Faculty Opinions recommendation of Treatment with the Bcl-xL inhibitor ABT-737 in combination with interferon α specifically targets JAK2V617F-positive polycythemia vera hematopoietic progenitor cells.

Author(s):  
Alessandro Vannucchi ◽  
Paola Guglielmelli
Blood ◽  
2012 ◽  
Vol 120 (15) ◽  
pp. 3098-3105 ◽  
Author(s):  
Min Lu ◽  
Xiaoli Wang ◽  
Yan Li ◽  
Joseph Tripodi ◽  
Goar Mosoyan ◽  
...  

Abstract Interferon (IFN-α) is effective therapy for polycythemia vera (PV) patients, but it is frequently interrupted because of adverse events. To permit the long-term use of IFN, we propose combining low doses of IFN with Nutlin-3, an antagonist of MDM2, which is also capable of promoting PV CD34+ cell apoptosis. Combination treatment with subtherapeutic doses of Peg IFN-α 2a and Nutlin-3 inhibited PV CD34+ cell proliferation by 50% while inhibiting normal CD34+ cells by 30%. Combination treatment with Nutlin-3 and Peg IFN-α 2a inhibited PV colony formation by 55%-90% while inhibiting normal colony formation by 22%-30%. The combination of these agents also decreased the proportion of JAK2V617F-positive hematopoietic progenitor cells in 6 PV patients studied. Treatment with low doses of Peg IFN-α 2a combined with Nutlin-3 increased phospho-p53 and p21 protein levels in PV CD34+ cells and increased the degree of apoptosis. These 2 reagents affect the tumor suppressor p53 through different pathways with Peg IFN-α 2a activating p38 MAP kinase and STAT1, leading to increased p53 transcription, whereas Nutlin-3 prevents the degradation of p53. These data suggest that treatment with low doses of both Nutlin-3 combined with Peg IFN-α 2a can target PV hematopoietic progenitor cells, eliminating the numbers of malignant hematopoietic progenitor cells.


2010 ◽  
Vol 38 (6) ◽  
pp. 472-480 ◽  
Author(s):  
Min Lu ◽  
Wei Zhang ◽  
Yan Li ◽  
Dmitriy Berenzon ◽  
Xiaoli Wang ◽  
...  

2015 ◽  
Vol 43 (10) ◽  
pp. 912-918.e2 ◽  
Author(s):  
Katherine Y. King ◽  
Katie A. Matatall ◽  
Ching-Chieh Shen ◽  
Margaret A. Goodell ◽  
Sabina I. Swierczek ◽  
...  

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 796-796 ◽  
Author(s):  
Windy D. Berkofsk-Fessler ◽  
Jonathan D. Licht ◽  
Melanie-Jane McConnell ◽  
Donna S. Neuberg ◽  
Timothy S. Bowler ◽  
...  

Abstract Polycythemia vera (PV) is a myeloproliferative disease characterized by accumulation of erythrocytes and cells of the myeloid and megakaryocyte lineages. Although genes like PRV-1 and PTP-MEG2 have been implicated in the pathology of PV, there is no consensus on their importance in the disease process. Progenitor cells from PV patients can grow in the absence of erythropoietin, and are hypersensitive to a variety of other growth factors. This suggests that polycythemic hematopoietic progenitor cells possess a significantly different genetic program. We tested this idea by molecular profiling hematopoietic progenitor cells (CD34+) from PV specimens and normal donors. We purified CD34+ cells from the marrow of 10 PV patients and harvested total RNA. Biotinylated cRNA was made through two rounds of linear amplification, and hybridized to Affymetrix HGU133A genechips. CD34+ cells from marrow mononuclear cells of 5 normal controls were processed similarly. The resulting datasets were normalized to the median across chips and across genes. Unsupervised hierarchical clustering showed that PV samples had a distinct gene expression profile from the controls. We then performed supervised clustering using a non-parametric t-test (Wilcoxon rank sum test) using the Benjamini and Hochberg multiple testing correction held to a p-value of 0.01 to determine genes that were significantly different between disease and normal samples. Using these stringent criteria, there were 331 genes that reached significance. Strikingly most of these were decreased in expression compared with control CD34+ cells and only 34 genes were upregulated in PV. A 35 gene predictor set was discovered through the use of a k-nearest neighbor metric. This set was 100% accurate for the prediction of PV in a leave one out cross-validation approach. Among these genes are EVI1, a known oncogene and one of only two genes upregulated in PV on this list, and the putative tumor suppressor genes TUSC4 (NPR2), NDRG1 and KLF4. Also among the predictor genes is BAALC, a gene expressed in normal CD34+ cells and known to be a prognostic indicator gene for acute myeloid leukemia.


Blood ◽  
2010 ◽  
Vol 116 (20) ◽  
pp. 4284-4287 ◽  
Author(s):  
Min Lu ◽  
Jiapeng Wang ◽  
Yan Li ◽  
Dmitriy Berenzon ◽  
Xiaoli Wang ◽  
...  

Abstract Polycythemia vera (PV) treatment with interferon α (IFNα) is frequently limited by dose-related toxicity. PV CD34+ cells are characterized by overexpression of Bcl-xL, which can be antagonized by ABT-737 leading to apoptosis. We explored the effects of ABT-737 and IFNα on PV hematopoiesis. Both IFNα and ABT-737 alone or in combination had a modest effect on normal hematopoiesis but each individually were able to markedly induce PV CD34+ cell apoptosis and suppress hematopoietic colony formation. The inhibitory activities of these agents in combination were greater against PV hematopoiesis than either agent alone. The exposure of PV CD34+ cells to low doses of IFNα and ABT-737 in combination resulted in the reduction of the proportion of JAK2V617F+ colonies similar to that observed with higher doses of IFNα. These data provide the rationale for combination therapy with low doses of IFNα and a BH3 mimetic for patients with PV.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3916-3916 ◽  
Author(s):  
Min Lu ◽  
Wei Zhang ◽  
Daniel Yoo ◽  
Dmitriy Berenzon ◽  
Yan Li ◽  
...  

Abstract Abstract 3916 Poster Board III-852 Polycythemia vera (PV) is a Philadelphia chromosome negative chronic myeloproliferative neoplasm (MPN) which is characterized by acquisition of a mutation in JAK2 (JAK2V617F). The administration of a pegylated form of interferon-alpha-2a (Peg IFNa-2a) to patients with PV has recently been reported to lead to hematological remissions and a reduction of the JAK2V617F allele burden in most patients receiving this modality of therapy. The mechanism underlying this profound clinical response of PV patients to Peg IFNa-2a has been the subject of a great deal of speculation. In order to evaluate the mechanism by which Peg IFNa-2a affects hematopoiesis in PV patients, CD34+ cells isolated from cord blood and the peripheral blood of patients with PV were cultured in semisolid media in the presence and absence of 200 and 500 U of Peg IFNa-2a. These relatively low doses of Peg IFNa-2a did not alter hematopoietic colony formation by CB CD34+ cells but inhibited PV CFU-GM colony formation by 35% and 50%, and BFU-E colony formation by 60% and 80%, respectively. Furthermore, the hematopietic colonies that formed in the presence of Peg IFNa-2a were composed of far fewer cells than those cultured in the presence of cytokines alone. In addition, individual hematopoietic colonies were plucked and the JAK2 genotype was assessed by nested allele-specific PCR assay. Exposure of PV CD34+ cells to Peg-IFNa-2a (500U) resulted in a reduction in the proportion of JAK2V617F-positive hematopoietic progenitor cells from 81.7±16.3% to 50.3±27.6% (p=0.004). Samples from 81.9% of the PV patients (9 of 11 samples) responded in this fashions to Peg IFNa 2a treatment. We then showed that incubation of PV CD34+ cells but not CB CD 34+ cells with 200 and 500U of Peg IFNa-2a resulted in increased rates of apoptosis by 4.3% and 15.3%, respectively. Erythroblasts and megakayoctes from patients with PV have been previously shown to be characterized by over-expression of the anti-apoptotic proteins Bcl-xL. We then examined if the effects of IFNa-2a could be enhanced by addition of the Bcl-xL inhibitor-ABT-737. After 2 days of treatment, Peg IFNa 2a plus ABT-737 induced significantly greater degree of apoptosis (∼50%) of a JAK2V617F positive erythroleukemia cell line (HEL cells) as compared to treatment with each agent alone, (Peg-IFNa-2a, <5%; ABT-737, 20%). PV CD34+ cells were incubated with Peg IFNa 2a (500 U) alone, ABT-737 (0.25 uM) alone or ABT-737 plus Peg IFNa 2a for 4 days and the numbers of cells were decreased by 35%, 40% and 65 %, respectively; and the corresponding percentage of apoptotic cells was 20%, 15% and 60%, respectively. Western blot analysis showed that the Bcl-xL protein level in PV but not CB mononuclear cells was reduced by treatments with ABT-737 alone or in combination with Peg IFNa 2a. Furthermore, treatment of PV CD34+ cells with ABT-737 plus Peg IFNa 2a (200U) lead to the appearance of a smaller proportion of JAK2 V617F-positive (46.7±26%) hematopoietic progenitor cells as compared to cells incubated with cytokines alone (81.7±17%) or cytokines plus Peg IFNa 2a (69±20%). These data suggest that low doses of Peg IFNa 2a selectively and directly eliminate Jak2V617F hematopietic progenitor cells which likely accounts for the therapeutic responses that have been observed with the use of this agent in the clinic. The enhanced elimination of JAK2V617F hematopoietic progenitor cells observed with the combination of ABT-737 and Peg-IFNa-2a suggests that this strategy might be an even more optimal approach for the treatment of JAK2V617F positive MPN which merits further testing in the clinic. Disclosures: No relevant conflicts of interest to declare.


1995 ◽  
Vol 71 (3) ◽  
pp. 119-121
Author(s):  
G. Castello ◽  
R. Lerza ◽  
A. Cerruti ◽  
D. Cavallini ◽  
G. Bogliolo ◽  
...  

1995 ◽  
Vol 71 (3) ◽  
pp. 119-121 ◽  
Author(s):  
G. Castello ◽  
R. Lerza ◽  
A. Cerruti ◽  
D. Cavallini ◽  
G. Bogliolo ◽  
...  

Blood ◽  
2019 ◽  
Vol 133 (16) ◽  
pp. 1729-1741 ◽  
Author(s):  
Helen M. McRae ◽  
Alexandra L. Garnham ◽  
Yifang Hu ◽  
Matthew T. Witkowski ◽  
Mark A. Corbett ◽  
...  

Abstract Somatically acquired mutations in PHF6 (plant homeodomain finger 6) frequently occur in hematopoietic malignancies and often coincide with ectopic expression of TLX3. However, there is no functional evidence to demonstrate whether these mutations contribute to tumorigenesis. Similarly, the role of PHF6 in hematopoiesis is unknown. We report here that Phf6 deletion in mice resulted in a reduced number of hematopoietic stem cells (HSCs), an increased number of hematopoietic progenitor cells, and an increased proportion of cycling stem and progenitor cells. Loss of PHF6 caused increased and sustained hematopoietic reconstitution in serial transplantation experiments. Interferon-stimulated gene expression was upregulated in the absence of PHF6 in hematopoietic stem and progenitor cells. The numbers of hematopoietic progenitor cells and cycling hematopoietic stem and progenitor cells were restored to normal by combined loss of PHF6 and the interferon α and β receptor subunit 1. Ectopic expression of TLX3 alone caused partially penetrant leukemia. TLX3 expression and loss of PHF6 combined caused fully penetrant early-onset leukemia. Our data suggest that PHF6 is a hematopoietic tumor suppressor and is important for fine-tuning hematopoietic stem and progenitor cell homeostasis.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1894-1894 ◽  
Author(s):  
Ricardo Amaru ◽  
Martin Villarroel ◽  
Hortencia Miguez ◽  
Rosario Peñaloza ◽  
Gina Torres ◽  
...  

Abstract Abstract 1894 Poster Board I-917 Background Chronic Mountain Sickness (CMS) is a clinical entity that occurs in native or long-life residents above 2500 meters of altitude. The disease is characterized by massive erythrocytosis, hyperviscosity syndrome (headache, dyspnea and cyanosis) severe hypoxemia and cardiopulmonary symptoms. The etiology is unknown and no association has been found with Erythropoietin (EPO), Epo receptor (EpoR), Hypoxia Inducible Factor 1a (HIF-1a), von Hippel Lindau (VHL), as well as PHD1, PHD2, PHD3 or PTEN genes. Therapy relies on phlebotomy and oxygen support. Acetazolamide, Medroxyprogesterone and Enalapril have also been tested, but their use has not been largely implemented. Since HMG-CoA inhibitors such as farnesyltransferase inhibitors (Larghero, Blood 2005) may inhibit the in vitro autonomous erythropoiesis of polycythemia vera patients, we studied in CMS the therapeutic potential of statins that have similar pharmacologic activity. Patients and Methods Normal controls (NC, n= 10) and patients were native Bolivians from the city of La Paz, Bolivia (3600–4000 mt altitude). The diagnosis of CMS (n=15) was made according to the consensus statement on this disease (Leon-Velarde, 2005). The diagnosis of Polycythemia Vera (PV, n= 5) or secondary erythrocytosis (SE, n= 10) was done according to WHO criteria or established clinical guidelines. Serum Erythropoietin (sEpo) was assessed by chemiluminescent assay. Burst forming units-erythroid (BFU-E) assay was performed by plating 105 BM mononuclear cells in methylcellulose with or without recombinant human rhEpo (2IU/ml) and Simvastatin (20 mM). Evaluation of apoptosis by Annexin V/7-AAD and JAK2V617F mutational analyses were performed as described (Guerini et al, Leukemia 2008). Results CMS patients (median age 48 years, range 29–58) had median values of hemoglobin and hematocrit (Hb 20.3 gr/dl, range: 19.1–22 and Hct, 62%) significantly higher than observed in NC (Hb 16.2 gr/dl, range 14.8–16.5 and Htc 52%), respectively (p< 0.001) and significantly lower than SE patients (Hb: 22.8 gr/dl, range 20.2–25 and Htc: 71%), (p<0.001). Hb and Hct were not different in CMS and PV patients (p= 0.875). In CMS sEpo values (median 22 mIU/mL, range: 16.1–45.1) were significantly higher compared to NC (median 10.7 mIU/mL, range 7–18.8) (p<0.001) and lower compared to SE patients (median 82.9 mIU/mL, range 44.8–135) (p<0.001); as expected, PV patients showed very low sEpo levels (median 3 mIU/ml, range 2.5–5.2). The JAK2V617F mutation analysis proved negative in all NC, CMS and SE patients and positive in PV. In the absence of exogenous rhEpo, a median of 0, 10, 0, 45 BFU-E colonies were obtained from NC, CMS, SE and PV patients. When rhEpo was added, 21, 40, 47 and 130 BFU-E were counted, respectively; this difference was significant when comparing NC and PV to CMS (p<0.001; p< 0.001 respectively), but not in the case of SE vs. CMS (p= 0.227). Interestingly, in PV and CMS patients, BFU-E colonies remain remarkably viable between day 14–21 while viability declined rapidly in NC and SE colonies after day 14. The prolonged viability and higher sensitivity to rhEpo of BFU-E obtained from CMS and PV erythroid progenitors was also confirmed by plating BM mononuclear cells with suboptimal doses of rhEpo (0.03 to 1 IU/ml). Moreover, when simvastatin (20 μM) was added in vitro to rhEpo driven BFU-E colonies, it induced a median inhibition of 29% in NC as compared to 37, 56 and 44 in CMS, SE and PV (p <0.013: p<0.001; p<0.001, respectively). Finally, 11 CMS patients who had a concomitant hypercholesterolemia (median cholesterol level 238 mg/dl, range 206– 310) had the opportunity to be treated with statins (atorvastatin, 20–40 mg/day). Before starting treatment with atorvastatin, all patients, who had median Hb and Htc values of 19.9 gr/dl and 63 % respectively, performed phlebotomy. After a median follow up of 18 months with atorvastatin, the median Hb and Htc values were 17.1 gr/dl and 54.6 %, respectively. The need of phlebotomy was apparently reduced, from 4–6 sessions/year to 1. Conclusions Our results underline that a) hematopoietic progenitor cells from CMS patients may promote an autonomous erythroid colony growth and show hypersensitivity to hrEpo b) statins may induce in vitro a significant inhibition of this accelerated erythropoiesis so that they could play a therapeutic role in the treatment of this and other chronic myeloproliferative disorders. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document