Analysis of Platelets By Flow Cytometry in Patients with Paroxysmal Nocturnal Hemoglobinuria (PNH)

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1252-1252
Author(s):  
David Araten ◽  
Daniel Boxer ◽  
Michael A Nardi

Abstract Paroxysmal Nocturnal Hemoglobinuria (PNH) is characterized by a clonal population of hematopoietic stem cells with an acquired somatic mutation in the PIG-A gene, giving rise to populations of circulating mature cells that are unable to synthesize glycosylphosphatidylinositol (GPI). The disease is most readily diagnosed by flow cytometry analysis of red blood cells, using antibodies specific for the GPI-linked protein CD59, or analysis of granulocytes, using antibodies specific for the GPI-linked protein CD24, along with the FLAER reagent, a fluorescent protein that binds to the GPI structure and which is detected only on the surface of GPI (+) cells. However, other mature blood lineages can be derived from the PNH clone. Notably, thrombosis is a major life threatening complication of PNH and may be triggered by complement activation on platelets that belong to the GPI-negative stem cell clone. The PNH clone size generally predicts thrombosis, but sometimes the proportion of PNH red cells and granulocytes are highly discordant, in which case there might be a role for the determination of the proportion of PNH platelets. Historically, flow cytometry analysis of platelets in patients with PNH has been technically difficult. Here is described a method to do this that avoids technical challenges by using aspirin and sepharose gel filtration of platelets to prevent their activation as well as simultaneous determination of CD59 expression and uptake of the FLAER reagent. Red cells were analyzed based on CD59 expression and granulocytes based on CD24 and FLAER. We analyzed blood samples from 48 patients with PNH and or AA/PNH who provided informed consent, 16 of whom had a prior history of thrombosis. To separate platelet rich plasma (PRP), whole blood collected in EDTA tubes was centrifuged at 200g for 7 minutes at room temperature with the brake turned off. After this step, there was no further centrifugation or vortexing of the platelets. A solution of aspirin was made up immediately prior to use and was added to the PRP at a final concentration of 0.5mMolar. Aspirinated PRP was then loaded on top of a sepharose-2B column using Tyrode's buffer. The platelet-rich turbid drops were collected, to isolate platelets from red cells and coagulation proteins. 50 ul of platelet rich buffer was then incubated with FLAER-Alexa-488 (Pinewood, 1:20 dilution) and CD59-PE (Serotec, 1:10 dilution) in the dark for 30' at room temperature. To prevent doublet events from confounding the analysis, the platelet suspension was diluted 1:200 in Hanks with 0.1% BSA. The sample was passed through a 35 uM Falcon cell strainer, and platelets were identified by forward/side scatter acquired on a log-log scale on a BD Facscan. The median proportion of PNH red cells, granulocytes and platelets was 24%, 86%, 76% respectively in the group without a history of thrombosis and 23% ,82%, and 65% in the group with a history of thrombosis. The proportion of PNH platelets was highly correlated with the proportion of PNH granulocytes (r=0.84). In two patients with almost undetectable PNH red cells and over 90% PNH granulocytes, the proportion of PNH platelets was over 90%; both were on prophylaxis and neither had thrombosis. It is predicted that this technique may be useful for determining thrombosis risk, particularly when the results from the analysis of rbc's and granulocytes are discordant. Disclosures No relevant conflicts of interest to declare.

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.


2018 ◽  
Vol 150 (3) ◽  
pp. 273-282 ◽  
Author(s):  
Alina E Dulau-Florea ◽  
Neal S Young ◽  
Irina Maric ◽  
Katherine R Calvo ◽  
Cynthia E Dunbar ◽  
...  

Abstract Objectives To determine fluorescently labeled aerolysin (FLAER) binding and glycophosphatidylinositol–anchored protein expression in bone marrow (BM) cells of healthy volunteers and patients with paroxysmal nocturnal hemoglobinuria (PNH) detected in peripheral blood (PB); compare PNH clone size in BM and PB; and detect PNH in BM by commonly used antibodies. Methods Flow cytometry analysis of FLAER binding to leukocytes and expression of CD55/CD59 in erythrocytes. Analysis of CD16 in neutrophils and CD14 in monocytes in BM. Results FLAER binds to all normal BM leukocytes, and binding increases with cell maturation. In PNH, lymphocytic clones are consistently smaller than clones of other BM cells. PNH clones are detectable in mature BM leukocytes with high specificity and sensitivity using common antibodies. Conclusions PNH clone sizes measured in mature BM leukocytes and in PB are comparable, making BM suitable for PNH assessment. We further demonstrate that commonly used reagents (not FLAER or CD55/CD59) can reliably identify abnormalities of BM neutrophils and monocytes consistent with PNH cells.


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 ◽  
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.


2016 ◽  
Vol 14 (3) ◽  
pp. 366-373 ◽  
Author(s):  
Rodolfo Patussi Correia ◽  
Laiz Cameirão Bento ◽  
Ana Carolina Apelle Bortolucci ◽  
Anderson Marega Alexandre ◽  
Andressa da Costa Vaz ◽  
...  

ABSTRACT Objective: To discuss the implementation of technical advances in laboratory diagnosis and monitoring of paroxysmal nocturnal hemoglobinuria for validation of high-sensitivity flow cytometry protocols. Methods: A retrospective study based on analysis of laboratory data from 745 patient samples submitted to flow cytometry for diagnosis and/or monitoring of paroxysmal nocturnal hemoglobinuria. Results: Implementation of technical advances reduced test costs and improved flow cytometry resolution for paroxysmal nocturnal hemoglobinuria clone detection. Conclusion: High-sensitivity flow cytometry allowed more sensitive determination of paroxysmal nocturnal hemoglobinuria clone type and size, particularly in samples with small clones.


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.


2012 ◽  
Vol 83A (1) ◽  
pp. 27-37 ◽  
Author(s):  
Allison Mayle ◽  
Min Luo ◽  
Mira Jeong ◽  
Margaret A. Goodell

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3676-3676
Author(s):  
Kazuhiko Ikeda ◽  
Hideyoshi Noji ◽  
Masaki Yasukawa ◽  
Akiko Shichishima ◽  
Kazuko Akutsu ◽  
...  

Abstract It is unclear how a paroxysmal nocturnal hemoglobinuria (PNH) clone expands and bone marrow failure (BMF) occurs in PNH patients, although an immunologic mechanism by human leukocyte antigen (HLA)-restricted cytotoxic T lymphocytes (CTLs) has been suggested. It has been also reported that immunization with HLA-binding peptides of Wilms’ tumor gene (WT1) in hematopoietic cells induces a WT1 peptide-specific CTL response, and WT1 RNA is highly expressed in BM mononuclear cells (MNCs) in PNH patients (Shichishima T et al., Blood, 2002). In this study, to clarify some roles of WT1 peptide-specific and HLA-restricted CTLs, the frequencies of peripheral blood (PB) WT1 peptide-specific and HLA-A*2402-restricted CTLs by flow cytometric tetramer analysis and WT1 peptide-stimulated interferon (IFN)-γ-producing MNCs by enzyme-linked immunospot assay in 5 PNH patients with the HLA-A*2402 allele were examined. We also investigated cytotoxicity of WT1 peptide-specific and HLA-A*2402-restricted CTL clone (TAK-1) cells on BM MNCs by 51Cr-releasing assay, colony forming-unit granulocyte-macrophage colony formation of CD34+CD59+ and CD34+CD59− cells, and CD59 expression in viable 7AAD−CD34+ cells by flow cytometry in those patients, and expression of IFN-γ in TAK-1 cells by flow cytometry, after co-incubation of BM cells from them with TAK-1 cells. As controls, 8 healthy volunteers (HV) with the HLA-A*2402 allele and 2 PNH patients and HV without the allele were examined. We found that the frequencies of PB WT1 peptide-specific and HLA-A*2402-restricted CD8+ cells (p&lt;0.005) and WT1 peptide-stimulated IFN-γ-producing MNCs (p&lt;0.02) were significantly higher in PNH patients with the HLA-A*2402 allele (0.255 ± 0.164% and 25.2 ± 15.4 / 5 x 105 cells, respectively) than HV with the allele (0.052 ± 0.025% and 6.6 ± 6.8 / 5 x 105 cells, respectively). In PNH patients or HV, TAK-1 cells significantly killed BM MNCs, suppressed colony formations of CD34+CD59+ and CD34+CD59− cells, and expressed IFN-γ in the absence and presence of a WT1 peptide or only in the presence of the peptide, respectively, in an HLA-A*2402-restricted manner. Reduction rates of colony formation of CD34+CD59− cells from the patients with the HLA-A*2402 allele by TAK-1 cells were significantly less than those of CD34+CD59+ cells in PNH patients, in the absence (38.3 ± 23.0% and 59.0 ± 28.0%, respectively, p&lt;0.01) and presence (74.7 ± 12.8% and 90.6 ± 11.1%, respectively, p&lt;0.002) of a WT1 peptide. After co-incubation of BM MNCs from the patients with TAK-1 cells, proportions of viable CD34+CD59− cells from PNH patients significantly increased in the absence (62.87 ± 27.29%; p&lt;0.01) and presence (62.32 ± 25.73%; p&lt;0.01) of a WT1 peptide compared with those of the controls incubated without TAK-1 cells (52.40 ± 24.58%) in an HLA-A*2402-restricted manner. In conclusion, WT1 peptide-specific and HLA-restricted CTLs may play important roles in the expansion of a PNH clone during immunologic selection and in the occurrence of BMF via IFN-γ in PNH.


Sign in / Sign up

Export Citation Format

Share Document