Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
1.
Hum Pathol ; 149: 66-74, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38879086

ABSTRACT

CSF3R activating mutation is a genetic hallmark of chronic neutrophilic leukemia (CNL), and is also present in a subset of atypical chronic myeloid leukemia (aCML), but infrequent in other myeloid neoplasms. However, the occurrence of CSF3R mutations in various myeloid neoplasms is not well studied. Here we evaluate the spectrum of CSF3R mutations and the clinicopathologic features of CSF3R mutated myeloid neoplasms. We retrospectively identified CSF3R mutations in a variety of myeloid neoplasms: two CNL, three atypical chronic myeloid leukemia (aCML), nine acute myeloid leukemia (AML), one chronic myelomonocytic leukemia, and one myeloproliferative neoplasm. The prototypic T618I mutation was found in 50% of cases: CNL (2/2), aCML (2/3) and AML (4/9). We observed a new recurrent CSF3R mutation Q776* in 25% of cases, and a potential-germline mutation in a 20-year-old patient. Co-occurring mutations were often in epigenetic modifier and spliceosome. IDH/RUNX1 and tumor suppressor mutations were frequent in AML but absent in CNL/aCML. All CNL/aCML patients succumbed within 2-years of diagnosis. We demonstrate that CSF3R mutations are not restricted to CNL. CNL and aCML show similar clinicopathologic and molecular features, suggesting that CNL may be best classified as myelodysplastic/myeloproliferative neoplasm rather than myeloproliferative neoplasm.


Subject(s)
Leukemia, Neutrophilic, Chronic , Mutation , Receptors, Colony-Stimulating Factor , Humans , Receptors, Colony-Stimulating Factor/genetics , Male , Middle Aged , Female , Aged , Leukemia, Neutrophilic, Chronic/genetics , Leukemia, Neutrophilic, Chronic/pathology , Retrospective Studies , Adult , Young Adult , Aged, 80 and over , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , DNA Mutational Analysis , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Genetic Predisposition to Disease , Biomarkers, Tumor/genetics , Phenotype
2.
Ann Diagn Pathol ; 71: 152317, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38642470

ABSTRACT

We report a series of patients with CSF3R-mutant (CSF3Rmut) atypical chronic myeloid leukemia (aCML), chronic neutrophilic leukemia (CNL) or other hematologic malignancies. We included 25 patients: 5 aCML and 4 CNL CSF3Rmut patients; 1 aCML, 2 CNL, and 2 myelodysplastic/myeloproliferative neoplasm, not otherwise specified patients without CSF3R mutation; and 11 CSF3Rmut patients with other diseases [8 acute myeloid leukemia (AML), 1 chronic myelomonocytic leukemia (CMML), 1 myelodysplastic syndrome (MDS), and 1 acute lymphoblastic leukemia (ALL)]. Patients with aCML or CNL were tested by Sanger sequencing and pyrosequencing to identify CSF3R T618I. Twenty-two patients underwent gene panel analysis. CSF3R mutations, mostly T618I (8/9), were found at high frequencies in both aCML and CNL patients [5/6 aCML and 4/6 CNL]. Two aCML patients in early adulthood with CSF3R T618I and biallelic or homozygous CEBPA mutations without other mutations presented with increased blasts and exhibited remission for >6 years after transplantation. The other 7 CSF3Rmut aCML or CNL patients were elderly adults who all had ASXL1 mutations and frequently presented with SEBP1 and SRSF2 mutations. Five AML patients had CSF3R exon 14 or 15 point mutations, and 6 other patients (3 AML, 1 CMML, 1 MDS, and 1 ALL) had truncating mutations, demonstrating differences in leukocyte counts and mutation status. In conclusion, CSF3R mutations were found at a higher frequency in aCML patients than in previous studies, which might reflect ethnic differences. Additional studies are needed to confirm these findings and the relationship between CSF3R and CEBPA mutations.


Subject(s)
Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative , Mutation , Receptors, Colony-Stimulating Factor , Humans , Receptors, Colony-Stimulating Factor/genetics , Male , Female , Middle Aged , Aged , Adult , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Aged, 80 and over , Leukemia, Neutrophilic, Chronic/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology
3.
Leuk Lymphoma ; 64(9): 1566-1573, 2023 09.
Article in English | MEDLINE | ID: mdl-37395413

ABSTRACT

Chronic myelomonocytic leukemia (CMML) is a myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) chacaterized by persistent peripheral blood monocytosis, hypercellular bone marrow and dysplasia at least in one myeloid lineage. CMML shares much of its molecular landscape with other myeloid neoplasms, while differs from others such as chronic neutrophilic leukemia (CNL), given the high frequency of CSF3R mutations in the latter. In this article, we report a case of CSF3R-mutated CMML and dissect this rare entity by reviewing the medical literature, with the intent to understand how this rare mutation shapes CMML's clinical and morphological phenotype. CSF3R-mutated CMML emerges as a rare entity meeting the ICC/WHO diagnostic criteria for CMML and simultaneously showing clinical-pathological and molecular traits of CNL and atypical chronic myeloid leukemia, rising an important and difficult diagnostic and therapeutical issue.


Subject(s)
Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative , Leukemia, Myelomonocytic, Chronic , Leukemia, Neutrophilic, Chronic , Myeloproliferative Disorders , Humans , Leukemia, Myelomonocytic, Chronic/diagnosis , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/pathology , Leukemia, Neutrophilic, Chronic/diagnosis , Leukemia, Neutrophilic, Chronic/genetics , Mutation , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Prognosis , Receptors, Colony-Stimulating Factor/genetics
4.
Mod Pathol ; 35(4): 470-479, 2022 04.
Article in English | MEDLINE | ID: mdl-34775472

ABSTRACT

Classification of myeloid neoplasms with isolated isochromosome i(17q) [17p deletion with inherent monoallelic TP53 loss plus 17q duplication] is controversial. Most cases fall within the WHO unclassifiable myelodysplastic/myeloproliferative neoplasms (MDS/MPN-U) category. The uniformly dismal outcomes warrant better understanding of this entity. We undertook a multi-institutional retrospective study of 92 adult MDS/MPN-U cases from eight institutions. Twenty-nine (32%) patients had isolated i(17q) [MDS/MPN-i(17q)]. Compared to MDS/MPN without i(17q), MDS/MPN-i(17q) patients were significantly younger, had lower platelet and absolute neutrophil counts, and higher frequency of splenomegaly and circulating blasts. MDS/MPN-i(17q) cases showed frequent bilobed neutrophils (75% vs. 23%; P = 0.03), hypolobated megakaryocytes (62% vs. 20%; P = 0.06), and a higher frequency of SETBP1 (69% vs. 5%; P = 0.002) and SRSF2 (63% vs. 5%; P = 0.006) mutations that were frequently co-existent (44% vs. 0%; P = 0.01). TP53 mutations were rare. The mutation profile of MDS/MPN-U-i(17q) was similar to other myeloid neoplasms with i(17q) including atypical chronic myeloid leukemia, chronic myelomonocytic leukemia, myelodysplastic/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis, myelodysplastic syndrome and acute myeloid leukemia, with frequent concomitant SETBP1/SRSF2 mutations observed across all the diagnostic entities. Over a median follow-up of 52 months, patients with MDS/MPN-i(17q) showed a shorter median overall survival (11 vs. 28 months; P < 0.001). The presence of i(17q) retained independent poor prognostic value in multivariable Cox-regression analysis [HR 3.686 (1.17-11.6); P = 0.026] along with splenomegaly. We suggest that MDS/MPN-i(17q) warrants recognition as a distinct subtype within the MDS/MPN-U category based on its unique clinico-biologic features and uniformly poor prognosis.


Subject(s)
Biological Products , Isochromosomes , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative , Adult , Bone Marrow/pathology , Humans , Isochromosomes/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/diagnosis , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Mutation , Retrospective Studies
5.
Cancer Genet ; 258-259: 69-73, 2021 11.
Article in English | MEDLINE | ID: mdl-34551378

ABSTRACT

Myeloid/lymphoid neoplasm with t(8;22)(p11.2;q11.2)/BCR-FGFR1 is an extremely rare diagnosis, with few reported cases to date. In contrast to other FGFR1-partner rearrangements that are associated with chronic eosinophilic leukemia, acute myeloid leukemia, and/or lymphoblastic lymphoma, patients with BCR-FGFR1 have a myeloproliferative disorder that closely resembles chronic myeloid leukemia (CML). The current report describes a rare case of a 61 year old man with an atypical CML phenotype associated with t(8;22)(p11.2;q11.2)/BCR-FGFR1. A literature review is presented to enhance the awareness of this rare diagnostic entity.


Subject(s)
Chromosomes, Human, Pair 22/genetics , Chromosomes, Human, Pair 8/genetics , Gene Rearrangement , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Proto-Oncogene Proteins c-bcr/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Translocation, Genetic , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Male , Middle Aged , Prognosis
6.
Int J Mol Sci ; 22(2)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33429941

ABSTRACT

The last decade has been very important for the quantity of preclinical information obtained regarding chronic myeloproliferative neoplasms (MPNs) and the following will be dedicated to the translational implications of the new biological acquisitions. The overcoming of the mechanistic model of clonal evolution and the entry of chronic inflammation and dysimmunity into the new model are the elements on which to base a part of future therapeutic strategies. The innate immune system plays a major role in this context. Protagonists of the initiation and regulation of many pathological aspects, from cytokine storms to fibrosis, the NLRP3 and AIM2 inflammasomes guide and condition the natural history of the disease. For this reason, MPNs share many biological and clinical aspects with non-neoplastic diseases, such as autoimmune disorders. Finally, cardiovascular risk and disturbances in iron metabolism and myelopoiesis are also closely linked to the role of inflammasomes. Although targeted therapies are already being tested, an increase in knowledge on the subject is desirable and potentially translates into better care for patients with MPNs.


Subject(s)
DNA-Binding Proteins/genetics , Inflammation/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Clonal Evolution , Humans , Inflammasomes/genetics , Inflammation/pathology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology
7.
BMC Cancer ; 20(1): 650, 2020 Jul 13.
Article in English | MEDLINE | ID: mdl-32660441

ABSTRACT

BACKGROUND: The identification of pathologically altered neutrophil granulocyte migration patterns bears strong potential for surveillance and prognostic scoring of diseases. We recently identified a strong correlation between impaired neutrophil motility and the disease stage of myelodysplastic syndrome (MDS). Here, we apply this assay to study quantitively increased neutrophils of a patient suffering from a rare leukemia subtype, atypical chronic myeloid leukemia (aCML). METHODS: A 69-year-old male was analyzed in this study. Besides routine analyses, we purified the patient's neutrophils from peripheral whole blood and studied their migration behavior using time-lapse video microscopy in a standardized assay. These live cell migration analyses also allowed for the quantification of cell morphology. Furthermore, the cells were stained for the markers CD15, CD16, fMLPR, CXCR1 and CXCR2. RESULTS: Despite cytoreductive therapy with hydroxyurea, the patient's WBC and ANC were poorly controlled and severe dysgranulopoiesis with hypogranularity was observed. Neutrophils displayed strongly impaired migration when compared to healthy controls and migrating cells exhibited a more flattened-out morphology than control neutrophils. Because of a detected CSF3R (p.T618I) mutation and constitutional symptoms treatment with ruxolitinib was initiated. Within 1 week of ruxolitinib treatment, the cell shape normalized and remained indistinguishable from healthy control neutrophils. However, neutrophil migration did not improve over the course of ruxolitinib therapy but was strikingly altered shortly before a sinusitis with fever and bleeding from a gastric ulcer. Molecular work-up revealed that under ruxolitinib treatment, the CSF3R clone was depleted, yet the expansion of a NRAS mutated subclone was promoted. CONCLUSION: These results demonstrate the usefulness of neutrophil migration analyses to uncover corresponding alterations of neutrophil migration in rare myeloid neoplasms. Furthermore, in addition to monitoring migration the determination of morphological features of live neutrophils might represent a useful tool to monitor the effectiveness of therapeutic approaches.


Subject(s)
Biomarkers, Tumor/genetics , Cell Movement , Granulocytes/pathology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/drug therapy , Neutrophils/pathology , Pyrazoles/adverse effects , Aged , Case-Control Studies , Female , Granulocytes/drug effects , Granulocytes/metabolism , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Longitudinal Studies , Male , Neutrophils/drug effects , Neutrophils/metabolism , Nitriles , Prognosis , Pyrimidines
8.
Best Pract Res Clin Haematol ; 33(2): 101133, 2020 06.
Article in English | MEDLINE | ID: mdl-32460981

ABSTRACT

Atypical chronic myeloid leukemia is an esoteric myeloid malignancy with features of both myeloproliferative and myelodysplastic syndromes. This disease is characterized primarily by morphologic-based criteria, and has clinical and molecular features overlapping with other myeloid malignancies. No one molecular abnormality is specific, and multiple mutations are often present in various combinations, due to the malignant multi-step clonal evolution of myeloid malignancies. In this review, we will address what we know about atypical chronic myeloid leukemia; evaluate how the molecular landscape in myeloid malignancies overlaps, and discuss what we can learn by incorporating individualized precision genomic strategies.


Subject(s)
Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Myelodysplastic Syndromes/genetics , Genomics , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/metabolism , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology
9.
Intern Med ; 59(12): 1549-1553, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32188810

ABSTRACT

Patients with myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) are often asymptomatic and thus can remain undiagnosed until they become symptomatic due to progression to the accelerated phase (AP) or transformation to acute leukemia (leukemic transformation; LT). We herein report the case of a previously healthy 38-year-old man who had hyperleukocytosis with dysplastic myeloid precursor cells and severe disseminated intravascular coagulation. Hematopoietic recovery with features of atypical chronic myeloid leukemia (aCML) after induction chemotherapy was a diagnostic clue. Although rare, this case highlights the limitation of the diagnostic approach for aCML with AP or LT at the initial presentation.


Subject(s)
Disseminated Intravascular Coagulation/complications , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/complications , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Adult , Antineoplastic Agents/therapeutic use , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/drug therapy , Leukocytosis/complications , Male
14.
Leuk Res ; 75: 50-57, 2018 12.
Article in English | MEDLINE | ID: mdl-30458320

ABSTRACT

Atypical chronic myeloid leukemia (aCML) and chronic neutrophilic leukemia (CNL) are rare BCR-ABL1 fusion gene-negative myeloid neoplasms with a predominance of neutrophils. Since no standard therapeutic strategy currently exists for these diseases, we retrospectively evaluated the outcomes of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for aCML and CNL. Data from 14 aCML and 5 CNL patients as their diagnoses were collected using a nationwide survey. Allo-HSCT was performed between 2003 and 2014. Preconditioning regimens included myeloablative (n = 15), reduced-intensity (n = 3), and non-myeloablative (n = 1) regimens. Transplanted stem cells were obtained from HLA-matched related donors (n = 5) and alternative donors (n = 14). Neutrophil engraftment was successfully achieved in 17 patients. One-year overall survival rates (OS) were 54.4% (95% confidence interval [CI], 24.8 to 76.7%) and 40.0% (95% CI, 5.2 to 75.3%) in patients with aCML and CNL, respectively. Among aCML patients, 1-year OS were 76.2% (95% CI, 33.2 to 93.5%) and 20.0% (95% CI, 0.8 to 58.2%) in patients with <5% myeloblasts (n = 9) and ≥5% myeloblasts (n = 5) in peripheral blood before allo-HSCT, respectively. These results suggest that allo-HSCT achieves long-term survival in patients with aCML and CNL. Better pre-transplant management is required to improve the outcomes of aCML patients with ≥5% blasts in peripheral blood.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/therapy , Leukemia, Neutrophilic, Chronic/therapy , Adolescent , Adult , Aged , Child , Disease-Free Survival , Female , Humans , Japan , Kaplan-Meier Estimate , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/mortality , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Leukemia, Neutrophilic, Chronic/mortality , Leukemia, Neutrophilic, Chronic/pathology , Male , Middle Aged , Retrospective Studies , Transplantation, Homologous , Young Adult
15.
Eur J Haematol ; 101(1): 57-67, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29645296

ABSTRACT

OBJECTIVE: Philadelphia-negative myeloproliferative neoplasms (MPNs) commonly share hyperactive JAK-STAT signaling affecting hematopoietic stem cells (HSC) and their progeny. The JAK1/2 inhibitor Ruxolitinib has remarkable clinical efficacy, including spleen reduction, improvement of constitutional symptoms, and bone marrow (BM) fibrosis reversal. Whether this is due to inhibition of JAK2-mutated HSC only, or whether Ruxolitinib also affects BM stroma is not known. METHODS: This study investigated potential effects of Ruxolitinib on BM mesenchymal stromal cells (MSC), which are not only major regulators of hematopoiesis but also contribute to fibrosis, from 10 healthy donors and 7 JAK2V617F -positive MPN patients. RESULTS: Ruxolitinib moderately inhibited the growth of healthy donor MSC (HD-MSC) and MSC from JAK2V617F+ MPN patients (P-MSC) in short- and long-term assays. The clonogenic potential of HD-MSC was not affected by Ruxolitinib. JAK-STAT signaling, however, was markedly inhibited in both HD-MSC and P-MSC, the latter of which showed higher expression of fibrosis-associated and hematopoiesis-maintenance genes. Moreover, Ruxolitinib reduced MSC secretion of MCP-1 and IL-6. CONCLUSION: Ruxolitinib affected JAK2 signaling in MSC at clinically relevant doses, which is likely to contribute to the normalization of the inflammatory milieu in MPNs. Thus, combined HSC and stroma-directed interventions have the potential to improve constitutional symptoms and reduce stromal proliferation in MPNs.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/drug effects , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Aged , Aged, 80 and over , Bone Marrow/enzymology , Bone Marrow/immunology , Bone Marrow/pathology , Case-Control Studies , Cell Proliferation/drug effects , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Female , Fibrosis , Hematopoietic Stem Cells/enzymology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Interleukin-6/genetics , Interleukin-6/immunology , Janus Kinase 1/genetics , Janus Kinase 1/immunology , Janus Kinase 2/genetics , Janus Kinase 2/immunology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/enzymology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/immunology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Male , Middle Aged , Mutation , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/immunology , Myeloproliferative Disorders/pathology , Nitriles , Primary Cell Culture , Pyrimidines , Signal Transduction
17.
Psychosomatics ; 59(5): 472-480, 2018.
Article in English | MEDLINE | ID: mdl-29506868

ABSTRACT

BACKGROUND: The physical symptom burden of patients with myeloproliferative neoplasms (MPNs) may last for extended periods during their disease trajectories and lead to psychologic distress, anxiety, or depression or all of these. OBJECTIVE: This study evaluated the relationship between physical symptom burden captured by the Physical Problem List (PPL) on the Distress Thermometer and Problem List and psychologic outcomes (distress, anxiety, and depression) in the MPN setting. METHODS: Patients (N = 117) with MPNs completed questionnaires containing the Distress Thermometer and Problem List and the Hospital Anxiety and Depression Scale in a dedicated MPN clinic within an academic medical center. They reported symptoms from any of 22 physical problems on the PPL. Items endorsed by more than 10% of participants were assessed for their associations with distress (Distress Thermometer and Problem List), anxiety (Hospital Anxiety and Depression Scale-Anxiety), and depression (Hospital Anxiety and Depression Scale-Depression). The total number of endorsed PPL items per participant was also evaluated. RESULTS: Nine of 22 PPL items (fatigue, sleep, pain, dry skin/pruritus, memory/concentration, feeling swollen, breathing, and sexual) were reported by >10% of participants. In univariate analyses, all PPL items but one were associated with distress and depression, and all but 2 were associated with anxiety. In multivariate analyses, the total number of PPL items was associated with depression only (p < 0.001) when controlling for covariates. CONCLUSION: Physical symptom burden in MPN patients was clearly associated with psychologic symptoms. Depression was uniquely associated with overall physical symptom burden. As such, the endorsement of multiple PPL items on the Distress Thermometer and Problem List should prompt an evaluation for psychologic symptoms to improve MPN patients' overall morbidity and quality of life.


Subject(s)
Anxiety/etiology , Depression/etiology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/psychology , Stress, Psychological/etiology , Anxiety/psychology , Depression/psychology , Female , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Male , Middle Aged , Psychiatric Status Rating Scales , Stress, Psychological/psychology , Surveys and Questionnaires
18.
Future Oncol ; 14(2): 177-185, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29226717

ABSTRACT

The aim of our study was to review the clinicopathologic features and management of atypical chronic myeloid leukemia (aCML). Relevant manuscripts published in English were searched using PubMed. aCML is diagnosed as per WHO 2016 classification in the presence of leukocytosis ≥13 × 109/l with circulating neutrophil precursors ≥10%, monocytes less than 10%, minimal basophils, hypercellular bone marrow with granulocytic proliferation and dysplasia, bone marrow blast less than 20% and absence of BCR/ABL fusion gene. Common cytogenetic features and mutations include trisomy 8, and mutations in SETBP1 and ETNK1. Median survival is 1-2 years. Hematopoietic stem cell transplant may be the only curative option. Ruxolitinib and dasatinib are emerging therapeutic options. Thus, aCML is a rare entity with poor survival. Novel therapies are needed.


Subject(s)
Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/epidemiology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Molecular Targeted Therapy , Neoplastic Cells, Circulating/pathology , Carrier Proteins/genetics , Dasatinib/therapeutic use , Disease Management , Disease-Free Survival , Female , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/therapy , Mutation , Nitriles , Nuclear Proteins/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Pyrazoles/therapeutic use , Pyrimidines
19.
Am J Dermatopathol ; 39(6): 479-484, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27930381

ABSTRACT

One of the pathogenic causes of cutaneous inflammatory pseudotumors is chronic localized fibrosing leukocytoclastic vasculitis (CLFLCV), a vasculitic reaction pattern seen in granuloma faciale (GF), a localized vasculitis, and erythema elevatum diutinum (EED), a generalized vasculitis. Patients with myelodysplastic syndromes (MDSs) are at risk for a diverse spectrum of cutaneous neutrophilic dermatoses such as EED. Herein, we report a 74-year-old man who presented with a large ulcerative, fungating tumor affecting the right flexor ankle caused by CLFLCV. During his workup and management, MDS and Philadelphia chromosome-negative chronic myeloid leukemia was diagnosed. Surgical excision of the inflammatory mass promptly triggered tumor recurrence, whereas antineutrophil therapy (dapsone coupled with hydroxyurea, and prednisone) lead to tumor regression. Histopathologic examination revealed an eosinophilic-rich small-vessel neutrophilic vasculitis associated with storiform and angiocentric fibrosis (GF-like). In the regions of fibrosis, dilated lymphatic and vascular spaces were numerous, some of which were congested with small CD3-positive lymphocytes (intralymphatic and intravascular lymphocytosis). These findings indicate coexisting localized lymphedema. By direct immunofluorescence, IgM and C4d vessel deposits were detected. The pathogenesis of the fibrotic nodules and plaques of CLFLCV is suspected to be due to recurring bouts of immune-complex vasculitis, creating a cycle of vessel damage followed by reparative granulation tissue. Poor lymphatic drainage may be the underlying factor initiating and maintaining recurrent, localized episodes of immune-complex vasculitis and progressive fibrosis. Although his tumor histopathology resembled GF-eosinophilic rich CLFLCV-the clinical context points to a solitary and paraneoplastic case of EED.


Subject(s)
Granuloma, Plasma Cell/pathology , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Lymphedema/pathology , Lymphocytosis/pathology , Paraneoplastic Syndromes/pathology , Vasculitis, Leukocytoclastic, Cutaneous/pathology , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , Dapsone/therapeutic use , Fatal Outcome , Granuloma, Plasma Cell/immunology , Granuloma, Plasma Cell/therapy , Humans , Hydroxyurea/therapeutic use , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/drug therapy , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/immunology , Lymphedema/immunology , Lymphedema/therapy , Lymphocytosis/immunology , Lymphocytosis/therapy , Male , Paraneoplastic Syndromes/immunology , Paraneoplastic Syndromes/therapy , Pentoxifylline/therapeutic use , Prednisone/therapeutic use , Remission Induction , Treatment Outcome , Vasculitis, Leukocytoclastic, Cutaneous/immunology , Vasculitis, Leukocytoclastic, Cutaneous/therapy
20.
Blood ; 129(6): 715-722, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28028025

ABSTRACT

Chronic neutrophilic leukemia (CNL) is a distinct myeloproliferative neoplasm with a high prevalence (>80%) of mutations in the colony-stimulating factor 3 receptor (CSF3R). These mutations activate the receptor, leading to the proliferation of neutrophils that are a hallmark of CNL. Recently, the World Health Organization guidelines have been updated to include CSF3R mutations as part of the diagnostic criteria for CNL. Because of the high prevalence of CSF3R mutations in CNL, it is tempting to think of this disease as being solely driven by this genetic lesion. However, recent additional genomic characterization demonstrates that CNL has much in common with other chronic myeloid malignancies at the genetic level, such as the clinically related diagnosis atypical chronic myeloid leukemia. These commonalities include mutations in SETBP1, spliceosome proteins (SRSF2, U2AF1), and epigenetic modifiers (TET2, ASXL1). Some of these same mutations also have been characterized as frequent events in clonal hematopoiesis of indeterminate potential, suggesting a more complex disease evolution than was previously understood and raising the possibility that an age-related clonal process of preleukemic cells could precede the development of CNL. The order of acquisition of CSF3R mutations relative to mutations in SETBP1, epigenetic modifiers, or the spliceosome has been determined only in isolated case reports; thus, further work is needed to understand the impact of mutation chronology on the clonal evolution and progression of CNL. Understanding the complete landscape and chronology of genomic events in CNL will help in the development of improved therapeutic strategies for this patient population.


Subject(s)
Gene Expression Regulation, Leukemic/drug effects , Genomics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Leukemia, Neutrophilic, Chronic/genetics , Neutrophils/metabolism , Antineoplastic Agents/therapeutic use , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Proliferation/drug effects , Clonal Evolution , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Disease Progression , Humans , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/drug therapy , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/metabolism , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Leukemia, Neutrophilic, Chronic/drug therapy , Leukemia, Neutrophilic, Chronic/metabolism , Leukemia, Neutrophilic, Chronic/pathology , Mutation , Neutrophils/drug effects , Neutrophils/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Practice Guidelines as Topic , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Colony-Stimulating Factor/genetics , Receptors, Colony-Stimulating Factor/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...