scholarly journals Anti-IL-6 cytokine treatment has no impact on elevated hematocrit and splenomegaly in a polycythemia vera mouse model

Author(s):  
Conny Katrin Baldauf ◽  
Peter Müller ◽  
Tobias Ronny Haage ◽  
Stephanie Adam-Frey ◽  
Juliane Lokau ◽  
...  

Somatic mutations in JAK2, MPL and Calreticulin and inflammation play a key role in pathophysiology of chronic myeloproliferative neoplasia (CMN). One of the most prominent cytokines elevated in serum of Polycythemia vera patients is interleukin-6 (IL-6). Currently, it is being discussed whether suppression of inflammation by anti-cytokine approaches as anti-IL-6 treatment may be therapeutically useful in CMN. We here sought to investigate the efficacy of anti-IL-6 treatment on inflammatory cytokines, hematocrit and splenomegaly in CMN like disease. JAK2-V617F knock-in mice (JAK2+/V617F) were treated for three weeks with anti-IL-6 antibody (Ab) or IgG-control. Upon anti-IL-6 Ab treatment, serum levels of CXCL2 and CXCL10 were significantly reduced. In addition, CXCL1, CCL11, M-CSF, G-CSF, IL-17, IL-12p40 and CCL2 were reduced by a factor of 0.3 - 0.8. Partly, this was also achieved by applying high-dose IgG. Hematocrit, erythrocyte and leukocyte counts were elevated in JAK2+/V617F mice but were not reduced by anti-IL6 Ab treatment. In addition, there was no apparent amelioration of splenomegaly and spleen histopathology. In conclusion, anti-IL-6 Ab treatment did not result in improvement of hematological disease parameters but was shown to modulate the serum cytokine signature.

2020 ◽  
Vol 10 (4) ◽  
pp. 259
Author(s):  
Diana L. Lighezan ◽  
Anca S. Bojan ◽  
Mihaela Iancu ◽  
Raluca M. Pop ◽  
Ștefana Gligor-Popa ◽  
...  

Background: The complexity of myeloproliferative neoplasms (MPNs) cannot be characterized by acquired somatic mutations alone. Individual genetic background is thought to contribute to the development of MPNs. The aim of our study was to assess the association between the TET2 rs1548483 single nucleotide polymorphism (SNP) and the susceptibility to polycythemia vera (PV), essential thrombocythemia (ET), primary myelofibrosis (PMF) or chronic myeloid leukemia (CML). Methods: We evaluated the TET2 rs1548483 SNP through real-time PCR in 1601 MPN patients out of which 431 with PV, 688 with TE, 233 with PMF, 249 with CML and 197 controls. We included only patients with a molecularly proven driver mutation, such as JAK2 V617F, CALR or BCR-ABL1. Results: Significant association between TET2 rs154843 variant allele and JAK2 V617F-positive PV and PMF (OR = 1.70; 95% CI: 1.01–2.91; p-value = 0.046, and OR = 2.04; 95% CI: 1.10–3.77; p-value = 0.024, respectively), and type 2 CALR-positive PMF (OR = 2.98; 95% CI: 1.12–7.93; p-value = 0.035) was noted. Conclusions: The TET2 rs1548483 SNP is associated with the susceptibility to molecularly annotated PV and PMF.


Blood ◽  
2011 ◽  
Vol 117 (10) ◽  
pp. 2813-2816 ◽  
Author(s):  
Francesco Passamonti ◽  
Chiara Elena ◽  
Susanne Schnittger ◽  
Radek C. Skoda ◽  
Anthony R. Green ◽  
...  

Abstract Although approximately 95% of patients with polycythemia vera (PV) harbor the V617F mutation in JAK2 exon 14, several mutations in exon 12 have been described in the remaining patients. We conducted a European collaborative study to define the molecular and clinical features of patients harboring these mutations. Overall, 106 PVs were recruited and 17 different mutations identified. Irrespective of the mutation, two-thirds of patients had isolated erythrocytosis, whereas the remaining subjects had erythrocytosis plus leukocytosis and/or thrombocytosis. Compared with JAK2 (V617F)-positive PV patients, those with exon 12 mutations had significantly higher hemoglobin level and lower platelet and leukocyte counts at diagnosis but similar incidences of thrombosis, myelofibrosis, leukemia, and death. In a multivariable analysis, age more than 60 years and prior thrombosis predicted thrombosis. These findings suggest that, despite the phenotypical difference, the outcome of JAK2 exon 12 mutations-positive PV is similar to that of JAK2 (V617F)-positive PV.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 1979-1979
Author(s):  
Kai Wang ◽  
Sabina Swierczek ◽  
Kimberly Hickman ◽  
Hakon Hakonarson ◽  
Josef T. Prchal

Abstract Abstract 1979 Polycythemia vera (PV) is an acquired clonal blood disorder characterized by variable increased number of clonal myeloid cells (erythrocytes, granulocytes and platelets) and mostly normal polyclonal T lymphocytes. Most patients carry a gain-of function JAK2 V617F mutation in 2 copies acquired by a uniparenteral disomy; i.e. a copy-neutral loss of heterozygosity (LOH) in chromosome 9p in PV pathogenesis. Yet, JAK2 V617F mutation is not a PV-initiating event and the family clustering of PV suggests a contribution of inherited genetic events to its pathogenesis. Interestingly, majority of PV subjects acquire the JAK2 V617F mutation on a distinct a germline SNP 9p24.1 haplotype. Using whole-genome SNP arrays, we assayed 34 T-cells and 66 granulocytes from PV patients (including 32 pairs from the same patient). Comparison to a control population identified multiple germline SNPs around JAK2 as being associated with PV susceptibility (rs11999802, P=1.8×10-8, OR=4.4). Analysis of somatic genomic changes in granulocytes revealed strong genetic heterogeneity: we detected 9p LOH in 36% of the patients, 9p chromosomal gain in 1 patient, as well as “fractional LOH” in 14% of the patients where a small fraction of granulocytes have LOH. Interestingly, the magnitude of somatic LOH in 9p was strongly associated with somatic V617F allelic dosage (r=0.74, P=4.8×10-12), suggesting that LOH preferentially increase V617 PV subclone without altering genomic copy number. Adjusting for the fractional LOH, we did not have sufficient power to detect association between rs11999802 germline genotypes and increased dosage of somatic V617F mutations. Analysis of granulocytes with heterozygous rs11999802 genotypes demonstrated that LOH increases the relative fraction of germline risk alleles versus non-risk alleles. In summary, germline risk variants surrounding JAK2 predispose to somatic SNVs within JAK2, whose allelic dosage can be further increased by a serial subclonal expansion of allele-specific copy-neutral LOH or chromosomal duplications. Our study illustrates how convergent mechanisms of somatic mutations increase the genetic load of gain-of-function mutations contributing to the clonal expansion and disease pathogenesis. Figure 1 Box plot of the percentage of somatic V617F mutation in JAK2 over each of the fractional LOH measures in granulocytes. Strong correlation between these two measures was detected (r=0.74, P=4.8×10-12). Figure 1. Box plot of the percentage of somatic V617F mutation in JAK2 over each of the fractional LOH measures in granulocytes. Strong correlation between these two measures was detected (r=0.74, P=4.8×10-12). Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 4 (12) ◽  
pp. 2567-2576 ◽  
Author(s):  
Theodoros Karantanos ◽  
Shruti Chaturvedi ◽  
Evan M. Braunstein ◽  
Jerry Spivak ◽  
Linda Resar ◽  
...  

Abstract The factors underlying the variable presentation and clinical course of myeloproliferative neoplasms (MPNs) remain unclear. The aim of this study was to evaluate the independent effect of sex on MPN presentation and outcomes. A total of 815 patients with essential thrombocytosis, polycythemia vera, or primary myelofibrosis were evaluated between 2005 and 2019, and the association of sex with presenting phenotype, JAK2 V617F burden, progression, and survival was examined. Men presented more often with primary myelofibrosis vs essential thrombocytosis (relative risk, 3.2; P < .001) and polycythemia vera (relative risk, 2.1; P < .001), had higher rates of transformation to secondary myelofibrosis (hazard ratio [HR], 1.55; P = .013) and acute myeloid leukemia (HR, 3.67; P < .001), and worse survival (HR, 1.63; P = .001) independent of age, phenotype at diagnosis, and MPN-specific mutation. Men had higher JAK2 V617F allele burdens in their CD34+ cells (P = .001), acquired more somatic mutations (P = .012) apart from the MPN-specific mutations, and had an increased frequency of 1 (odds ratio, 2.35; P = .017) and 2 (odds ratio, 20.20; P = .011) high-risk mutations independent of age, phenotype, and driver mutation. Male sex is an independent predictor of poor outcomes in MPNs. This seems to be due to an increased risk of non–MPN-specific somatic mutations, particularly high-risk mutations, rather than MPN-specific mutation allele frequency. Conversely, disease progression in female subjects is more dependent on JAK2 mutation allele burden than on acquisition of other somatic mutations. Sex should be considered in prognostic models and when evaluating therapeutic strategies in MPNs.


2005 ◽  
Vol 2005 (5) ◽  
pp. 298-303 ◽  
Author(s):  
Joanna Narbutt ◽  
Aleksandra Lesiak ◽  
Malgorzata Skibinska ◽  
Anna Wozniacka ◽  
Anna Sysa-Jedrzejowska ◽  
...  

The aim of our study was to compare serum concentration of IL-1β, IL-6, IL-8, IL-10, and TNF-αin 105 healthy volunteers before and after exposure to UVR: 25 subjects (10 days of UVB), 55 (10 days of UVB or solar-simulated radiation, followed by acute UVB dose), and 25 (local high dose of UVB). In all the individuals blood samples were analyzed before and after final irradiation by chemiluminescence assay. After 10 days of UVB irradiation a statistically significant increase in serum concentration only in IL-8(P<.05)and strong tendency in TNF-α(P=.05)were observed. The applied schedules of irradiation have minor impact on serum cytokine level and still a threshold dose of UVR causing systemic immune impairment is unknown.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4322-4322
Author(s):  
Jared S Fowles ◽  
Chi-Joan How ◽  
Stephen T Oh

Abstract Introduction: Polycythemia vera (PV) is a myeloproliferative neoplasm (MPN) characterized by overproduction of mature erythrocytes and a propensity for transformation to myelofibrosis (MF) and acute myeloid leukemia (AML). The defining genetic feature of PV is the JAK2 V617F mutation, present in 95-97% of patients, resulting in overactive JAK-STAT signaling. Secondary mutations have been identified which may modulate PV disease phenotype. Although typically diagnosed in older individuals, PV occurs in young patients (≤ 45 years old) as well. Compared with old patients (≥ 65 years old), a significant association with young PV and female gender, lower white blood cell count, lower JAK2 V617F allele burden, palpable splenomegaly, and splanchnic vein thrombosis has been reported. In the same study, the frequency of transformation to MF or AML was found to be similar, although the median time to transformation was shorter in old PV. The reasons for these age-related associations are not clearly understood. Since somatic mutations have been shown to accumulate with age, we hypothesized that the mutational profile of young PV patients may differ from that of old PV, which could contribute to the differences in clinical phenotypes observed. Methods: DNA from tumor (bulk peripheral blood mononuclear cells or granulocytes) and matched normal tissue (skin or sorted CD3+ T cells) was isolated from samples from 10 young (≤ 45 years old) and 11 old (≥ 65 years old) PV patients. Enhanced exome sequencing utilizing the standard IDT exome capture panel plus additional probes for genes known to be recurrently mutated in MPN and/or AML was performed. To determine order of mutation acquisition (and to rule out the possibility of biclonal disease) qPCR-based genotyping was performed on single cell-derived colonies from patients with ≥ 1 secondary mutation (in addition to JAK2 V617F). Plasma concentrations of 10 cytokines were assayed in 15 young and 23 old PV patient samples compared with age-matched healthy controls. Results: Comparative analysis of 7 young and 10 old patients (four samples were removed due to sample contamination/artifact issues) identified a total of 133 somatic mutations. A significantly higher overall mutational load was observed in the old group (mean 10.9 vs 3.4; p = 0.0025). A trend toward higher JAK2 V617F variant allele frequency (VAF) was observed in old versus young PV in the sequenced patients, and in a separate clinical cohort the JAK2 V617F VAF was significantly higher in old PV patients (p = 0.0067). Notably, putative secondary driver mutations were identified in 9/10 old PV patients (including ASXL1, TET2, and DNMT3A), whereas JAK2 appeared to be the only driver mutation present in each of the young PV patients. Secondary mutations were validated as being acquired before, after, or coincident with JAK2 V617F, with no specific predominant pattern observed. MPN-predisposing germline JAK2 46/1 haplotype occurrence did not differ with age, and additional germline mutations in MPN genes were not enriched in either cohort. Supranormal levels of multiple cytokines (e.g. IFNγ, IL-2Rα, IP-10, MIP-1α) were observed in both young and old PV, with a slight enhancement observed with increased age. Conclusions: These data indicate a striking age-based distinction in the near uniform absence (young) or presence (old) of secondary non-JAK2 mutations in PV patients. The finding that putative secondary driver mutations were acquired before JAK2 V617F in only a minority of old PV suggests that the overall increase of secondary mutations in this cohort may not merely be a function of aging, implying that these mutations do in fact contribute to PV development and/or progression. As transformation can be induced by accrual of disease-modifying mutations, the presence of secondary mutations in old PV patients is consistent with the reported shorter time to transformation in this age group, since young PV patients would likely need additional time to acquire secondary mutations. Abnormal inflammatory cytokine production was common to both young and old PV, suggesting this is not the predominant factor distinguishing these two cohorts. The contribution of additional non-genomic factors (e.g. epigenetic, hormonal) will be the subject of future study. Disclosures Oh: Janssen: Research Funding; Incyte: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; CTI Biopharma: Research Funding; Gilead: Research Funding; Takeda: Research Funding.


Cell Research ◽  
2008 ◽  
Vol 18 (S1) ◽  
pp. S141-S141
Author(s):  
Alvin CH Ma ◽  
Alice MS Cheung ◽  
Alister C Ward ◽  
Wing-Yan Au ◽  
Yok-Lam Kwong ◽  
...  

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3827-3827
Author(s):  
Francesca Ferraro ◽  
Christopher A Miller ◽  
Amy Abdalla ◽  
Nichole Helton ◽  
Nathan Salomonis ◽  
...  

Currently, it is not clear why some patients with acute myeloid leukemia (AML) can be "cured" with chemotherapy alone; are they living with small amounts of disease that is held in check by immunologic (or other) mechanisms, or is their disease really eradicated? The percentage of cytogenetically normal AML patients who have long (>5 years) first remissions (LFRs) after chemotherapy alone is low (about 9.1% in patients <60 years and 1.6% in >60 years1). For this reason, most intermediate risk patients are offered allogeneic transplantation to decrease their risk for relapse. To better understand mechanisms of chemotherapy sensitivity in AML, we performed an analysis of the mutation landscape and persistence, using samples from 8 normal karyotype LFR patients (without CEBPA mutations) who received standard "7+3" induction and high dose cytarabine consolidation as their only therapy. The mean age at diagnosis was 43.5 years, and the mean follow up in first remission is 7.6 years; none of these patients has relapsed to date. For each case, we performed enhanced exome sequencing at diagnosis (235x coverage of the entire exome, and ~1008x coverage of recurrently mutated AML genes). Each case had at least one documented AML driver mutation, with a median of 29 somatic mutations in the exome space. We created probes for 225 mutations (mean 28 per case), and performed error-corrected sequencing (Haloplex) for all available remission samples. The mean depth of Haloplex coverage was 1607x, and each sample had at least one AML-specific mutation assayed, with a sensitivity of 1 cell in 1,750 (0.06%). 7/8 patients demonstrated complete clearance of all mutations in all remission samples tested, which was confirmed with digital droplet PCR for 5 cases, with a sensitivity of detection of 1 cell in 100,000. In one case, we detected a persistent ancestral clone harboring DNMT3AR882H, which can be associated with long first remissions for some patients2. Strikingly, the founding clone in all 8 cases had one or more somatic mutations in genes known to drive cell proliferation (e.g. MYC, FLT3, NRAS, PTPN11, Figure 1 top panel). These are usually subclonal mutations that occur late during leukemic progression, suggesting that the presence of a "proliferative hit" in the founding clone might be important for chemotherapy clearance of all the AML cells in a given patient. To support this hypothesis, we analyzed the mutational clearance of 82 AML cases with paired diagnosis and day 30 post-chemotherapy bone marrow samples. We observed that, whether present in the founding clone or in subclones, mutations in MYC, CEBPA, FLT3, NRAS, and PTPN11 cleared after induction chemotherapy in all samples, while other mutations were often persistent at day 30 (e.g. DNMT3A, IDH1, IDH2, NPM1, TET2; Figure 1 bottom panel). Compared to other published sequencing studies of AML, MYC and NRAS mutations were significantly enriched in this small cohort (MYC p= 0.002, and NRAS p= 0.034), with MYC enrichment being particularly striking (37.5% versus 1.8%). All MYC mutations were canonical single base substitutions occurring in the highly conserved MYC Box 2 domain at the N-terminus of MYC (p.P74Q or p.T73N). Overexpression of MYCP74Q in murine hematopoietic progenitors prolonged MYC half life (89 min vs. 44 min for wild type), and enhanced cytarabine sensitivity at all concentrations tested (range 10-1000 nM, p=0.0003), both in vitro and in a MYC-driven leukemia model in vivo. MYC expression measured with flow cytometry in the blasts of the LFR samples was significantly higher (p=0.045) compared to unfavorable risk (complex karyotype) or other intermediate risk categories, but similar to good risk AML (biallelic CEBPA mutations, core binding factor fusion-associated AML, and AML with isolated NPMc), suggesting that activation of the MYC pathway may represent a shared feature of chemosensitive patients. Taken together, these data suggest that some intermediate patients who are effectively "cured" with chemotherapy alone may not have persistent subclinical disease, nor retained ancestral clones that could potentially contribute to relapse. Importantly, these patients often have mutations driving cell proliferation in the founding clone, indicating that the presence of specific mutations in all malignant cells may be critical for complete AML cell clearance with chemotherapy. 1. Blood Adv. 2018 Jul 10; 2(13): 1645-1650 2. N Engl J Med 2018; 378:1189-1199 Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4588-4588 ◽  
Author(s):  
Yongbao Wang ◽  
Albert K Ho ◽  
Qiulu Pan ◽  
Frederick Karl Racke ◽  
Dan Jones

Abstract Introduction: Mutations in the chaperone gene calreticulin (CALR) have been recently identified in essential thrombocythemia (ET) and primary myelofibrosis (PMF), and are essentially mutually exclusive with JAK2 or MPL mutations. Normal and mutant CALR proteins may differentially affect the subcellular trafficking of JAK-STAT signaling components. CALR mutations previously reported in ET and PMF have been +1 frameshift (fs) mutations localized to exon (E) 9 that generate a novel C-terminal protein sequence with a shift from acidic to basic residues. CALR E9 in-frame (IF) deletions have been recently rarely reported as polymorphisms such as TMP_ESP_19_13054686_13054688 and TMP_ESP_19_13054650_13054658 (Ensembl database entries). We sought to determine the frequency and associated clinical features of CALR with E9 IF alterations in samples submitted for suspicion of a myeloproliferative neoplasm (sMPN). We also assessed whether CALR IF alterations are differentially associated with +1fs mutations or with JAK2 V617For other somatic mutations in MPN-associated genes. Materials and Methods: CALR mutation analysis of E9 was performed on genomic DNA extracted from blood, bone marrow (BM) aspirate or fixed BM biopsy sections using a Sanger sequencing assay with an analytic sensitivity of at least 15%. E9 IF cases were further assessed and mutations quantified by an Ion torrent sequencing panel assessing CALR, CSF3R, JAK2 and MPL, a second panel containing ASXL1, EZH2, IDH1, IDH2, KRAS, NRAS and TET2 and an Illumina MiSeq extended panel with 20 additional MPN-associated genes. These assays had a sensitivity of approximately 5%. JAK2 V617Fmutations were quantitated using a pyrosequencing assay with an analytic sensitivity of 1%. Results: We assessed CALR E9 mutation status in 733 sMPN samples that were negative for JAK2 V617F mutation. 148 (20.1%) had typical +1fs mutations (95 type 1 and variants, 53 type 2 and variants); 2 (0.3%) had point mutations (E381A and D7373M); 7 (1.0%) had IF deletions including E381_A382>A, D397_D400>D (n =4), D400_K401>D and E405_V409>V. All E9 IF deletions were present at ~50% of reads. Clinical diagnoses were cytopenia/BM fibrosis, ET, thrombocytosis/anemia, and sMPN unspecified. Mutation analysis for 27 additional MPN-associated genes revealed mutations in 5/7 (71.4%) IF deletion cases including in MPL (W515L,40%; D163Y,12%), CSF3R (A470T 46%), ASXL1 (D954fs*26, 45%) and ZRSR2 (S449_R450dup, 27%). No additional mutations were found in the 2 cases with non-synonymous CALR point mutations/SNPs. In a parallel set of 76 MPN samples that had JAK2 V617F at varying levels, we noted 1 E9 IF deletion (D397_D400>D) in a sMPN case with 21.6% JAK2 V617F, and a typical +1fs mutation (K385fs*47) in a case with low (4.2%) JAK2 V617F. All other JAK2 V617F cases had no E9 CALR alterations. Conclusions: CALR E9 in-frame deletions occur in up to 1% of sMPN samples and involve a variety of codons in the acidic domain. Therefore, sizing assays without DNA sequencing are not sufficient to unequivocally distinguish IF deletions from the characteristic +1 frameshift somatic mutations associated with ET and PMF. Given their level, these CALR IF deletions are likely germline sequence variants but are associated with a high frequency of somatic mutations in other MPN-associated genes but not with CALR +1fs mutations. Their co-occurrence with pathogenic somatic mutations in JAK2, MPL and CSF3R affecting the JAK-STAT pathway raises the possibility for a contributory role of altered CALR proteins produced by these E9 deletions in the pathogenesis of MPN. Disclosures Wang: Quest Diagnostics: Employment. Ho:Quest Diagnostics: Employment. Pan:Quest Diagnostics: Employment. Racke:Quest Diagnostics: Employment. Jones:Quest Diagnostics: Employment.


Sign in / Sign up

Export Citation Format

Share Document