New Insights in Bone Marrow Failure - European Medical Journal

New Insights in Bone Marrow Failure

8 Mins
Hematology
Download PDF
Chairperson:
Gérard Socié1
Speakers:
Austin Kulasekararaj,2 Gérard Socié,1 Alexander Röth3
Disclosure:

Prof Socié is a consultant for Alexion Pharmaceuticals. Dr Kulasekararaj has received funding and speaker’s honoraria from Alexion, Celgene, Novartis, and Amgen. Dr Röth has received honoraria, has membership of an entity’s board of directors and advisory committees, and has research funding from Alexion Pharmaceuticals.

Acknowledgements:

Writing assistance was provided by Dr Joan Thomas of ApotheCom.

Support:

The symposium and publication of this article was sponsored by Alexion Pharmaceuticals Inc. Authors received honoraria for preparation and delivery of their presentations. The views and opinions expressed are those of the authors and not necessarily Alexion Pharmaceuticals Inc.

Citation:
EMJ Hematol. ;4[1]:47-54. DOI/10.33590/emjhematol/10312028. https://doi.org/10.33590/emjhematol/10312028.

Each article is made available under the terms of the Creative Commons Attribution-Non Commercial 4.0 License.

Meeting Summary

Several rare haematological diseases are linked to bone marrow failure (BMF). This symposium provided the latest scientific insights into the different pathophysiological mechanisms and clinical advances in the management of these conditions, with a specific focus on the clinical management of patients with paroxysmal nocturnal haemoglobinuria (PNH) in the context of aplastic anaemia (AA), and the pathophysiology, consequences, and identification of PNH in the context of BMF.

Prof Gérard Socié chaired the symposium and overviewed BMF. Dr Austin Kulasekararaj gave a presentation on new paradigms in BMF, followed by Prof Gérard Socié, who reviewed the diagnosis and management of AA. Dr Alexander Röth then discussed the diagnosis and management of PNH in the context of BMF. The symposium was concluded by a short question and answer session.

Overview of Bone Marrow Failure

Professor Gérard Socié

Several different diseases overlap within BMF syndromes and share the propensity to develop into myelodysplastic syndromes (MDS) and acute myeloid leukaemia (AML) (Figure 1).1 For any BMF or AA, especially in children or young adults, a congenital disorder should be ruled out during diagnosis. Patients with congenital BMF will not respond to immunosuppressive therapy (IST).

Figure 1: Overlapping entities in bone marrow failure syndromes.1
AA: aplastic anaemia; PNH: paroxysmal nocturnal haemoglobinuria; FA: Fanconi anaemia; VSAA: very severe aplastic anaemia; SAA: severe aplastic anaemia; NSAA: non-severe aplastic anaemia; DC: dyskeratosis congenita; RBDS: ribosomal dysgenesis syndrome; 5q-: q arm of chromosome 5 deletion; MDS: myelodysplastic syndrome; MPD: myeloproliferative disorder; LGL: large granular lymphocytic leukaemia; AML: acute myeloid leukaemia.

Anaemia is the most common clinical sign of BMF, others include neutropenia, monocytopenia, and thrombocytopenia. Blood analysis is important for differential diagnosis to detect abnormalities in neutrophils, platelets, blast cells, and other cells. During bone marrow analysis mast cells and plasma cells are frequently detected in patients with severe AA, increased erythroblasts frequently detected in PNH, and dyserythropoiesis in both.

Bone marrow aspirate and trephine biopsy are fundamental to a differential diagnosis. However, it is important to consider that ageing is also associated with reduced bone marrow cellularity.2 Theoretically, distinguishing between AA and hypoplastic MDS is relatively simple, as dysplastic neutrophils and, in particular, dysplastic megakaryocytes are key features of hypoplastic MDS but not of AA.3 CD34+ cells may also be used. Cytogenetic analysis plays a key role in diagnosis, as hypocellular bone marrow can lead to insufficient cellular metaphase. Fluorescence in situ hybridisation for chromosomes 5, 8, and 7 should be considered for differential diagnosis. However, an abnormal cytogenetic clone does not imply a diagnosis of MDS, particularly as trisomy 8 is frequently detected in patients who are otherwise typical for AA. Flow cytometry is a key diagnostic tool for PNH and will be discussed in depth later. Other diagnostic tools may include screening for viral hepatitis or autoimmune disease (although cases linked to these diseases are rare) and vitamin B12 dosing.

Two rare diseases to consider in children, and adolescents and young adults, are Fanconi anaemia, which can easily be discounted using a chromosomal breakage test, and dyskeratosis congenita. Over recent years, the number of patients with a diagnosis of dyskeratosis congenita has increased, even though the disease development presents a wide spectrum of clinical features. A family history of pulmonary fibrosis, unknown pulmonary complications, or non-alcohol-related cirrhosis should be investigated, and dyskeratosis congenita has a frequent association with mutations of the telomerase RNA compartment (TERC) or telomerase reverse transcriptase (TERT). Despite the variety of tests available, in practice differential diagnosis between MDS and severe AA/PNH is not necessarily easy.

New Paradigms in Bone Marrow Failure

Doctor Austin Kulasekararaj

AA is an immune-mediated non-malignant disease, and clonal haematopoiesis in AA is linked to the evolution of disorders such as PNH and MDS/AML, which are associated with acquired genetic abnormalities.3 Recently, studies have identified inherited GATA2 mutations in patients presenting with AA, MDS, and also paediatric neutropenia, but invariably with severe monocytopenia.4

AA is associated with CD8+ autoreactive cytotoxic T cells which inhibit haematopoietic stem cells, leading to apoptosis and BMF; a significant reduction in CD4+ regulatory T cells, which are also non-functional and have an abnormal cytokine profile; and an expansion of CD4+ T helper (Th) 1 and Th17 cells.5-7 These anomalies can be identified by routine flow cytometry or cytokine profiling. Recent studies using newer techniques such as single cell mass cytometry (also called Cy-TOF) have identified aberrant subsets of T regulatory cells with pro-inflammatory properties in AA that predict poor response to IST.8

PNH in the context of BMF is characterised by progressive expansion of one or more PNH stem cell clones (immunological escape clones), which are deficient (partial or complete) in the expression of glycosylphosphatidylinositol (GPI)-anchored proteins.9 Other examples of such escape clones include trisomy 8, which leads to expansion of Wilms’ tumour 1-specific cytotoxic CD8+ T cells,10 uniparental disomy on chromosome 6p, the human leukocyte antigen (HLA) locus, and del 13q, which has been increasingly associated with BMF with PNH clones and responsive to IST, particularly in the context of AA rather than MDS.11

Several somatic mutations have been identified in a subgroup of patients with AA that can predict malignant transformation to MDS and monosomy 7.12 In a study of 150 patients with AA and no morphological evidence of MDS, acquired somatic mutations were identified in approximately 20%.12 ASXL1 (8% of patients) and DNMT3A (5% of patients) were the most common mutations, while BCOR was present in 4% of patients. In this study, 41% of patients had <10% mutation clonal load.12 In patients with AA with a disease duration of >6 months, these somatic mutations were associated with a 40% risk of transformation to MDS versus 4% risk without the mutations.12 Similar findings have been reported by the National Institutes of Health (NIH), Cleveland, and Japanese groups.13 The acquisition of these mutations increases with age, with each mutation demonstrating a different pattern of change over time.13 Importantly, the identification of these mutations may allow prediction of response to IST.13 Interestingly, presence of the BCOR mutation seems to predict a better response to IST.12 Additionally, studies investigating the mutational landscape of PNH indicate a stepwise pattern of MDS-type mutations, with these mutations arising either as a sub-clone within the PIG-A mutant population or prior to PIG-A mutation.14

Telomere length/attrition is another important factor in transformation to MDS in AA.12,15 Patients with AA and somatic mutations have been shown to have reduced telomere length. In a study of 13 patients with severe AA, patients who evolved to MDS and AML had marked telomere attrition that preceded the emergence of monosomy 7.15 In a study of patients with refractory AA treated with eltrombopag (a small molecule agonist of the c-mpl [thrombopoietin] receptor), approximately 40% of patients showed an improved haematologic response; however, they also had an increased risk of clonal evolution to monosomy 7, with a shorter time to evolution.16

There are several different ways in which monosomy 7 can occur, including familial origin, myeloid neoplasia, following haematopoietic stem cell transplantation (HSCT), AA, and following treatment with granulocyte colony-stimulating factor.17 In a recently reported Phase I/II study, patients with telomere diseases were treated with the synthetic steroid danazol 800 mg/day for 24 months, with the aim of attenuating accelerated telomere attrition. After just 3 months of treatment, almost all patients had a gain in telomere length and 79% had haematological response. Adverse effects included elevated liver enzymes (41%) and cramps (33%).18

Reports of spontaneous remission in patients with PNH have been described in the literature.19,20 In the UK PNH Service, where 303 patients have been treated with eculizumab (a humanised monoclonal antibody to complement protein C5), five patients have stopped treatment due to spontaneous remission,21 although the recent case report indicates that there is no recovery of normal haematopoiesis.19

A Phase III trial (A Prospective Randomized Multicenter Study Comparing Horse Antithymocyte Globuline [hATG] + Cyclosporine A [CsA] With or Without Eltrombopag as Front-line Therapy for Severe Aplastic Anemia Patients; RACE) is in progress to evaluate the efficacy and safety of eltrombopag versus placebo combined with IST.

Diagnosis and Management of Patients with Aplastic Anaemia

Professor Gérard Socié

AA is characterised by pancytopenia and persistent unexplained marrow hypoplasia. There is no specific marker and the diagnosis is mainly consequent with the differential diagnosis of hypoplastic MDS.22 Careful blood analysis, bone marrow aspirate, and bone marrow biopsy are all important aspects in correct diagnosis.22 Differential diagnosis versus congenital disorders should always be considered, especially with regard to dyskeratosis congenita.23 Finally, differentiation from MDS should be considered, as outlined earlier in this report.

Severe AA is defined as hypocellularity <30% and at least two criteria from: <0.5×109/L polymorphonuclear neutrophils, <20×109/L platelets, and <20×109/L reticulocytes; very severe AA is defined as <0.2×109/L polymorphonuclear neutrophils.24 All patients with severe or very severe AA require treatment.

For patients <40 years of age with severe AA, HSCT is mandatory, as it has been shown to be effective with low treatment-related mortality. At a median follow-up of 73 months in 61 patients (mean age: 21 years) who underwent HSCT from an HLA-matched sibling donor after irradiation-based conditioning with cyclophosphamide and ATG, 6-year overall survival was 87% (95% confidence interval: 78–97).25 Only one patient developed secondary malignancy. Osteonecrosis (observed in 10 patients) remains a concern in this disease.25

In patients >40 years of age or without a sibling donor, IST with ATG plus cyclosporine is an effective alternative to HSCT and improves blood counts and survival. A Phase III randomised trial comparing hATG and rabbit ATG (rATG) in 60 patients with severe AA showed that hATG was superior to rATG as a first-line treatment for severe AA. At 6-month follow-up, the haematologic response was 68% for patients treated with hATG versus 37% with rATG (p<0.001). Overall survival at Day 800 was 86% in the hATG group versus 68% in the rATG group (p=0.009). Transplant-free survival was 52% for rATG versus 76% for hATG (p=0.002).26 Similar results were demonstrated in a Phase II study conducted in 35 patients with AA treated with rATG and compared with 105 age and disease severity-matched patients from the European Blood and Marrow Transplant (EBMT) registry.27 The ongoing RACE study (mentioned earlier) is also investigating hATG with or without eltrombopag as first-line therapy for severe AA.

In patients refractory to IST, the decision to transplant stem cells from unrelated donors often represents a dilemma for physicians. Evaluations of outcomes in patients <30 years of age without a sibling donor and refractory to IST demonstrate that survival after unrelated HSCT for severe AA has improved significantly over the past 15 years, due to better HLA matching and improved conditioning regimens.28 Similar improvements in survival rates have been shown in other studies in patients with severe AA and unrelated donors receiving fludarabine, cyclophosphamide, and ATG.29

For patients >30 years of age and refractory to IST and with no suitable unrelated donor, there are three options: second-line IST, androgens (20–30% response rate for each), or currently still an investigational drug, eltrombopag. Long-term follow-up of a cohort of 43 patients with severe AA treated with eltrombopag demonstrated durable tri and bi-lineage responses, with a 40% response rate at 3–4 months.16

In conclusion, in the diagnosis of AA, inherited AA and MDS must be excluded. First-line treatment remains IST or bone marrow transplant, according to the availability of a donor. In patients who are refractory to first-line therapy, HSCT from a matched unrelated donor may be used in patients <30 years of age, and in patients >30 years of age, eltrombopag, androgen, or second-line IST.

Diagnosis and Management of Paroxysmal Nocturnal Haemoglobinuria in the Context of Bone Marrow Failure

Doctor Alexander Röth

The classical clinical triad of PNH includes haemolytic anaemia, thrombophilia, and cytopenia. Haemoglobinuria is present in approximately one-third of patients at the time of diagnosis. All patients have some degree of BMF, from isolated thrombocytopenia to AA, which typically precedes PNH. Thromboembolic complications, usually involving the brain, liver, or abdomen, are the leading cause of morbidity and mortality in patients with PNH.30-32 PNH originates from a somatic mutation in the PIG-A gene in a HSC, which is essential for biosynthesis of GPI-anchors for proteins, two of which (CD55 and CD59) are complement regulatory proteins. Selection and expansion of the mutant stem cell are both necessary for clinical development of PNH, which may explain the rarity of this disease.33 The absence of CD55 and CD59 on affected red blood cells leads to the formation of a membrane attack complex, which activates complement-mediated haemolysis and the release of free haemoglobin.34 Large amounts of free haemoglobin causes depletion of nitric oxide. The resulting reduction of nitric oxide levels leads to the activation of haemostasis with thromboembolic complications and smooth muscle dystonias that occur in PNH, which manifest as abdominal pain, dysphagia, pulmonary and systemic hypertension, and erectile dysfunction caused by impaired regulation of smooth muscle contractions.35

Patients are typically diagnosed with PNH in their early 30s36 and 5-year mortality is 35%, despite best supportive care.19 Thrombosis occurs in 18–40% of patients with PNH and increases the risk of death 7-fold compared with patients without thrombosis.19,37 Studies have reported a 21% incidence of a thrombotic event prior to PNH diagnosis.19,38 Silent thromboembolic complications are also possible in PNH and may be severe. Evaluation of the entire blood vessel might be helpful for diagnosis and may be conducted using magnetic resonance angiography with the angiographic system for unlimited rolling fields of view (AngioSURF).39

Markers for risk of thrombosis include D-dimers and tissue factor microparticles, both of which may be elevated in patients with PNH.40-43 Patients with thrombocytopenia also have an elevated risk of thromboembolism.40 Data from the South Korean National PNH Registry identified several haemolysis and clinical symptoms associated with increased risk of thromboembolism.37 Patients with elevated haemolysis (lactate dehydrogenase [LDH] levels ≥1.5-times the upper limit of normal [ULN] at diagnosis) were at significantly higher risk for thromboembolism than patients with LDH <1.5-times ULN (odds ratio: 7.0; p=0.013). The combination of LDH ≥1.5-times ULN with the clinical symptoms of abdominal pain, chest pain, dyspnoea, or haemoglobinuria was associated with a greater increased risk for thromboembolism than elevated haemolysis or clinical symptoms alone.37

Flow cytometry is the gold standard for diagnosis of PNH.44 Detection of GPI-anchor proteins such as CD59 or CD55, or fluorescent aerolysin reagent (FLAER) on haematopoietic cells using monoclonal antibodies forms the basis of a specific PNH diagnostic test.44,45 Identification of FLAER may be particularly useful as it selectively binds to the glycan core of GPI.46 Patients at risk who should be tested for PNH include those with haemolysis (e.g. Coombs negative haemolytic anaemia, haemoglobinuria, or renal dysfunction), BMF (e.g. AA, MDS, or cytopenia), and patients with unexplained thrombosis.44,47

Supportive care for patients with PNH may include blood transfusion, folic acid and vitamin B12 supplementation, oral iron supplementation (if the patient is iron deficient), early treatment of bacterial infections with antibiotics, hydration (in critical haemolysis), and anticoagulants (although these may be non-effective in many patients).44,48-50 Supportive care however, does not affect disease progression and may be associated with additional adverse risk factors.

The monoclonal antibody eculizumab blocks the terminal complement cascade, eliminates the formation of the membrane attack complex, and prevents cell lysis.51,52 In clinical trials, eculizumab improves survival and has demonstrated significant efficacy in reducing haemolysis, the need for blood transfusions, and the risk of thromboembolic complications versus controls. Patients treated with eculizumab also had reduced levels of fatigue, independent of haemoglobin levels, and a lower incidence of renal complications.30,38,53-59 Eculizumab is indicated in adults and children for the treatment of patients with PNH and atypical haemolytic uraemic syndrome (aHUS).29 In patients ≥40 kg, the recommended dose of eculizumab is 900 mg every 14±2 days.30,60 Patients on eculizumab are susceptible to meningococcal infections, and vaccination with Meningococcal Group A, C, W135, Y conjugate vaccine (e.g. Menveo®) followed by Meningococcal Group B Vaccine (Bexsero®) should be considered.58 Predictors of response to eculizumab include the degree of underlying BMF, concurrent inflammatory conditions such as bacterial and viral infections that can cause breakthrough haemolysis and autoimmune disease such as Crohn’s disease, genetic factors such as missense C5 heterozygous mutation, and polymorphisms of CR1 that lead to a suboptimal response due to extravascular haemolysis. Figure 2 shows a proposed treatment algorithm for PNH; in the haemolytic setting, symptomatic patients can be treated with eculizumab in combination with supportive therapy.61

Figure 2: Proposed treatment algorithm for paroxysmal nocturnal haemoglobinuria.61
TE: thromboembolic event; W&W: wait and watch.

In a recently published case-report, a 64-year old female with PNH, thromboembolic complications, and subsequent transition to severe AA was treated with IST (hATG + cyclosporin A) in combination with eculizumab. No reduced ATG efficacy or severe adverse events were observed. Re-occurrence of PNH symptoms was prevented, while T cell depletion was similar to non-eculizumab-treated patients, with partial remission being evident by Day 83.62

In conclusion, PNH is a chronic, life-threatening disease associated with chronic complement-mediated haemolysis. Thrombosis is multifactorial, may occur unexpectedly, and is the primary cause of death in patients with PNH. The gold standard test for PNH is high-sensitivity flow cytometry performed on peripheral blood. Terminal complement inhibition with eculizumab has become the standard of care for symptomatic PNH.

References
Young NS et al. Current concepts in the pathophysiology and treatment of aplastic anemia. Blood. 2006;15;108(8):2509-19. Ogawa T et al. Age-related changes of human bone marrow: a histometric estimation of proliferative cells, apoptotic cells, T cells, B cells and macrophages. Mech Ageing Dev. 2000;117(1-3):57-68. Barrett J et al. Myelodysplastic syndrome and aplastic anemia: distinct entities or diseases linked by a common pathophysiology? Semin Hematol. 2000;37(1):15-29. Ogawa S. Clonal hematopoiesis in acquired aplastic anemia. Blood. 2016. [Epub ahead of print]. Marsh J. ‘Aplastic anaemia and pure red cell aplasia’. Warrell D et al. (eds.) Oxford Textbook of Medicine. 2013. Oxford University Press. Solomou EE et al. Deficient CD4+ CD25+ FOXP3+ T regulatory cells in acquired aplastic anemia. Blood. 2007;110(5):1603-6. Kordasti S et al. Functional characterization of CD4+ T cells in aplastic anemia. Blood. 2012;119(9):2033-43. Kordasti S. In press. Araten DJ et al. Dynamics of hematopoiesis in paroxysmal nocturnal hemoglobinuria (PNH): no evidence for intrinsic growth advantage of PNH clones. Leukemia. 2002;16(11):2243-8. Sloand EM et al. Preferential suppression of trisomy 8 compared with normal hematopoietic cell growth by autologous lymphocytes in patients with trisomy 8 myelodysplastic syndrome. Blood. 2005;106(3):841-51. Hosokawa K et al. Favorable outcome of patients who have 13q deletion: a suggestion for revision of the WHO ‘MDS-U’ designation. Haematologica. 2012; 97(12):1845-9. Kulasekararaj AG et al. Somatic mutations identify a subgroup of aplastic anemia patients who progress to myelodysplastic syndrome. Blood. 2014; 124(17):2698-704. Yoshizato T et al. Somatic Mutations and Clonal Hematopoiesis in Aplastic Anemia. N Engl J Med. 2015;373(1):35-47. Shen W et al. Deep sequencing reveals stepwise mutation acquisition in paroxysmal nocturnal hemoglobinuria. J Clin Invest. 2014;124(10):4529-38. Dumitriu B et al. Telomere attrition and candidate gene mutations preceding monosomy 7 in aplastic anemia. Blood. 2015;125(4):706-9. Desmond R et al. Eltrombopag restores trilineage hematopoiesis in refractory severe aplastic anemia that can be sustained on discontinuation of drug. Blood. 2014;123(12):1818-25. Sloand EM et al. Granulocyte colony-stimulating factor preferentially stimulates proliferation of monosomy 7 cells bearing the isoform IV receptor. Proc Natl Acad Sci U S A. 2006;103(39):14483-8. Townsley DM et al. Danazol Treatment for Telomere Diseases. N Engl J Med. 2016;374(20):1922-31. Hillmen P et al. Natural history of paroxysmal nocturnal hemoglobinuria. N Engl J Med. 1995;333(19):1253-8. Babushok DV et al. Clonal Replacement Underlies Spontaneous Remission in Paroxysmal Nocturnal Haemoglobinuria. Br J Haematol. 2016. [Epub ahead of print]. Kulasekararaj AG. Personal communication. Killick SB et al. Guidelines for the diagnosis and management of adult aplastic anaemia. Br J Haematol. 2016; 172(2):187-207. Savage SA, Bertuch AA. The genetics and clinical manifestations of telomere biology disorders. Genet Med. 2010;12(12):753-64. Camitta BM et al. Severe aplastic anemia: a prospective study of the effect of early marrow transplantation on acute mortality. Blood. 1976;48(1):63-70. Konopacki J et al. Long-term follow up after allogeneic stem cell transplantation in patients with severe aplastic anemia after cyclophosphamide plus antithymocyte globulin conditioning. Haematologica. 2012;97(5):710-6. Scheinberg P et al. Horse versus rabbit antithymocyte globulin in acquired aplastic anemia. N Engl J Med. 2011; 365(5):430-8. Marsh JC et al. Prospective study of rabbit antithymocyte globulin and cyclosporine for aplastic anemia from the EBMT Severe Aplastic Anaemia Working Party. Blood. 2012;119(23):5391-6. Maury S et al. Unrelated stem cell transplantation for severe acquired aplastic anemia: improved outcome in the era of high-resolution HLA matching between donor and recipient. Haematologica. 2007;92(5):589-96. Bacigalupo A et al. Fludarabine, cyclophosphamide, antithymocyte globulin, with or without low dose total body irradiation, for alternative donor transplants, in acquired severe aplastic anemia: a retrospective study from the EBMT-SAA Working Party. Haematologica. 2010;95(6):976-82. Soliris®. Summary of Product Characteristics. Available at: https://www.medicines.org.uk/emc/medicine/19966. Last accessed: 20 July 2016. Brodsky RA. Advances in the diagnosis and therapy of paroxysmal nocturnal hemoglobinuria. Blood Rev. 2008;22(2):65-74. Röth A. Paroxysmale nächtliche Hämoglobinurie. Dtsch Arztebl. 2007;104(4):192-7. Inoue N et al. Molecular genetics of paroxysmal nocturnal hemoglobinuria. Int J Hematol. 2003;77(2):107-12. Rosse WF. New insights into paroxysmal nocturnal hemoglobinuria. Curr Opin Hematol. 2001;8(2):61-7. Rother RP et al. The clinical sequelae of intravascular hemolysis and extracellular plasma hemoglobin: a novel mechanism of human disease. JAMA. 2005;293(13):1653-62. Moyo VM et al. Natural history of paroxysmal nocturnal haemoglobinuria using modern diagnostic assays. Br J Haematol. 2004;126(1):133-8. Lee JW et al. Clinical signs and symptoms associated with increased risk for thrombosis in patients with paroxysmal nocturnal hemoglobinuria from a Korean Registry. Int J Hematol. 2013;97(6):749-57. Hillmen P et al. Effect of the complement inhibitor eculizumab on thromboembolism in patients with paroxysmal nocturnal hemoglobinuria. Blood. 2007;110(12):4123-8. Ruehm SG et al. [Whole-body MRA on a rolling table platform (AngioSURF)]. Rofo. 2000;172(8):670-4. Socie G. Terminal complement inhibitor eculizumab improves complement-mediated platelet consumption and thrombocytopenia in patients with paroxysmal nocturnal hemoglobinuria (PNH). Abstract 4030. 51st American Society of Hematology Annual Meeting, New Orleans, Louisiana, USA, 5-8 December 2009. Hill A et al. Recent developments in the understanding and management of paroxysmal nocturnal haemoglobinuria. Br J Haematol. 2007;137(3):181-92. Watt SG et al. Purpura fulminans in paroxysmal nocturnal haemoglobinuria. Br J Haematol. 2007;137(4):271. Helley D et al. Evaluation of hemostasis and endothelial function in patients with paroxysmal nocturnal hemoglobinuria receiving eculizumab. Haematologica. 2010;95(4):574-81. Parker C et al. Diagnosis and management of paroxysmal nocturnal hemoglobinuria. Blood. 2005;106(12):3699-709. Richards SJ et al. Application of flow cytometry to the diagnosis of paroxysmal nocturnal hemoglobinuria. Cytometry. 2000;42(4):223-33. Brodsky RA et al. Improved detection and characterization of paroxysmal nocturnal hemoglobinuria using fluorescent aerolysin. Am J Clin Pathol. 2000;114(3):459-66. Sharma VR. Paroxysmal nocturnal hemoglobinuria: pathogenesis, testing, and diagnosis. Clin Adv Hematol Oncol. 2013;11 Suppl 13(9):2-8. Marotta S et al. Hematopoietic stem cell transplantation for aplastic anemia and paroxysmal nocturnal hemoglobinuria: current evidence and recommendations. Expert Rev Hematol. 2014;7(6):775-89. Santarone S et al. Hematopoietic stem cell transplantation for paroxysmal nocturnal hemoglobinuria: long-term results of a retrospective study on behalf of the Gruppo Italiano Trapianto Midollo Osseo (GITMO). Haematologica. 2010;5(6):983-8. Bhatia S et al. Late mortality after allogeneic hematopoietic cell transplantation and functional status of long-term survivors: report from the Bone Marrow Transplant Survivor Study. Blood. 2007;110(10):3784-92. Rosse WF et al. Immune-mediated hemolytic anemia. Hematology Am Soc Hematol Educ Program. 2004:48-62. DeZern AE, Brodsky RA. Paroxysmal nocturnal hemoglobinuria: a complement-mediated hemolytic anemia. Hematol Oncol Clin North Am. 2015;29(3):479-94. Hillmen P et al. The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med. 2006;355(12):1233-43. Schubert J et al. Eculizumab, a terminal complement inhibitor, improves anaemia in patients with paroxysmal nocturnal haemoglobinuria. Br J Haematol. 2008;142(2):263-72. Brodsky RA et al. Multicenter phase 3 study of the complement inhibitor eculizumab for the treatment of patients with paroxysmal nocturnal hemoglobinuria. Blood. 2008;111(4):1840-7. Hillmen P et al. Long-term effect of the complement inhibitor eculizumab on kidney function in patients with paroxysmal nocturnal hemoglobinuria. Am J Hematol. 2010;85(8):553-9. de Latour RP et al. Paroxysmal nocturnal hemoglobinuria: natural history of disease subcategories. Blood. 2008;112(8):3099-106. Loschi M et al. Impact of eculizumab treatment on paroxysmal nocturnal hemoglobinuria: a treatment versus no-treatment study. Am J Hematol. 2016;91(4):366-70. Kelly RJ et al. Long-term treatment with eculizumab in paroxysmal nocturnal hemoglobinuria: sustained efficacy and improved survival. Blood. 2011;117(25):6786-92. Röth A, Dührsen U. Treatment of paroxysmal nocturnal hemoglobinuria in the era of eculizumab. Eur J Haematol. 2011;87(6):473-9. Schubert J. Paroxysmale nächtliche Hämoglobinurie (PNH). 2015. Available at: https://www.onkopedia.com/de/onkopedia/guidelines/paroxysmale-naechtliche-haemoglobinurie-pnh/@@view/html/index.html. Last accessed: 20 July 2016. Alashkar F et al. Horse anti-thymocyte globulin and eculizumab as concomitant therapeutic approach in an aplastic paroxysmal nocturnal hemoglobinuria patient: go or no-go? Eur J Haematol. 2016. [Epub ahead of print].

Please rate the quality of this content

As you found this content interesting...

Follow us on social media!

We are sorry that this content was not interesting for you!

Let us improve this content!

Tell us how we can improve this content?