Natural History of Paroxysmal Nocturnal Hemoglobinuria Clones In Patients Presenting as Aplastic Anemia

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4428-4428
Author(s):  
Jeffrey J Pu ◽  
Galina Mukhina ◽  
Hao Wang ◽  
William Savage ◽  
Robert A Brodsky

Abstract Abstract 4428 Introduction: Acquired aplastic anemia (AA) and paroxysmal nocturnal hemoglobinuria (PNH) are closely related bone marrow failure disorders. Most AA results from an autoimmune attack directed against hematopoietic stem/progenitor cells. PNH originates from a multipotent hematopoietic stem cell (HSC) that acquires a PIG-A mutation. The PIG-A gene mutation leads to glycosylphosphatidylinositol-anchor protein (GPI-AP) biosynthesis deficiency and subsequent hemolysis secondary to the absence of complement regulatory proteins (CD55 and CD59). Both PNH and AA can be cured by allogeneic bone marrow transplantation (alloBMT), but only a minority of patients is offered this approach due to the potential morbidity and mortality. AA can be treated with immunosuppressive therapy (IST) and PNH can be controlled by eculizumab. It has been estimated that more than 50% of AA patients harbor small, but expandable PNH populations at diagnosis. The natural history of PNH clones in AA patients following non-transplant therapy is not well studied. The purpose of this study is to determine the fate and clinical relevance of these PNH clones in patients with AA who did not receive an alloBMT. Patients and Method: Twenty-seven patients with AA and a detectable PNH clone were monitored for a median of 5.3 years (range,1.5 to 11.5 years). The PNH granulocyte clone sizes were measured using flow cytometry and analyzed via CellQuest software. PE-conjugated anti-CD15 and fluoresceinated aerolysin variant (FLAER) staining were used to define granulocytes and GPI-AP deficient cells respectively. Serum lactate dehydrogenase (LDH) value was used as a surrogate for monitoring hemolysis and 1.5× the upper limit of normal LDH value (330mg/dL) as a cut-off point to define clinically apparent hemolysis. A PNH size change <2.5% was considered as stable. Patients were treated with IST, HiCy, or both. Result: We found a linear relationship between PNH granulocyte clone size and LDH values (Pearson correlation coefficient=0.73; P<0.0001). A PNH clone size above 23% was the threshold to identify hemolysis as measured by LDH (ROC analysis with AUC=0.88). Higher LDH values over the period of follow-up were associated with larger PNH granulocyte clone size at diagnosis (P=0.03). Patients with small (≤15%) initial PNH granulocytes had lower LDH levels at 5 years after diagnosis (mean±SD: 236.9±109.9 vs 423.1±248.8; P=0.02), and were less likely to develop hemolysis (13.3% vs 55.6%, P=0.06) comparing to those with larger (>15%) initial PNH granulocytes. Of 9 patients who initially were treated with traditional IST (ATG, CsA, and prednisone), 7 did not respond to treatment and eventually received high-dose cyclophosphamide (HiCy) salvage therapy, 2 achieved a remission and did not require further treatment though one demonstrated PNH clone size expansion to 50% after 37 months. After HiCy salvage, all 7 patients became transfusion independent and 4 of them had no further PNH clone expansion. PNH clone expansion was observed in 7 of 9 patients at a median time of 3 (range: 2 to 87) months after treatment. Of 15 patients who received HiCy as initial therapy, 14 achieved remissions. Later expansion of PNH size was observed in 7 patients, of which 5 eventually required intermittent blood transfusion but only 1 developed symptomatic hemolysis necessitating eculizumab therapy. The median time to PNH granulocyte clone expansion after HiCy was 52 (range: 18 to 106) months. In 5 patients who received HiCy and then relapsed, their PNH clone size only increased (1±0.7)% in (71±31) months observation during post treatment remission; however, their PNH clone size increase accelerated to (38±14)% in (34±21) months after AA relapse (P=0.04). Two nSAA patients with an initial PNH clone size ≤15% spontaneously recovered hematopoiesis at 84 and 56 months respectively, neither had PNH clone size expansion. In this study, 25.9% patients kept a stable PNH size, 48.1% patients increased the size, and 26% patients decreased the size. The group with small initial PNH clone sizes (≤15%) was the most stable over time. Conclusion: The risk of developing clinically significant PNH over 10 years appears to be low in AA patients with PNH clones, especially for those with small initial PNH granulocyte clones (≤15%) and for those who maintain remission following therapy. Disclosures: No relevant conflicts of interest to declare.

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5016-5016
Author(s):  
Wenrui Yang ◽  
Xin Zhao ◽  
Guangxin Peng ◽  
Li Zhang ◽  
Liping Jing ◽  
...  

Aplastic anemia (AA) is an immune-mediated bone marrow failure, resulting in reduced number of hematopoietic stem and progenitor cells and pancytopenia. The presence of paroxysmal nocturnal hemoglobinuria (PNH) clone in AA usually suggests an immunopathogenesis in patients. However, when and how PNH clone emerge in AA is still unclear. Hepatitis associated aplastic anemia (HAAA) is a special variant of AA with a clear disease course and relatively explicit immune pathogenesis, thus serves as a good model to explore the emergence and expansion of PNH clone. To evaluate the frequency and clonal evolution of PNH clones in AA, we retrospectively analyzed the clinical data of 90 HAAA patients that were consecutively diagnosed between August 2006 and March 2018 in Blood Diseases Hospital, and we included 403 idiopathic AA (IAA) patients as control. PNH clones were detected in 8 HAAA patients (8.9%,8/90) at the time of diagnosis, compared to 18.1% (73/403) in IAA. Eight HAAA patients had PNH clone in granulocytes with a median clone size of 3.90% (1.09-12.33%), and 3 patients had PNH clone in erythrocytes (median 4.29%, range 2.99-10.8%). Only one HAAA patients (1/8, 12.5%) had a PNH clone larger than 10%, while 24 out of 73 IAA patients (32.9%) had larger PNH clones. Taken together, we observed a less frequent PNH clone with smaller clone size in HAAA patients, compared to that in IAAs. We next attempted to find out factors that associated with PNH clones. We first split patients with HAAA into two groups based on the length of disease history (≥3 mo and < 3mo). There were more patients carried PNH clone in HAAA with longer history (21.4%, 3/14) than patients with shorter history (6.6%, 5/76), in line with higher incidence of PNH clone in IAA patients who had longer disease history. Then we compared the PNH clone incidence between HAAA patients with higher absolute neutrophil counts (ANC, ≥0.2*109/L) and lower ANC (< 0.2*109/L). Interestingly, very few VSAA patients developed PNH clone (5%, 3/60), while 16.7% (5/30) of non-VSAA patients had PNH clone at diagnosis. We monitored the evolution of PNH clones after immunosuppressive therapy, and found increased incidence of PNH clone over time. The overall frequency of PNH clone in HAAA was 20.8% (15/72), which was comparable to that in IAA (27.8%, 112/403). Two thirds of those new PNH clones occurred in non-responders in HAAA. In conclusion, PNH clones are infrequent in HAAA compared to IAA at the time of diagnosis, but the overall frequency over time are comparable between the two groups of patients. In SAA/VSAA patients who are under the activated abnormal immunity, longer clinical course and relatively adequate residual hematopoietic cells serve as two important extrinsic factors for HSCs with PIGA-mutation to escape from immune attack and to expand. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4215-4215
Author(s):  
Sandra van Bijnen ◽  
Konnie Hebeda ◽  
Petra Muus

Abstract Abstract 4215 Introduction Paroxysmal Nocturnal Hemoglobinuria (PNH) is a disease of the hematopoietic stem cell (HSC) resulting in a clone of hematopoietic cells deficient in glycosyl phosphatidyl inositol anchored proteins. The clinical spectrum of PNH is highly variable with classical hemolytic PNH at one end, and PNH in association with aplastic anemia (AA/PNH) or other bone marrow failure states at the other end. It is still largely unknown what is causing these highly variable clinical presentations. Immune-mediated marrow failure has been suggested to contribute to the development of a PNH clone by selective damage to normal HSC. However, in classic PNH patients with no or only mild cytopenias, a role for immune mediated marrow failure is less obvious. No series of trephine biopsies has been previously documented of patients with PNH and AA/PNH to investigate the similarities and differences in these patients. Methods We have reviewed a series of trephine biopsies of 41 PNH patients at the time the PNH clone was first detected. The histology was compared of 27 patients with aplastic anemia and a PNH clone was compared to that of 14 patients with classic PNH. Age related cellularity, the ratio between myeloid and erythroid cells (ME ratio), and the presence of inflammatory cells (mast cells, lymphoid nodules and plasma cells) were evaluated. The relation with clinical and other laboratory parameters of PNH was established. Results Classic PNH patients showed a normal or hypercellular marrow in 79% of patients, whereas all AA/PNH patients showed a hypocellular marrow. Interestingly, a decreased myelopoiesis was observed not only in AA/PNH patients but also in 93% of classic PNH patients, despite normal absolute neutrophil counts (ANC ≥ 1,5 × 109/l) in 79% of these patients. The number of megakaryocytes was decreased in 29% of classic PNH patients although thrombocytopenia (< 150 × 109/l) was only present in 14% of the patients. Median PNH granulocyte clone size was 70% (range 8-95%) in classic PNH patients, whereas in AA/PNH patients this was only 10% (range 0.5-90%). PNH clones below 5% were exclusively detected in the AA/PNH group. Clinical or laboratory evidence of hemolysis was present in all classical PNH patients and in 52% of AA/PNH patients and correlated with PNH granulocyte clone size. Bone marrow iron stores were decreased in 71% of classic PNH patients. In contrast, increased iron stores were present in 63% of AA/PNH patients, probably reflecting their transfusion history. AA/PNH patients showed increased plasma cells in 15% of patients and lymphoid nodules in 37%, versus 0% and 11% in classic PNH. Increased mast cells (>2/high power field) were three times more frequent in AA/PNH (67%) than in PNH (21%). Conclusion Classic PNH patients were characterized by a more cellular bone marrow, increased erythropoiesis, larger PNH clones and clinically by less pronounced or absent peripheral cytopenias and more overt hemolysis. Decreased myelopoiesis and/or megakaryopoiesis was observed in both AA/PNH and classic PNH patients, even in the presence of normal peripheral blood counts, suggesting a role for bone marrow failure in classic PNH as well. More prominent inflammatory infiltrates were observed in AA/PNH patients compared to classical PNH patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3426-3426 ◽  
Author(s):  
Andrew Shih ◽  
Ian H. Chin-Yee ◽  
Ben Hedley ◽  
Mike Keeney ◽  
Richard A. Wells ◽  
...  

Abstract Abstract 3426 Introduction: Paroxysmal Nocturnal Hemoglobinuria (PNH) is a rare disorder due to a somatic mutation in the hematopoietic stem cell. The introduction of highly sensitive flow cytometric and aerolysin testing have shown the presence of PNH clones in patients with a variety of other hematological disorders such as aplastic anemia (AA) and myelodysplasic syndrome (MDS). It is hypothesized that patients with these disorders and PNH clones may share an immunologic basis for marrow failure with relative protection of the PNH clone, due to their lack of cell surface expression of immune accessory proteins. This is supported by the literature showing responsiveness in AA and MDS to immunosuppressive treatments. Preliminary results from a recent multicenter trial, EXPLORE, notes that PNH clones can be seen in 70% of AA and 55% of MDS patients, and therefore there may be utility in the general screening of all patients with bone marrow failure (BMF) syndromes. Furthermore, it has been suggested that the presence of PNH cells in MDS is a predictive biomarker that is clinically important for response to immunosuppressive therapy. Methods: Our retrospective cohort study in a tertiary care center used a high sensitivity RBC and FLAER assay to detect PNH clones as small as 0.01%. Of all patients screened with this method, those with bone marrow biopsy and aspirate proven MDS, AA, or other BMF syndromes (defined as unexplained cytopenias) were analysed. Results from PNH assays were compared to other clinical and laboratory parameters such as LDH. Results: Overall, 102 patients were initially screened over a 12 month period at our center. 30 patients were excluded as they did not have biopsy or aspirate proven MDS, AA, or other BMF syndromes. Of the remaining 72 patients, four patients were found to have PNH clones, where 2/51 had MDS (both RCMD, IPSS 0) [3.92%] and 2/4 had AA [50%]. The PNH clone sizes of these four patients were 0.01%, 0.01%, 0.02%, and 1.7%. None of the MDS patients with known recurrent karyotypic abnormalities had PNH clones present. Only one of the four patients had a markedly increased serum LDH level. Conclusions: Our retrospective study indicates much lower incidence of PNH clones in MDS patients or any patients with BMF syndromes when compared to the preliminary data from the EXPLORE trial. There is also significant disagreement in other smaller cohorts in regards to the incidence of PNH in AA and MDS. Screening for PNH clones in patients with bone marrow failure needs further study before adoption of widespread use. Disclosures: Keeney: Alexion Pharmaceuticals Canada Inc.: Consultancy, Membership on an entity's Board of Directors or advisory committees. Wells:Alexion Pharmaceuticals Canada Inc: Honoraria. Sutherland:Alexion Pharmaceuticals Canada Inc.: Consultancy, Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5072-5072
Author(s):  
Alexander Kulagin ◽  
Olesya Klimova ◽  
Alexey Dobronravov ◽  
Elena Pavlyuchenko ◽  
Elena Babenko ◽  
...  

Abstract Introduction: PNH is an orphan clonal hematopoietic stem cell disorder characterized by intravascular hemolysis, cytopenias and TE. TE is the leading cause of death in the natural course of PNH. The data on epidemiology, clinical characteristics and outcomes of TE are limited to a few studies and not available in the Russian population of patients with PNH. Methods: We conducted a retrospective and prospective analysis of TE in a large cohort of patients with classic PNH and hemolytic PNH associated with aplastic anemia (AA/PNH) in one reference center in Russia. Hemolytic PNH was defined as a granulocyte clone size ≥ 10% and lactate dehydrogenase (LDH) level ≥ 1.5 of upper limit of normal (ULN). The analysis included only the documented episodes of symptomatic TE. Cases of subclinical thrombosis detected using sensitive methods were not considered. Cumulative incidence of TE was calculated using unrelated to TE death and allogeneic HSCT as competing risks. Patients without TE were censored at the last follow-up or date of first treatment with eculizumab. Results: A total of 276 patients with classic PNH (n=141) and hPNH associated with aplastic anemia (n=135) were included in analysis between 1996 and 2016 (Table 1). Sixty seven patients (24.3%) developed a total of 118 TE (median, 1; range, 1-7) with the median time between PNH onset and first TE of 2.7 years (range, 0-29). Cumulative incidence of first TE reached 29.7% and 45.5% at 10 and 20 years respectively (Fig. 1). Recurrent TEs were documented in 30 of 67 patients (45%) with the cumulative incidence of second TE of 27.7% and 46.7% at 1 and 5 years respectively (Fig. 2). The median time between the initial TE and the first recurrence was 0.74 years (range, 0.05-10). Routine secondary anticoagulant prophylaxis carried out in 34 patients had no significant impact on 5-year cumulative incidence of recurrent TE (43% vs 50%, p=0.919). The proportion of TE(+) patients was significantly higher in the classical PNH compared to AA / PNH, but taking into account the duration of the disease, these differences were not found in the cumulative incidence analysis. TE occurred in venous (64% of patients), arterial (22%) and both venous and arterial sites (14%). The most common sites of venous TE were intra-abdominal veins (n=26, 22%), deep veins (n=22, 18.6%), superficial veins (n=16, 13.5%), pulmonary embolism (n=15, 12.7%) and cerebral veins (n=5, 4.2%). Arterial TE included TIA/ischemic stroke (n=23, 19.5%) and myocardial infarction (n=7, 5.9%). Patients with TE were also characterized by 2-fold increase in the incidence of acute kidney injury (AKIN) episodes, the presence of chronic kidney disease (CKD) ≥ 2 stage and pulmonary hypertension. Seventeen of 67 (25%) TE(+) patients died which resulted in 10-year overall survival of 66% (CI 95%, 51-82). The mortality rate was highest in eculizumab naïve cohort: 11 of 24 (46%) patients died and all deaths were directly caused by TEs. In contrast, among the 43 patients with a history of TE treated with eculizumab there was only one death related to the consequences of TE, but 5 patients died due to neutropenic infections (2), solid tumor (1), MDS/AML (1) and complications after HSCT (1). Conclusions: These data confirm the high incidence and poor prognosis of symptomatic TE in patients with natural history of PNH. TE are often associated with organ dysfunction (AKIN, CKD, pulmonary hypertension), and characterize the highest burden of disease. Early targeted control of intravascular hemolysis is required in all cases at high risk or history of TE in PNH. Disclosures Kulagin: Alexion: Speakers Bureau.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4866-4866 ◽  
Author(s):  
Igor Lisukov ◽  
Alexander Kulagin ◽  
Alexey Maschan ◽  
Elena Shilova ◽  
Kudrat Abdulkadyrov ◽  
...  

Abstract Introduction Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hematopoietic stem cell disease that can lead to life-threatening complications including thrombotic events (TE), chronic kidney disease (CKD) and pulmonary hypertension. An international PNH Registry was implemented in 2008 to enhance understanding of the natural history of PNH, to describe treatment outcomes, and to evaluate the long term safety of eculizumab in treated patients. Methods This Registry is a non-interventional, prospective, multicenter, observational study. All patients with a diagnosis of PNH (confirmed in accordance with international diagnostic guidelines) or a detected PNH clone are enrolled irrespective of age or therapy. Data on patient demographics, medical histories, disease characteristics and treatment are collected at enrollment, every 6 months thereafter and/or at discontinuation. Descriptive statistics are used to describe the data; n, median and range (min–max) for continuous variables and percentages for categorical parameters. Results As of May 1, 2013, the Registry has enrolled 248 patients from Russia, over 50% of whom have a history of aplastic anemia or other bone marrow disorder (BMD) (Table 1). Disease characteristics for the overall population and by clone size or LDH level are presented in table1.A total of 25 patients have received eculizumab and have available follow-up data after starting treatment; median (range) follow-up time 4.4 (0.3–8.3) months. Among the 11 patients treated with eculizumab and with available LDH levels, the median LDH ratio was 5.7 X ULN before treatment and 1.0 X ULN at last follow-up assessment. Conclusion Russian patients included in the International PNH Registry show broad ranges of age, clone size, and degrees of hemolysis. History of TE, impaired renal function, and signs of chronic hemolysis are present among these patients regardless of PNH clone size. History of TE was recorded more frequently in patients with PNH clone sizes ≥20%, and was also more frequent among patients with LDH levels ≥1.5 x ULN. Among patients treated with eculizumab there was a marked decrease in hemolysis (as measured by LDH levels). Disclosures: Lisukov: Alexion: Honoraria. Kulagin:Alexion: Honoraria. Shilova:Alexion: Honoraria. Afanasyev:Alexion: Honoraria.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5150-5150
Author(s):  
Ugur Sahin ◽  
Mustafa Merter ◽  
Pinar Ataca ◽  
Erden Atilla ◽  
Berna Atesagaoglu ◽  
...  

Abstract Introduction: Flow cytometric assays (FCA), especially FLAER based methods, have become important recently in means of diagnosis of paroxysmal nocturnal hemoglobinuria (PNH). In this retrospective study, we aimed to describe diagnostic features of PNH patients along with disease characteristics. Methods: FCA results of patients ordered for PNH diagnosis between November 2007 and July 2014 were retrospectively evaluated. FCA results were expressed as percentage of granulocytes and monocytes negative for CD55/59. The FLAER/CD24-CD14 assay has been available in our center since December 2011 and used for confirmation of FCA results since then. The distribution of erythrocyte types according to CD59 expression (type 1, 2 and 3) were also given in percentages. Results: FCA for PNH diagnosis was performed for a total of 175 patients. FLAER method was available in 136 of these assays (77.7%). A PNH clone was not detected in 136 (77.7%) of the FCAs. PNH clone free patients were diagnosed with unclassified anemia (n=34, 19.4%), other unclassified cytopenias (n=30, 17.2%), non-PNH hemolytic anemias (n=7, 4.0%), myelodysplastic syndrome (n=23, 13.1%), aplastic anemia (n=9, 5.1%), primary myelofibrosis (n=1, 0.6%), leukemia (n=7, 4.0%), lymphoma (n=3, 1.7%), thrombotic incidents (n=14, 8.0%), rheumatological disorders (n=4, 2.3%) and unknown diagnosis (n=4, 2.3%). A PNH clone was detected in 39 patients (22.3%). Of these, 27 (69.2%) constituted classical PNH and 12 (30.8%) PNH accompanying other bone marrow disorders. In the PNH clone positive group, the accompanying bone marrow disorders were aplastic anemia (n=9) and myelodysplastic syndrome (n=3). Median age at diagnosis was 31 (10-82) for classical PNH and 35 (18-77) for PNH accompanying other bone marrow disorders. Erythrocyte transfusion requirement was prominent in 48.7% (n=19) of the patients and thrombotic incidents occured in 20.5% (n=8). Leukemic transformation did not take place in any of the patients. In one patient with aplastic anemia, the FCA became positive for PNH clone within one year of follow-up. Among classical PNH patients, thrombosis was observed only among patients with a clone size of > 50%. Clone size (both for granulocyte and monocyte clones) correlated with corrected reticulocyte and serum lactate dehydrogenase levels (for granulocytes p=0.001 and p<0.001, for monocytes p=0.001 and p<0.001, respectively). Treatment modalities of the PNH clone positive patients included eculizumab (n=12, 30.8%), allogeneic stem cell transplantation (n=13, 33.3%) and none/other (CsA, steroids, ATG, danazol) (n=16, 41.0%). Conclusion: FCA is an important modality for the diagnosis and follow-up of PNH patients. Clone size might be used to assess the severity of the disease and the risk of thrombosis in classical PNH. In patients with PNH accompanying other bone marrow disorders clone sizes are generally small and complications seen in classical PNH are rare. However, aplastic anemia and myelodysplastic syndrome might evolve into PNH during the disease course, which is reflected as an increase in clone size. Thus, FCA might be used in selected patients with aplastic anemia and myelodysplastic syndrome, especially when treatment failure or unexpected complications are suspected. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1976 ◽  
Vol 47 (4) ◽  
pp. 611-619 ◽  
Author(s):  
J Whang-Peng ◽  
T Knutsen ◽  
EC Lee ◽  
B Leventhal

Abstract Cytogenetic studies showed both 45XO and 46XY clones in the bone marrow of a 76-yr-old male with a 17-yr history of paroxysmal nocturnal hemoglobinuria (PNH). 55Fe incorporation studies demonstrated that both clones involved the hematopoietic stem cells. The loss of the Y chromosome may reflect an aging phenomenon, rather than be related to the PNH.


Blood ◽  
1985 ◽  
Vol 66 (6) ◽  
pp. 1247-1250 ◽  
Author(s):  
JH Antin ◽  
D Ginsburg ◽  
BR Smith ◽  
DG Nathan ◽  
SH Orkin ◽  
...  

Abstract Paroxysmal nocturnal hemoglobinuria (PNH) involves the proliferation of an abnormal and possibly premalignant hematopoietic stem cell. Successful treatment of PNH by marrow grafting requires that the PNH clone be eradicated by the pretransplant conditioning regimen. Four patients with PNH-associated marrow aplasia were transplanted with marrow from their HLA-matched, MLR-nonreactive siblings. Three patients were conditioned with cyclophosphamide, procarbazine, and antithymocyte serum (CTX/PCZ/ATS), and one was conditioned with busulfan/CTX/PCZ/ATS. Persistent complete engraftment of myeloid, lymphoid, and erythroid cell lines was demonstrated in all four patients by DNA sequence polymorphism analysis or cytogenetics, and RBC typing. There was no recurrence of the abnormal clone of cells for up to five years after transplantation despite the use of a conditioning regimen in three of them, which is not usually associated with permanent marrow aplasia. Bone marrow transplantation is a curative therapy in patients whose illness is severe enough to warrant the risk.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4213-4213
Author(s):  
Blanche P. Alter ◽  
Neelam Giri ◽  
William Hogan ◽  
Monique Johnson ◽  
Susan Olson ◽  
...  

Abstract Abstract 4213 INTRODUCTION Fanconi Anemia (FA) is a primarily autosomal recessive disorder with a defective DNA repair pathway associated with mutations in any of 13 genes. The majority of patients reported in the literature have one or multiple congenital anomalies, including low birth weight, short stature, café au lait spots, abnormal radii and/or thumbs, structural renal abnormalities, microcephaly, and deafness, among others. About 25% of reported patients had few or none of these findings. The hazard of severe aplastic anemia peaks at 10 years of age, and patients have very high risks of acute myeloid leukemia and specific solid tumors, such as head and neck and gynecologic squamous cell carcinomas. Only six patients have been reported who were diagnosed between 40 and 50 years of age. However, patients may present as adults with neoplasms, or remain asymptomatic and undiagnosed. METHODS We report the diagnosis of FA in the oldest known patient, an asymptomatic 55 year old woman (Case 2), identified only because she was a potential stem cell donor for her 42 year old sister (Case 1) with severe aplastic anemia. RESULTS Case 1 had a history of thrombocytopenia at 26, and anemia and thrombocytopenia during pregnancy; physical exam was normal except for a slightly deformed thumb; blood lymphocyte chromosome aberrations were increased with both diepoxybutane (DEB) and mitomycin C (MMC). The patient died following an HLA-matched bone marrow transplant from a non-FA brother. Case 2, the other of two siblings of Case 1 who were HLA matches, had higher than normal chromosome breakage in blood (3 cells with multiple radials with MMC) but not in the FA range; skin fibroblasts were diagnostic of FA, confirming hematopoietic somatic mosaicism. She had a normal appearance, a history of hypothyroidism and mitral valve prolapse, five pregnancies with five children (one miscarriage, one set of twins), a near normal blood count (Hb 13.3 g/dl, MCV 99 fl, WBC 3500/ul, and platelets 139,000/ul), and was a regular blood donor. Bone marrow cellularity and morphology were normal, but cytogenetics showed a small clone (46,XX,add(11)(q23)[6]/46,XX[14]). Complementation analysis of Case 1 indicated group A (FA-A), and molecular analysis identified two mutations in the FANCA gene. One mutation, p.S1208S (c.3624C>T) was a splice site mutation occurring in exon 36 and has been previously described. The second mutation was a novel nonsense mutation in exon 23, p.S674X (c.2021C>A). Five siblings had normal breakage results; four were heterozygous for the nonsense mutation and one was negative for both mutations. Case 2, with mosaicism for FA, had the familial splice mutation in both blood and fibroblasts, and the familial nonsense mutation in fibroblasts, but was skewed heavily toward wild-type in blood. cDNA studies confirmed that the predicted splice mutation created an alternate splice site resulting in multiple transcripts, including exon skipping, which varied in different tissues. The molecular mechanism for the loss of the nonsense mutation in the blood is most likely due to back mutation at a hot spot, which occurred in a hematopoietic stem cell which then had a selective growth advantage. CONCLUSIONS Reversion of one FANCA mutation probably occurred in a hematopoietic stem cell which was selected for and repopulated the peripheral blood. A plausible explanation for the lack of FA clinical features is the leaky splice mutation which may provide sufficient levels of protein for normal function in DNA repair. Family members should be tested for FA by chromosome breakage analysis in blood (and/or fibroblasts to identify those who may be mosaics). Those who have FA are at risk of syndrome-specific solid tumors, as well as aplastic anemia, myelodysplastic syndrome, and leukemia, if a non-gene corrected hematopoietic stem cell were to emerge. Even if asymptomatic, they should not be used as stem cell transplant donors for siblings with FA, because they may fail to repopulate the recipient marrow. FA is undoubtedly underdiagnosed in adults at this time. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5886-5886 ◽  
Author(s):  
Miroslaw Markiewicz ◽  
Malwina Rybicka-Ramos ◽  
Monika Dzierzak-Mietla ◽  
Anna Koclega ◽  
Krzysztof Bialas ◽  
...  

Abstract Introduction: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired clonal abnormality of hematopoietic stem cell leading to lack of phosphatidylinositol glycoproteins, sensitizing cells to complement-mediated lysis. Despite the efficient symptomatic treatment of hemolytic PNH with eculizumab, allo-HCT is the only curative treatment for the disease, although outcomes presented in the past were controversial. Material and methods: We report 41 allo-HCTs: 37 from MUD and 4 from MRD performed for PNH in 2004-2016. Median age of recipients was 29(20-62) years and donors 30(19-53), median time from diagnosis to allo-HCT was 16(2-307) months. Median size of PNH clone was 80% granulocytes (0.5%-100%). Indication for allo-HCT was PNH with aplastic/hypoplastic bone marrow (19 pts), MDS (2 pts), overlapping MDS/aplasia (3 pts), severe course of PNH with hemolytic crises and transfusion-dependency without access to eculizumab (17 pts). Additional risk factors were Budd-Chiari syndrome and hepatosplenomegaly (1 pt), history of renal insufficiency requiring hemodialyses (2 pts), chronic hepatitis B (1 pt) and C (1 pt). The preparative regimen consisted of treosulfan 3x14 g/m2 plus fludarabine 5x30 mg/m2 (31 pts) or treosulfan 2x10 g/m2 plus cyclophosphamide 4x40 mg/kg (10 pts). Standard GVHD prophylaxis consisted of cyclosporine-A, methotrexate and pre-transplant ATG in MUD-HCT. 2 pts instead of cyclosporine-A received mycophenolate mofetil and tacrolimus. Source of cells was bone marrow (13 pts) or peripheral blood (28 pts) with median 6.3x108NC/kg, 5.7x106CD34+cells/kg, 24.7x107CD3+cells/kg. Myeloablation was complete in all pts with median 9(1-20) days of absolute agranulocytosis <0.1 G/l. Median number of transfused RBC and platelets units was 9(0-16) and 8(2-18). Results: All pts engrafted, median counts of granulocytes 0.5 G/l, platelets 50 G/l and Hb 10 g/dl were achieved on days 17.5(10-33), 16(9-39) and 19.5(11-34). Acute GVHD grade I,II and III was present in 16, 7 and 3 pt, limited and extensive chronic GVHD respectively in 11 and 3 pts. LDH decreased by 73%(5%-91%) in first 30 days indicating disappearance of hemolysis. 100% donor chimerism was achieved in all pts. In 1 patient donor chimerism decreased to 81% what was treated with donor lymphocytes infusion (DLI). 3 patients died, 1 previously hemodialysed pt died on day +102 due to nephrotoxicity complicating adenoviral/CMV hemorrhagic cystitis, two other SAA patients with PNH clone<10% died on days +56 due to severe pulmonary infection and +114 due to aGvHD-III and multi organ failure. Complications in survivors were FUO (10 pts), CMV reactivation (13), VOD (1), neurotoxicity (1), venal thrombosis (1), hemorrhagic cystitis (4) and mucositis (8). 38 pts (92.7%) are alive 4.2 (0.4-12) years post-transplant and are doing well without treatment. Complete disappearance of PNH clone was confirmed by flow cytometry in all surviving pts. Conclusions: Allo-HCT with treosulfan-based conditioning is effective and well tolerated curative therapy for PNH. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document