scholarly journals Hereditary Chronic Neutrophilic Leukemia in a Four Generation Family without Transformation to Acute Leukemia

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3594-3594
Author(s):  
Zane Chiad ◽  
Amanda Lance ◽  
Sara L. Seegers ◽  
Sarah-Catherine Paschall ◽  
Kendra Drummond ◽  
...  

Abstract Chronic neutrophilic leukemia (CNL) is a rare myeloproliferative neoplasm (MPN) characterized by peripheral blood leukocytosis consisting primarily of segmented neutrophils and band forms, hypercellular bone marrow with granulocytosis, hepatosplenomegaly, and the presence of activating colony-stimulating factor 3 receptor (CSF3R) mutations. Blast transformation occurs frequently in patients with acquired CNL, with a median overall survival of 21 months from diagnosis. Typically, CSF3R mutations in CNL are thought to be somatic; however, we and others have reported rare cases of germline activating CSF3R mutations producing a familial CNL. Here we report the clinical course of a patient with CNL along with definitive evidence of inherited germline transmission of the CSF3R T618I mutation. Spanning four generations, with affected family members of ages 1.6 - 51 years, this is the largest reported pedigree of a family with familial CNL (Figure 1A). The proband is a 49-year-old female referred to our center with a history of lifelong leukocytosis and leukocyte count of 115.1 x 10 9/L with 75% granulocytes and 11% bands, platelet count of 341 x 10 9/L, and hemoglobin of 12.5 g/dL with hematocrit of 38%. The family history was also remarkable for leukocytosis. Prior therapies for the proband included imatinib, splenectomy, and hydroxyurea. Additional testing by our center revealed a T618I CSFR3 mutation, and the absence of mutations in ASXL1 and SETBP1 or a BCR-ABL translocation. Treatment with ruxolitinib resulted in improvement of her leukocyte count to 43.0 x 10 9/L with 73% granulocytes, and reduction in her alkaline phosphatase from 732 IU/L to 296 IU/L. There has been no evidence of gain of any known deleterious somatic mutations that frequently co-occur with somatic T618I CSF3R mutations in CNL in the patient to date. Germline analysis of genomic DNA extracted from cultured mesenchymal stromal cells from the proband and Sanger sequencing demonstrated a heterozygous T618I mutation. Mutational analysis of the proband's family members confirmed a heterozygous CSF3R T618I mutation in all living affected family members, while all unaffected family members tested were homozygous wild type. There has been no evidence of leukemic transformation in any affected family members to date. Mutational analysis was not feasible on the proband's deceased mother and brother with a putative CNL diagnosis due to lack of DNA samples; however, there was no evidence of transformation to acute leukemia in either of the two deceased family members. Because CSF3R can produce anti-apoptotic signaling, we hypothesized that autoactivating T618I mutations could prolong neutrophil survival. Polymorphonuclear cells (PMNs) isolated from the proband and from normal donors were cultured in vitro and apoptosis assessed at 24-hour intervals. Neutrophils expressing the CSF3R T618I had prolonged survival with a >40% decrease in apoptosis after 48 hours in culture (Figure 1B). RNA-seq followed by pathway analysis demonstrated significant decreases in activation of canonical apoptotic pathways in PMNs, including both the extrinsic and mitochondrial dependent pathways. Immunoblotting for candidate anti- and pro-apoptotic proteins revealed increased expression of the anti-apoptotic BCL2 family member MCL1 in T618I-expressing PMNs. Notably, inhibition of MCL1 using S63845 reversed the anti-apoptotic effect induced by ligand-activation of the CSF3R receptor in PMNs (P < 0.001, Figure 1C). In conclusion, we demonstrate hereditary CNL within a large family tree with no observed transformation to acute leukemia in any affected individuals up to age 51, suggesting a potentially more indolent course. Nonetheless, our observations highlight the need for germline testing of patients with CNL to better understand the natural history of CNL. Moreover, our data provide further insight into the pathobiology of CNL and potential novel targets for therapy. Figure 1 Figure 1. Disclosures Voorhees: Bristol-Myers Squibb Company.: Other: Data Safety & Monitoring; AbbVie Inc, Bristol-Myers Squibb Company; Consulting Agreement: GlaxoSmithKline, Novartis, Oncopeptides: Other: Advisory Committee.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3141-3141
Author(s):  
Seamus B Hughes ◽  
David Spiciarich ◽  
Richard D. Press ◽  
Sarah L Thompson ◽  
Jerald P. Radich ◽  
...  

Abstract Mutations in CSF3R (aka GCSFR) occur in the majority of patients with Chronic Neutrophilic Leukemia (CNL) and also are rarely found in adult and pediatric Acute Myeloid Leukemia. The most common CSF3R mutation in CNL is T618I (aka T595I), a point mutation in the membrane-proximal extracellular domain that causes ligand independence. Mutations that lead to a premature stop in the cytoplasmic domain are also found in CNL and result in increased expression of CSF3R on the cell surface. Understanding the biology of other novel mutations in CSF3R may lead to insight into both receptor biology and oncogenesis. The CSF3R N610H mutation was identified in a patient with a myeloproliferative neoplasm, which was most consistent with a JAK2, CALR, MPL mutation negative primary myelofibrosis. This patient had a history of mild leukocytosis for several years with most recent white blood cell counts between 13.3 and 15.3 x 103/uL. A bone marrow biopsy revealed 90% cellularity with mildly increased reticulin fibrosis, increased myeloid to erythroid ratio, no overt dysplasia, and less than 5% blasts. The cells were karotypically normal with a micro deletion of the 3' end of PDGFRB (5q) identified by FISH at 59%. The patient has had minimal symptoms with no anemia or thrombocytopenia and is currently being monitored but not receiving any intervention. The patient's bone marrow was next-generation sequenced using a 42-gene myeloid malignancy targeted mutation hotspot panel which revealed a mutation at N610H in CSF3R at a 50% mutant allele frequency. Given its proximity to the most common CSF3R mutation found in Chronic Neutrophilic Leukemia (T618I, aka T595I), we were interested in understanding whether the N610H mutation might contribute to disease biology. N610 (also known as N586 in the traditional numbering system that does not include the signal peptide) is part of an N-X-T motif, which is a consensus sequence for N-linked glycosylation. Haniu et al (Biochemistry 1996) demonstrated that N610 is one of 8 sites that are N-glycosylated on CSF3R. We confirm by mass spectrometry (MS) based analysis that N610 is occupied with a bisecting complex N-glycan (in vitro). We further found that the N610H mutation and a more conservative N610Q substitution are highly activating in CSF3R, leading to cytokine-independent growth in the murine Ba/F3 cell line. Furthermore, like the T618I mutation, these mutations render the receptor ligand-independent. N610H and N610Q lead to a robust increase in downstream signaling through the JAK/STAT pathway as demonstrated by an increase in the levels of phospho-STAT3. The loss of N-glycosylation in the membrane-proximal region of CSF3R may therefore increase ligand-independent receptor activation and promote oncogenesis. This study highlights the insight that rare human mutations can provide into the relationship between receptor structure and function. Disclosures Radich: Ariad: Consultancy; Novartis: Consultancy, Research Funding; Gilliad: Consultancy; Incyte: Consultancy. Bertozzi:GlaxoSmithKline: Membership on an entity's Board of Directors or advisory committees; Catalent Biologics: Membership on an entity's Board of Directors or advisory committees; Verily: Membership on an entity's Board of Directors or advisory committees; Enable Bioscience: Equity Ownership, Membership on an entity's Board of Directors or advisory committees. Oh:CTI: Research Funding; Janssen: Research Funding; Gilead: Membership on an entity's Board of Directors or advisory committees, Research Funding; Incyte Corporation: Membership on an entity's Board of Directors or advisory committees, Research Funding.


2001 ◽  
Vol 86 (8) ◽  
pp. 3889-3899 ◽  
Author(s):  
Ingrid A. Holm ◽  
Anne E. Nelson ◽  
Bruce G. Robinson ◽  
Rebecca S. Mason ◽  
Deborah J. Marsh ◽  
...  

PHEX is the gene defective in X-linked hypophosphatemic rickets. In this study, analysis of PHEX revealed mutations in 22 hypophosphatemic rickets patients, including 16 of 28 patients in whom all 22 PHEX exons were studied. In 13 patients, in whom no PHEX mutation had been previously detected in 17 exons, the remaining 5 PHEX exons were analyzed and mutations found in 6 patients. Twenty different mutations were identified, including 16 mutations predicted to truncate PHEX and 4 missense mutations. Phenotype analysis was performed on 31 hypophosphatemic rickets patients with PHEX mutations, including the 22 patients identified in this study, 9 patients previously identified, and affected family members. No correlation was found between the severity of disease and the type or location of the mutation. However, among patients with a family history of hypophosphatemic rickets, there was a trend toward more severe skeletal disease in patients with truncating mutations. Family members in more recent generations had a milder phenotype. Postpubertal males had a more severe dental phenotype. In conclusion, although identifying mutations in PHEX may have limited prognostic value, genetic testing may be useful for the early identification and treatment of affected individuals. Furthermore, this study suggests that other genes and environmental factors affect the severity of hypophosphatemic rickets.


2021 ◽  
Vol 28 (3) ◽  
pp. 1790-1794
Author(s):  
Hans G. Drexler ◽  
Stefan Nagel ◽  
Hilmar Quentmeier

Chronic neutrophilic leukemia (CNL) is a rare myeloproliferative neoplasm that is genetically characterized by the absence of both the Philadelphia chromosome and BCR-ABL1 fusion gene and the high prevalence of mutations in the colony-stimulating factor 3 receptor (CSF3R). Additional disease-modifying mutations have been recognized in CNL samples, portraying a distinct mutational landscape. Despite the growing knowledge base on genomic aberrations, further progress could be gained from the availability of representative models of CNL. To address this gap, we screened a large panel of available leukemia cell lines, followed by a detailed mutational investigation with focus on the CNL-associated candidate driver genes. The sister cell lines CNLBC-1 and MOLM-20 were derived from a patient with CNL and carry CNL-typical molecular hallmarks, namely mutations in several genes, such as CSF3R, ASXL1, EZH2, NRAS, and SETBP1. The use of these validated and comprehensively characterized models will benefit the understanding of the pathobiology of CNL and help inform therapeutic strategies.


Blood ◽  
1956 ◽  
Vol 11 (1) ◽  
pp. 19-30 ◽  
Author(s):  
AUSTIN S. WEISBERGER ◽  
LEIF G. SUHRLAND

Abstract Selenium cystine was administered orally to 2 patients with acute leukemia and to 2 patients with chronic myeloid leukemia. In all patients there was a rapid decrease in the total leukocyte count as well as a decrease in spleen size. This effect was observed in patients refractory to other chemotherapeutic agents as well as in the usually resistant types of leukemia. In patients with chronic myeloid leukemia the immature granulocytes disappeared much more rapidly than the mature granulocytes. The most striking and consistent effects were observed in acute leukemia. One patient who had become resistant to 6-mercaptopurine appeared to reacquire sensitivity to this compound after receiving selenium cystine. These effects of selenium cystine on leukocytes correlate with the ability of selenium cystine to decrease the influx of S35 L-cystine by leukemic leukocytes in vitro. Other potentially effective analogues of cystine (or cysteine) may therefore be selected by this technic. No changes were detected in any of the organs attributable to selenium cystine toxicity. The nausea and vomiting associated with the oral administration of selenium cystine was so severe that it was not possible to administer selenium cystine for a sufficient period of time to determine whether an appreciable remission can be obtained in leukemia. Further study is necessary to determine whether selenium cystine has any practical applicability in the chemotherapy of leukemia. Although the mechanism of action of selenium cystine is not known, these striking effects of an analogue of cystine on leukemia are further suggestive of the importance of cystine (cysteine) in the metabolism of leukocytes.


1987 ◽  
Author(s):  
T R Andersson ◽  
M L Larsen ◽  
U Abildgaard

In our normal material of 379 blood donors,3 indivi-uals had values below mean −2.5 SD (below 56%).Further studies revealed hereditary deficiency in two of these individuals. In the family study,5 out of 7 individuals had heparin cofactor II (HC II) values below 56%. In only one of these 5 a history of DVT was obtained. In deceased members of her family, however, frequent episodes of thromboembolic disease had occurred.Crossed immunoelectrophoresis (CIE) was performed in plasma from deficiency individuals from both families. Heparin in the first dimension gave a pattern similar to that observed with normal pooled plasma. The inactivation of thrombin by HC II is preferentially accelerated by dermatan sulfate (DS).HC II consumption in in vitro coagulation is increased by DS rather than by heparin. This prompted the addition ofTBS to the first dimension, which makes the antigen move faster, but produced no alteration in the antigenic pattern or size, neither in pooled plasma nor in a family member with normal activity. In family members with low HC II activity, DS in the first dimension, resulted in an abnormal CIE pattern with two distinct precipitation arcs with identity pattern. Molecular heterogeneity of HC II has previously not been reported.Screening 70 individuals who had sustained thrombosis before the age of 50,no values suggesting congenital deficiency were encountered.Conclusion. The CIE findings suggest that plasma of affected family members contain two types of HC II molecules of immunological identity. One type shows the normal accelerated mobility in the presence of DS and the other lacks this effect. The abnormal electrophoretic pattern becomes, however, first apparent when DS is added to the first dimension. When looking for molecular defects in congenital HC II deficiency, it is important that the first dimension is run with DS.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4682-4682 ◽  
Author(s):  
Xin Xu ◽  
Wilhelm G Dirks ◽  
Hans G. Drexler ◽  
Zhenbo Hu

Abstract Background: Acute leukemia (AL) originates from both genetic and epigenetic changes that can be targeted to cure AL. Dysregulated DNA methylation has been shown to be associated with AL and demethylating agents 5-azacytidine and decitabine show favored improvement in secondary leukemia. Deficient histone acetylation has also been reported in AL and can be corrected to relieve leukemia. Histone methylation harbors more structural complexities compared to DNA methylation and histone acetylation and is broadly involved in AL. In particular, histone H3 lysine 9 (H3K9) methylation has been associated with AL. Di-methylation of H3K9 is reportedly involved in human hematopoietic stem cell lineage commitment. Moreover, tri-methylation of H3K9 predicts AML survival. H3K9 demethylation is catalyzed by exclusive KDM3 family members (KDM3A, KDM3B, and JMJD1C) that catalyze mono- and di-demethylation of H3K9, non-exclusive KDM4 family members (KDM4A, KDM4B, KDM4C, and KDM4D) that catalyze both H3K9 and H3K36 di- and tri-demethylation, KDM1A (LSD1) that catalyzes H3K4 and H3K9 mono- and di-demethylation, and PHF8 that catalyzes H3K9 mono- and di-demethylation and H4K20 demethylation. Among these, KDM3B, JMJD1C, KDM4C, LSD1, and PHF8 have been reported to be associated with AL in an enzymatic activity-dependent way. Furthermore, small molecular inhibitors of KDM4C and LSD1 have been developed for treatment of AML. H3K9 demethylase KDM3B is located at chromosome 5 band 31, a region frequently deleted or lost in acute myeloid leukemias (AML) and myelodysplasias (MDS). Different from other H3K9 demethylases that are usually responsible for leukemia maintenance, KDM3B harbors potential tumor-suppressive activities in acute myeloid leukemia and myelodysplastic syndromes. However, small molecular antagonists and agonists are lacking for KDM3B. Results: We aim to identify small molecular modulators of KDM3B. We focused on crystal structure of KDM3B Jumonji domain that catalyzes histone demethylation for virtual screening. From approximately 200,000 natural products and Chinese medicine components, we identified a potential KDM3B modulator, namely compound #7. Surface plasmon resonance technology showed that compound #7 binds to KDM3B with favorable affinity. In vitro and in vivo demethylation assay showed that compound #7 is able to increase H3K9 demethylating activity of KDM3B. We thus named compound #7 as KA-7 (KDM3B agonist #7). Interestingly, the identified KDM3B agonist KA-7 is able to selectively repress MLL-rearranged AL in cell proliferation and colony formation assays. Considering that KA-7 targets KDM3B that is located at chromosome 5q, a frequently deleted region in AML and MDS, we explored if KA-7 collaborates with Lenalidomide, an FDA approved drug for treating MDS with deletion at 5q where KDM3B is located. KA-7 was found to be able to synergistically increase the selective killing of AL cells by Lenalidomide. Conclusion: To sum up, physiologic H3K9 demethylase activity of KDM3B can be enhanced by a small molecular modulator KA-7 and causes selective killing against MLL-arranged AL cells. Disclosures. No relevant conflicts of interest to declare. Disclosures No relevant conflicts of interest to declare.


1968 ◽  
Vol 20 (03/04) ◽  
pp. 534-541 ◽  
Author(s):  
O Egeberg

SummarySevere hemorrhagic disorder due to congenital factor XIII deficiency is described in two unrelated Norwegian girls.Plasma cephalin time was for both patients extraordinarily short during episodes of bleeding and hematomas. No such hyperactivity reaction was demonstrable in unaffected condition some months later.Estimations of blood factor XIII levels revealed a partial defect in the parents of both children, and also in some other family members, consistent with an autosomal incompletely recessive inheritance of the defect. Some of the presumptive heterozygotes had a history of light bleeding phenomenons; whether this was related to their partial lack of factor XIII is so far uncertain.


2005 ◽  
Vol 83 (4) ◽  
pp. 497-504 ◽  
Author(s):  
Benoit Coulombe ◽  
Marie-France Langelier

High resolution X-ray crystal structures of multisubunit RNA polymerases (RNAP) have contributed to our understanding of transcriptional mechanisms. They also provided a powerful guide for the design of experiments aimed at further characterizing the molecular stages of the transcription reaction. Our laboratory used tandem-affinity peptide purification in native conditions to isolate human RNAP II variants that had site-specific mutations in structural elements located strategically within the enzyme's catalytic center. Both in vitro and in vivo analyses of these mutants revealed novel features of the catalytic mechanisms involving this enzyme.Key words: RNA polymerase II, transcriptional mechanisms, mutational analysis, mRNA synthesis.


Sign in / Sign up

Export Citation Format

Share Document