Long-term support of hematopoiesis by a single stem cell clone in patients with paroxysmal nocturnal hemoglobinuria

Blood ◽  
2002 ◽  
Vol 99 (8) ◽  
pp. 2748-2751 ◽  
Author(s):  
Jun-ichi Nishimura ◽  
Toshiyuki Hirota ◽  
Yuzuru Kanakura ◽  
Takashi Machii ◽  
Takashi Kageyama ◽  
...  

Abstract Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired hematopoietic stem cell disorder characterized by clonal blood cells that are deficient in glycosylphosphatidylinositol-anchored proteins because of somatic mutations of the PIG-A gene. Many patients with PNH have more than one PNH clone, but it is unclear whether a single PNH clone remains dominant or minor clones eventually become dominant. Furthermore, it is unknown how many hematopoietic stem cells (HSCs) sustain hematopoiesis and how long a single HSC can support hematopoiesis in humans. To understand dynamics of HSCs, we reanalyzed the PIG-A gene mutations in 9 patients 6 to 10 years after the previous analyses. The proportion of affected peripheral blood polymorphonuclear cells (PMNs) in each patient was highly variable; it increased in 2 (from 50% and 65% to 98% and 97%, respectively), was stable in 4 (changed less than 20%), and diminished in 3 (94%, 99%, and 98% to 33%, 57%, and 43%, respectively) patients. The complexity of these results reflects the high variability of the clinical course of PNH. In all patients, the previously predominant clone was still present and dominant. Therefore, one stem cell clone can sustain hematopoiesis for 6 to 10 years in patients with PNH. Two patients whose affected PMNs decreased because of a decline of the predominant PNH clone and who have been followed up for 24 and 31 years now have an aplastic condition, suggesting that aplasia is a terminal feature of PNH.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2195-2195
Author(s):  
Sung-Eun Lee ◽  
Sung Soo Park ◽  
Young-Woo Jeon ◽  
Jae-Ho Yoon ◽  
Byung Sik Cho ◽  
...  

Abstract Background: Although recently, Eculizumab, humanized monoclonal antibody directed against complement component C5, has used increasingly for the patients with hemolytic paroxysmal nocturnal hemoglobinuria (PNH), allogeneic stem cell transplantation (allo-SCT) can be curative treatment option especially for PNH patients with combined aplastic anemia (AA). The aim of the present study was to evaluate long-term outcome of allo-SCT in patients with AA/PNH. In addition, patients with classic PNH who underwent allo-SCT in the pre-eculizumab era were also evaluated. Methods: Total of 33 patients with PNH clones underwent allogeneic SCT at our institution between Jan 1998 and Jan 2016. Among them, seven patients had classic PNH and 26 patients with cytopenia had AA/PNH (with bone marrow evidence of a concomitant AA). Results: There were 21 male and 12 female patients with a median age of 34 years (range, 13-56 years). Pre-transplant GPI-AP deficient neutrophils and erythrocytes were 5.6% (0-92) and 21% (0-98.5), respectively. Median white blood cell, absolute neutrophil count, hemoglobin, and platelet at transplant were 2.4×109/L, 0.8×109/L, 7.7 g/dL, and 27×109/L, respectively. Median LDH level was 727 U/L (232-7721 U/L) and 19 (58%) patients had LDH ≥1.5x upper limit of normal. Classic PNH (n=7) and AA/PNH [SAA (n=15), VSAA (n=9), or non-SAA (n=2)] received SCT from HLA-matched sibling (MSD, n=24), unrelated (URD, n=7), or haplo-identical donor (Haplo-SCT, n=2). Since 2003, the conditioning regimen for MSD-SCT was changed from Busulfex (12.8 mg/kg) + cyclophosphamide (CY, 120 mg/kg) to fludarabine (180 mg/m2) + CY (100 mg/kg) + rATG (10 mg/kg). The conditioning regimen for URD-SCT and Haplo-SCT were TBI (800 cGy) + CY (100-120 mg/kg) ± rATG (2.5 mg/kg) and TBI 600cGy + Fludarabine (150 mg/m2) + rATG (5 mg/kg), respectively. After a median follow-up of 57 months (range 6.0-151.3), the 5-year estimated OS rates were 87.9 ± 5.7%. Four patients died of treatment-related mortality (TRM), including acute GVHD (n=1), pneumonia (n = 2), and cerebral hemorrhage (n=1), respectively. Except one patient with early TRM, 32 patients engrafted. Two patients who experienced delayed graft-failure received second transplant and recovered. The cumulative incidence of acute GVHD (≥grade II) and chronic GVHD was 27.3 ± 7.9% and 18.7 ± 7.0%, respectively. Among 25 patients with available follow-up data, PNH clone disappeared at median 3.0 months (range 0.7-45.5) after SCT and reemerging of PNH clones was observed in two patients; one patient showed re-appearance of 2.6% GPI-negative neutrophils at 12 months without PNH symptoms, but disappeared again at 21 months. Another patient suffered from labile graft and received a booster with peripheral blood stem cells. Conclusion: This study showed that long-term transplant outcome in patients with AA/PNH were comparable to that of allogeneic SCT in SAA (the 3-year estimated OS rates were 92.7 and 89 % for MSD-SCT and URD-SCT, respectively) at our institution (ASH Annual Meeting Abstracts 2012;120:4151). Reduced-intensity conditioning regimen was sufficient for the eradication of PNH clone in allogeneic SCT. Therefore, application of allogeneic SCT should be considered in PNH patients with AA in case of availability of well matched donor. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1251-1251
Author(s):  
Miroslaw Markiewicz ◽  
Anna Koclega ◽  
Monika Dzierzak-Mietla ◽  
Patrycja Zielinska ◽  
Ewa Mendek-Czajkowska ◽  
...  

Abstract Introduction: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare acquired clonal abnormality of the hematopoietic stem cell caused by somatic mutation in the phosphatidylinositol glycan-class A (PIG-A) gene located on the short arm of the X chromosome. Cells with lack phosphatidylinositol glycoproteins are more sensitive to complement-mediated lysis. Despite the efficient symptomatic treatment of hemolytic PNH with eculizumab, allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only curative treatment of the disease, although outcomes presented in the past were controversial. Material and methods: We report 32 allo-HSCTs: 31 from MUD and 1 from MRD performed for PNH in 2004-2014. Median age of recipients was 28 years (range 20-55) and donors 33(19-53), median time from diagnosis to allo-HSCT was 18(2-307) months. Median size of PNH clone was 80% granulocytes (0.41%-98%). Indication for allo-HSCT was aplastic/hypoplastic bone marrow (15 pts), overlapping MDS (2 pts), severe course of PNH with hemolytic crises and transfusion-dependency without access to eculizumab (15 pts). Additional risk factors were Budd-Chiari syndrome and hepatosplenomegaly (1 pt), history of renal insufficiency requiring hemodialyses (2 pts) and chronic hepatitis B (1 pt). The preparative regimen consisted of treosulfan 3x14 g/m2 plus fludarabine 5x30 mg/m2 (25 pts) or treosulfan 2x10 g/m2 plus cyclophosphamide 4x40 mg/kg (7 pts). Standard GVHD prophylaxis consisted of cyclosporine-A, methotrexate and pre-transplant ATG or thymoglobulin in MUD-HSCT. 2 pts instead cyclosporine-A received mycophenolate mofetil and tacrolimus. Source of cells was bone marrow (12 pts) or peripheral blood (20 pts) with median 7.7x10(8)NC/kg, 5.3x10(6)CD34+cells/kg, 24.2x10(6)CD3+cells/kg. Myeloablation was complete in all pts with median 9 days (6-13) of absolute agranulocytosis <0.1 G/l. Median number of transfused RBC and platelets units was 8.5(1-16) and 8(3-18). 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(13-33), 17.5(11-39) and 19.5(11-34). Acute GVHD grade I,II and III was present in 14, 6 and 1 pt, limited chronic GVHD in 11 pts. LDH decreased by 77%(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 83% what was treated with donor lymphocytes infusion (DLI). 2 patients died, 1 previously hemodialysed pt died on day +102 in a consequence of nephrotoxicity complicating adenoviral/CMV hemorrhagic cystitis and second on day +56 because of severe pulmonary infection. Complications in survivors were FUO (7 pts), CMV reactivation (8), VOD (1), neurotoxicity (1), venal thrombosis (1), hemorrhagic cystitis (1) and mucositis (8). 30 pts (93.7%) are alive 42 months (1-85) post-transplant and are doing well without treatment. Complete disappearance of PNH clone was confirmed by flow cytometry in all surviving pts. Conclusions: Our results indicate, that allo-HSCT with treosulfan-based conditioning is effective and well tolerated curative therapy in PNH. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1048-1048
Author(s):  
Kazuhiko Ikeda ◽  
Tsutomu Shichishima ◽  
Yoshihiro Yamashita ◽  
Yukio Maruyama ◽  
Hiroyuki Mano

Abstract Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired clonal hematological disorder which is manifested by complement-mediated hemolysis, venous thrombosis, and bone marrow failure. Deficiencies of glycosylphosphatidylinositol (GPI)-anchored proteins, due to mutations in the phosphatidylinositol glycan-class A (PIG-A) gene, contribute to complement-mediated hemolysis and affect all hematopoietic lineages in PNH. However, it is unclear how a PNH clone with a PIG-A gene mutation expands in bone marrow. Although some genes, including the Wilms’ tumor gene (Shichishima et al, Blood, 2002), the early growth response gene, anti-apoptosis genes, and the gene localized at breakpoints of chromosome 12, have been reported as candidate genes that may associate with proliferations of a GPI-negative PNH clone, previous studies were not intended for hematopoietic stem cell, indicating that the differences in gene expressions between GPI-negative PNH clones and GPI-positive cells from PNH patients remain unclear at the level of hematopoietic stem cell. To identify genes contributing to the expansion of a PNH clone, here we compared the gene expression profiles between GPI-negative and GPI-positive fractions among AC133-positive hematopoietic stem cells (HSCs). By using the FACSVantage (Becton Dickinson, San Jose, CA) cell sorting system, both of CD59+AC133+ and CD59− AC133+ cells were purified from bone marrow mononuclear cells obtained from 11 individuals with PNH. Total RNA was isolated from each specimen with the use of RNeasy Mini column (Qiagen, Valencia, CA). The mRNA fractions were amplified, and were used to generate biotin-labeled cDNAs by the Ovation Biotin system (NuGEN Technologies, San Carlos, CA). The resultant cDNAs were hybridized with a high-density oligonucleotide microarray (HGU133A; Affymetrix, Santa Clara, CA). A total of &gt;22,000 probe sets (corresponding to &gt;14,000 human genes) were assayed in each experiment, and thier expression intensities were analyzed by GeneSpring 7.0 software (Silicon Genetics, Redwood, CA). Comparison between CD59-negative and CD59-positive HSCs has identified a number of genes, expression level of which was statistically different (t-test, P &lt;0.001) between the two fractions. Interestingly, one of the CD59− -specific genes isolated in our data set turned out to encode a key component of the proteasome complex. On the other hand, a set of transcriptional factors were specifically silenced in the CD59− HSCs. These data indicate that affected CD59-negative stem cells have a specific molecular signature which is distinct from that for the differentiation level-matched normal HSCs. Our data should pave a way toward the molecular understanding of PNH.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 804-804
Author(s):  
Wenyi Shen ◽  
Bartlomiej P. Przychodzen ◽  
Michael J. Clemente ◽  
Brittney Dienes ◽  
Tetsuichi Yoshisato ◽  
...  

Abstract While PNH is characterized by clonality, it has not been considered a malignant disorder. Nevertheless, the similarities to some forms of MDS are clearly apparent, and include clonal hematopoiesis with the prescence of a somatic mutation, persistence and expansion of an aberrant stem cell clone, and frequent antedescent aplastic anemia. Somatic PIG-A gene mutations, the hallmark of PNH, lead to a defective GPI-anchor biosynthesis with a resultant deficiency of the GPI-anchored proteins, and is believed to be responsible for an extrinsic growth advantage. While this scenario is plausible, our research indicates that intrinsic factors may also be involved. Such factors may include additional, secondary genetic events, such as somatic mutations, which may coexist with PIG-A mutations, suggesting that the clonal architecture of PNH is more complex. For the purpose of this project we hypothesized that the evolution of a PNH clone may be associated with additional mutational events. Our genetic analysis involved 50 patients with PNH: the average PNH clone size by flow cytometry was 76%, 19 of these patients have history of antecedent aplastic anemia. We first performed paired whole exome sequencing (WES) of sorted PNH and wild type cells in 12 PNH patients and confirmed 34 somatic events in PNH-derived DNA, including 19 missense, 4 nonsense, 8 frameshift and 3 splice site mutations (a total of 22 genes). An additional 38 cases were used to examine the prevalence of these mutations. We detected somatic PIGA mutations (5 SNVs and 8 indels) in 9/12 PNH fractions (1 negative case contained a 616 kb delXp22.2 microdeletion involving the PIGA locus). Deep sequencing demonstrates the presence two independent PIGA mutations in 1/3 of the patients; semisolid culture experiments followed by sequencing of single CFUs confirmed that 2 independent PNH clones were present. Most significantly, by WES we found and confirmed additional somatic mutations (other than PIG-A) in PNH clones, including TET2 (p.E1250X), MAGEC1 (p.C747Y), BRPF1 (p.N797S), KDM3B (p.L125I), STAC3 (p.F97V) and NTNG1 (p.P24S). In 38 PNH cases studied by deep NGS sequencing, additional 2 somatic homozygous JAK2 (p.V617F), TET2 (p.S1556fs), SUZ12 (intron 2 splice), DHX29 (K498N), MECOM (P18S), and BCOR (Q1606X) mutations were found. Using targeted deep NGS of individual colonies, clonal architecture was analyzed in 9/12 WES cases. Clonal analysis of these cases revealed that PIGA mutations were often acquired in a later stage (6/9) preceeded by mutations in other genes (including NTNG1, CELSR1, STAC3, TET2, SLC20A1). For instance, in one PNH case, the PNH ancestral event was a novel MAN1A2 mutation, which was followed by the appearance of subclonal PIGA mutations, thus creating 2 independent subclones. In another illustrative case, somatic SYNE2 and PEX14 gene mutations were the initial events, followed by a PIGA mutation and an additional subclonal FBN1 mutation. Several somatic mutations were present in both PNH and WT cells and thus likely predated PIGA mutations. These mutations included TET2, SUZ12 and JAK2. In one case we determined that mutant fractions for TET2 and STAC3 mutations were larger than the PIGA mutant fraction with the TET2 mutation also present in the PNH- fraction (CD59+), indicating that PNH, in this case, evolved after the TET2 mutation as a subclone. However, in another case, dysplastic changes were identified along with trisomy 8. FISH analysis resolved that trisomy 8 was only present in the PNH- fraction, suggesting that in this patient, PNH evolved independent of the acquisition of trisomy 8. In sum, using whole exome sequencing, targeted deep NGS sequencing and single colony sequencing, we found that PNH, analogous to myeloid neoplasia, has a complex clonal architecture. Furthermore, the PIG-A mutation is frequently not the sole genetic lesion. Additional somatic mutations may help to further clarify the mechanism of clonal expansions, persistence of the mutated PNH stem cell, clinical diversity of PNH, and distinct behavior of PNH clones in individual patients. Disclosures: Maciejewski: NIH: Research Funding; Aplastic anemia&MDS International Foundation: Research Funding. Makishima:AA & MDS international foundation: Research Funding; Scott Hamilton CARES grant: Research Funding.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 10-10
Author(s):  
Fangfei Chen ◽  
Bing Han ◽  
Jian Li

Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a disease presented with hemolysis, cytopenia and thrombosis. Apart from PIGA gene on hematopoietic stem cells which accounts for the glycosylphosphatidylinositol (GPI) anchor deficiency on the cell membrane, other mutations have also been detected in PNH through whole-exome sequencing (WES). However, the characteristics of mutations in patients with PNH and genes which may contribute to PNH clonal expansion have not been well defined. Methods: Peripheral blood samples were collected from 41 patients with PNH, among them samples from 6 patients were further separated into CD59- and CD59+ fractions by CD59 magnetic beads. Gene mutations were tested by whole-exome sequencing(WES). 178 genes commonly mutated in myeloid cancer were analyzed in the sequencing results, as well as their correlation with clinical indicators. Mutated genes correlated with cell proliferation were compared between sorted CD59+ and CD59- cells. Results: The most frequently mutated myeloid cancer-related genes were MAP3K4 and CSMD1 (12.2% respectively). Among them, RUNX1T1 mutation was found to be correlated with larger clone size, higher level of uncombined bilirubin, and lower level of hemoglobin (P&lt;0.05). No other correlation between clinical parameters and gene mutations were found. The proportion of mutations (DNMT3A、RUNX1、JAK2、JAK3、CSMD1) which have been shown to indicate poor outcome in patients with aplastic anemia decreased as PNH clone increased (p=0.026). Mutations related to cell proliferation tended to happen more frequently in CD59- fractions compared with CD59+ fractions of the same patient (P=0.062). Conclusions: Myeloid cancer-related mutations can be detected in patients with PNH with some correlation with clinical manifestations. Larger PNH clone may "save" patients from mutation indicating poor prognosis. CD59- fractions seemed to carry more proliferation related mutations, which may contribute to PNH clonal expansion. Disclosures No relevant conflicts of interest to declare.


2012 ◽  
Vol 2012 ◽  
pp. 1-5
Author(s):  
Radha Raghupathy ◽  
Olga Derman

Paroxysmal nocturnal hemoglobinuria is caused by expansion of a hematopoietic stem cell clone with an acquired somatic mutation in the PIG-A gene. This mutation aborts the synthesis and expression of the glycosylphosphatidylinositol anchor proteins CD55 and CD59 on the surface of blood cells, thereby making them more susceptible to complement-mediated damage. A spectrum of disorders occurs in PNH ranging from hemolytic anemia and thrombosis to myelodysplasia, aplastic anemia and, myeloid leukemias. Aplastic anemia is one of the most serious and life-threatening complications of PNH, and a PNH clone is found in almost a third of the cases of aplastic anemia. While allogeneic bone marrow transplantation and T cell immune suppression are effective treatments for aplastic anemia in PNH, these therapies have significant limitations. We report here the first case, to our knowledge, of PNH associated with aplastic anemia treated with the anti-CD20 monoclonal antibody rituximab, which was associated with a significant reduction in the size of the PNH clone and recovery of hematopoiesis. We suggest that this less toxic therapy may have a significant role to play in treatment of PNH associated with aplastic anemia.


Blood ◽  
1995 ◽  
Vol 86 (5) ◽  
pp. 1736-1739 ◽  
Author(s):  
P Pramoonjago ◽  
W Wanachiwanawin ◽  
S Chinprasertsak ◽  
K Pattanapanayasat ◽  
J Takeda ◽  
...  

Paroxysmal nocturnal hemoglobinuria (PNH) is a hematopoietic stem cell disorder characterized by clonal blood cells that are deficient in the surface expression of glycosylphosphatidylinositol (GPI)-anchored proteins. In the affected cells, the X-chromosomal gene PIG-A, which participates in biosynthesis of the GPI anchor, is somatically mutated. Analyses of Japanese, British, and American patients with PNH have shown somatic mutations of PIG-A in all of them, indicating that PIG-A is responsible for PNH in most, if not all, patients in those countries. Twenty-nine of the reported somatic mutations are small, mostly involving 1 or 2 bases, except for one with a 4-kb deletion. Here we describe an analysis of PIG-A in neutrophils from 14 patients from Thailand where PNH is thought to be more common. We found small somatic PIG-A mutations in all patients. These consisted of six single base deletions, one each of 2-, 3-, 5- and 10-base deletions, two single base insertions and two base substitutions. Thus, the small somatic mutation in the PIG-A gene is also responsible for PNH in Thailand. However, base substitutions were rarer (2 of 14) than in Japan (8 of 16), and deletions of multiple bases were more common, suggesting various causes of mutation.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5469-5469
Author(s):  
Gulsun Karasu ◽  
Yilmaz Ay ◽  
Sinan Akbayram ◽  
Suar Caki Kilic ◽  
Fügen Pekün ◽  
...  

Abstract Introduction Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired clonal hematopoietic stem cell disorder characterized by complement-mediated hemolysis, thrombosis, and bone marrow failure. The clinical manifestations of PNH are usually seen in adulthood and are very rarely reported in children. The experience with transplantation in the management of children with PNH is also very limited. Here in this paper, we report a child with PNHpresenting with an episode of hemolytic anemia who was treated successfully with hematopoietic stem cell transplantation (HSCT). Case Presentation An eleven-year-old patient presented with a history of fatigue. On physical examination, there was no lymphadenopathy, and liver and spleen were not palpable. Laboratory analysis revealed a hemoglobin of 5.7 g/dL, platelets of 124.000/mL, and white blood cells were 1420/ml. She had increased lactate dehydrogenase and reticulocyte count and decreased haptoglobulin level. Bone marrow biopsy showed erythroid hyperplasia and relative hypocellularity in myeloid cell lines. Chromosomal karyotyping of bone marrow cells was normal. To look for PNH, immunophenotyping was performed. Flow cytometric analysis showed a PNH clone within the RBCs, granulocytes and monocytes. These findings were consistent with a diagnosis of PNH and she was started on anti-complement therapy with eculizumab. Since matched sibling donor was available, she was referred to our center for transplantation. The conditioning regimen of HSCT consisted of fludarabine (40 mg/m2day -9 to -6) and busulphan (4 mg/kg, day -5 to -2) combined with anti-thymocyte globulin (ATG) (Fresenius, Munich, Germany) (5 mg/kg, day -2 to 0). GVHD prophylaxis consisted of metotrexate plus cyclosporin. The infusion of bone marrow stem cells contained 11.8x106 CD34+ cells/kg and 7.9 x108total nucleated cells/kg. The patient had no significant complications in the post-transplant period. Neutrophil and platelet engraftment occurred on posttransplant day 11 and day 18, respectively. She is alive and is doing well over 14 months following BMT. Recovery is complete with full donor chimerism and the eradication of PNH clone. Conclusion PNH can occur in children but is often misdiagnosed and mismanaged. Although with the advent of anti-complement therapy, pure hemolytic anemia is no longer a clear indication for HSCT in adult patient, it is not licensed for use in pediatrics. HSCT is the only curative option for patients with PNH and if suitable matched sibling donor is available, transplant should be considered. However, the experience with tranplantation is also very limited. This case is worth mentioning as it shows that busulphan, fludarabine and ATG can be safely and effectively used for conditioning in PNH in children. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document