remove mutant complement factors and auto-antibodies, as in this case [7, 8]. It is recommended that plasma therapy be started within 24 hours of suspected diagnosis of aHUS. Efficacy of exchange should be monitored by daily creatinine, platelet count, LDH, and hemoglobin levels. Persistence of hemolysis or lack of improvement in thrombocytopenia after 3-5 days of plasma therapy should be considered no response to therapy, and an indication to stop plasma exchange and start complement inhibitor therapy with eculizumab. It is, however, important to note that patients with aHUS due to membrane cofactor protein mutations are unlikely to respond to plasma therapy because membrane cofactor protein is not a plasma protein. For anti-CFH-associated aHUS, plasma exchange can induce complete remission in 25%, and partial remission in another 50% of the patients [6]. There are reports that concomitant immunosuppression (with steroids, cyclophosphamide, rituximab, mycophenolate, or other drugs) along with plasma exchange can improve results [3, 7, 8]. The major limitation in the management of aHUS is the high recurrence rate and progression to chronic kidney failure in significant number of patients. Although kidney transplants have been tried, their efficacy was limited by the high recurrence rates in the transplanted kidneys. Eculizumab is a humanized monoclonal antibody to C5 that inhibits the terminal phase of complement activation. It has been approved by the American Food and Drug Administration for the treatment of paroxysmal nocturnal hemoglobinuria [9]. Subsequently, a few investigators have used eculizumab successfully in patients with aHUS, especially plasmapheresis-resistant cases [10, 11]. Since eculizumab acts by inhibiting the alternative pathway of complement, patients are at risk for meningococcal infections. Therefore, all patients treated with eculizumab should receive the meningococcal vaccine as well as prophylactic antibiotics for the first 2 weeks after immunization to protect them while their immune response develops. Despite its good efficacy in aHUS, data regarding its utility in anti-CFH-associated aHUS patients is sparse. In conclusion, this case highlights several points which clinicians would do well to remember: firstly, HUS must be strongly suspected in any patient who presents with non-specific abdominal or respiratory symptoms along with anemia and thrombocytopenia; secondly, the possibility of thrombotic microangiopathy should be investigated by using a battery of tests including reticulocyte count, LDH, and peripheral smear examination for schistocytes, and, if in doubt, confirmed with renal biopsy; thirdly, complement level should always be assessed before plasma therapy or exchange is initiated; and fourthly, in cases of suspected aHUS, samples should be sent to a reference laboratory to test for mutations.

          Related collections

          Most cited references9

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Atypical hemolytic uremic syndrome

          Hemolytic uremic syndrome (HUS) is defined by the triad of mechanical hemolytic anemia, thrombocytopenia and renal impairment. Atypical HUS (aHUS) defines non Shiga-toxin-HUS and even if some authors include secondary aHUS due to Streptococcus pneumoniae or other causes, aHUS designates a primary disease due to a disorder in complement alternative pathway regulation. Atypical HUS represents 5 -10% of HUS in children, but the majority of HUS in adults. The incidence of complement-aHUS is not known precisely. However, more than 1000 aHUS patients investigated for complement abnormalities have been reported. Onset is from the neonatal period to the adult age. Most patients present with hemolytic anemia, thrombocytopenia and renal failure and 20% have extra renal manifestations. Two to 10% die and one third progress to end-stage renal failure at first episode. Half of patients have relapses. Mutations in the genes encoding complement regulatory proteins factor H, membrane cofactor protein (MCP), factor I or thrombomodulin have been demonstrated in 20-30%, 5-15%, 4-10% and 3-5% of patients respectively, and mutations in the genes of C3 convertase proteins, C3 and factor B, in 2-10% and 1-4%. In addition, 6-10% of patients have anti-factor H antibodies. Diagnosis of aHUS relies on 1) No associated disease 2) No criteria for Shigatoxin-HUS (stool culture and PCR for Shiga-toxins; serology for anti-lipopolysaccharides antibodies) 3) No criteria for thrombotic thrombocytopenic purpura (serum ADAMTS 13 activity > 10%). Investigation of the complement system is required (C3, C4, factor H and factor I plasma concentration, MCP expression on leukocytes and anti-factor H antibodies; genetic screening to identify risk factors). The disease is familial in approximately 20% of pedigrees, with an autosomal recessive or dominant mode of transmission. As penetrance of the disease is 50%, genetic counseling is difficult. Plasmatherapy has been first line treatment until presently, without unquestionable demonstration of efficiency. There is a high risk of post-transplant recurrence, except in MCP-HUS. Case reports and two phase II trials show an impressive efficacy of the complement C5 blocker eculizumab, suggesting it will be the next standard of care. Except for patients treated by intensive plasmatherapy or eculizumab, the worst prognosis is in factor H-HUS, as mortality can reach 20% and 50% of survivors do not recover renal function. Half of factor I-HUS progress to end-stage renal failure. Conversely, most patients with MCP-HUS have preserved renal function. Anti-factor H antibodies-HUS has favourable outcome if treated early.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Genetics of HUS: the impact of MCP, CFH, and IF mutations on clinical presentation, response to treatment, and outcome.

            Hemolytic uremic syndrome (HUS) is a thrombotic microangiopathy with manifestations of hemolytic anemia, thrombocytopenia, and renal impairment. Genetic studies have shown that mutations in complement regulatory proteins predispose to non-Shiga toxin-associated HUS (non-Stx-HUS). We undertook genetic analysis on membrane cofactor protein (MCP), complement factor H (CFH), and factor I (IF) in 156 patients with non-Stx-HUS. Fourteen, 11, and 5 new mutational events were found in MCP, CFH, and IF, respectively. Mutation frequencies were 12.8%, 30.1%, and 4.5% for MCP, CFH, and IF, respectively. MCP mutations resulted in either reduced protein expression or impaired C3b binding capability. MCP-mutated patients had a better prognosis than CFH-mutated and nonmutated patients. In MCP-mutated patients, plasma treatment did not impact the outcome significantly: remission was achieved in around 90% of both plasma-treated and plasma-untreated acute episodes. Kidney transplantation outcome was favorable in patients with MCP mutations, whereas the outcome was poor in patients with CFH and IF mutations due to disease recurrence. This study documents that the presentation, the response to therapy, and the outcome of the disease are influenced by the genotype. Hopefully this will translate into improved management and therapy of patients and will provide the way to design tailored treatments.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Anti-Factor H autoantibodies associated with atypical hemolytic uremic syndrome.

              Several studies have demonstrated genetic predisposition in non-shigatoxin-associated hemolytic uremic syndrome (HUS), involving regulatory proteins of the complement alternative pathway: Factor H (FH) and membrane co-factor protein (CD46). Regarding the observations of thrombotic thrombocytopenic purpura patients, in whom a von Willebrand factor protease (ADAMST-13) deficiency may be inherited or acquired secondary to IgG antibodies, it was speculated that HUS might occur in a context of an autoimmune disease with the development of anti-FH antibodies leading to an acquired FH deficiency. The presence of FH autoantibodies was investigated by an ELISA method using coated purified human FH in a series of 48 children who presented with atypical HUS and were recruited from French university hospitals. Anti-FH IgG antibodies were detected in the plasma of three children who presented with recurrent HUS. The anti-FH specificity was conserved by the Fab'2 fraction. The plasma FH activity was found to be decreased, whereas plasma FH antigenic levels and FH gene analysis were normal, indicating that the presence of anti-FH antibodies led to an acquired functional FH deficiency. This report supports for the first time that HUS may occur in a context of an autoimmune disease with the development of anti-FH-specific antibody leading to an acquired FH deficiency. This new mechanism of functional FH deficiency may lead to the design of new approaches of diagnosis and treatment with a particular interest in plasma exchanges or immunosuppressive therapies.
                Bookmark

                Author and article information

                Journal
                Blood Res
                Blood Res
                BR
                Blood research
                Korean Society of Hematology; Korean Society of Blood and Marrow Transplantation; Korean Society of Pediatric Hematology-Oncology; Korean Society on Thrombosis and Hemostasis
                2287-979X
                2288-0011
                March 2015
                24 March 2015
                : 50
                : 1
                : 63-65
                Affiliations
                [1 ]Department of Hematology, NilRatan Sarkar (NRS) Medical College & Hospital, Kolkata, India.
                [2 ]Department of Nephrology, NilRatan Sarkar (NRS) Medical College & Hospital, Kolkata, India.
                Author notes
                Correspondence to Meet Kumar. Department of Hematology, NRS Medical College & Hospital, 138, AJC Bose Road, Kolkata 700014, India. kr_meet@ 123456yahoo.com
                Article
                10.5045/br.2015.50.1.63
                4377346
                c417f3a1-bfc7-41d9-9191-7c8cd18210c3
                © 2015 Korean Society of Hematology

                This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License ( http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 27 June 2014
                : 15 September 2014
                : 27 January 2015
                Categories
                Letter to the Editor

                Comments

                Comment on this article