Chronic eosinophilic leukaemia

From Compendium of Cancer Genome Aberrations
Jump to navigation Jump to search

Haematolymphoid Tumours (5th ed.)

editHAEM5 Conversion Notes
This page was converted to the new template on 2023-12-07. The original page can be found at HAEM4:Chronic Eosinophilic Leukemia, Not Otherwise Specified.

(General Instructions – The main focus of these pages is the clinically significant genetic alterations in each disease type. Use HUGO-approved gene names and symbols (italicized when appropriate), HGVS-based nomenclature for variants, as well as generic names of drugs and testing platforms or assays if applicable. Please complete tables whenever possible and do not delete them (add N/A if not applicable in the table and delete the examples). Please do not delete or alter the section headings. The use of bullet points alongside short blocks of text rather than only large paragraphs is encouraged. Additional instructions below in italicized blue text should not be included in the final page content. Please also see Author_Instructions and FAQs as well as contact your Associate Editor or Technical Support)

Primary Author(s)*

Chelsea D. Kramish; Daynna J.Wolff

Pending Review*

Cancer Category / Type

Myeloproliferative Neoplasm

Cancer Sub-Classification / Subtype

Definition / Description of Disease

Chronic Eosinophilic Leukemia (CEL) is an autonomous, clonal proliferation of eosinophil precursor cells leading to persistently increased eosinophils in peripheral blood, bone marrow and peripheral tissues. The eosinophil count must be greater than or equal to 1.5 x 10^9/L in the blood with less than 20% blasts in the peripheral blood and bone marrow. Diagnosis of CEL is based upon evidence for clonality of myeloid cells or an increase in myeloblasts in peripheral blood (at least 2%) or bone marrow (at least 5%), in order to distinguish from hypereosinophilic syndrome (HES). [1]

Synonyms / Terminology

Chronic Eosinophilic Leukemia, Not Otherwise Specified (CEL, NOS)

Epidemiology / Prevalence

The true incidence is unknown, due to the difficulty of differentiating CEL from idiopathic HES. However, CEL, NOS appears to be more common in men. The reported median age of occurrence is in the seventh decade of life. [1]

Clinical Features

Put your text here and fill in the table (Instruction: Can include references in the table)

Signs and Symptoms EXAMPLE Asymptomatic (incidental finding on complete blood counts)

EXAMPLE B-symptoms (weight loss, fever, night sweats)

EXAMPLE Fatigue

EXAMPLE Lymphadenopathy (uncommon)

Laboratory Findings EXAMPLE Cytopenias

EXAMPLE Lymphocytosis (low level)


editv4:Clinical Features
The content below was from the old template. Please incorporate above.

CEL, NOS is sometimes discovered incidentally in otherwise asymptomatic individuals. In other instances, patients experience constitutional symptoms including weight loss, night sweats, fever, fatigue, cough, angioedema, muscle pain, pruritis and diarrhea. Endomyocardial fibrosis with ensuing restrictive cardiomegaly is known to precipitate the most severe clinical presentation. Scarring of the bicuspid and mitral valves can lead to valvular regurgitation and intracardiac thrombi formation which may embolize to the brain and other organs. Cardiac failure may also occur. Peripheral neuropathy, central nervous system dysfunction and pulmonary symptoms due to lung infiltration as well as rheumatological findings are also common. [1]

Sites of Involvement

Peripheral blood and bone marrow are always involved. Tissue damage secondary to infiltration by eosinophils and the release of cytokines and humoral factors from eosinophil granules may lead to involvement of the heart, lungs, central nervous system, skin, liver, spleen and gastrointestinal tract. [1]

Morphologic Features

Microscopic evaluation of CEL shows eosinophilia in the peripheral blood with small numbers of eosinophilic myelocytes and promyelocytes. Eosinophils are mainly mature and may have sparse granulation, cytoplasmic vacuolation, nuclear hypersegmentation or hyposegmentation, and increased size. Of note, these characteristics alone may not be able to differentiate CEL, NOS from reactive eosinophilia. Significant dysplasia of other myeloid cells contributes toward the diagnosis of CEL, NOS. Neutrophilia and mild basophilia have been reported as well. Blast cells may be present but compose less than 20% of the cells. Greater or equal to 2% myeloblasts in peripheral blood is suggestive of CEL, NOS. [1]

Bone marrow is hypercellular and in most cases eosinophil maturation is orderly. Erythropoiesis and megakaryoctopoiesis are typically normal. Marrow fibrosis is present in approximately one third of cases. 5-20% myeloblasts in bone marrow is suggestive of CEL, NOS. Charcot-laden crystals are often present. [1]

Immunophenotype

No specific immunophenotype has been identified.

Finding Marker
Positive (universal) EXAMPLE CD1
Positive (subset) EXAMPLE CD2
Negative (universal) EXAMPLE CD3
Negative (subset) EXAMPLE CD4

Chromosomal Rearrangements (Gene Fusions)

Put your text here and fill in the table

Chromosomal Rearrangement Genes in Fusion (5’ or 3’ Segments) Pathogenic Derivative Prevalence Diagnostic Significance (Yes, No or Unknown) Prognostic Significance (Yes, No or Unknown) Therapeutic Significance (Yes, No or Unknown) Notes
EXAMPLE t(9;22)(q34;q11.2) EXAMPLE 3'ABL1 / 5'BCR EXAMPLE der(22) EXAMPLE 20% (COSMIC)

EXAMPLE 30% (add reference)

Yes No Yes EXAMPLE

The t(9;22) is diagnostic of CML in the appropriate morphology and clinical context (add reference). This fusion is responsive to targeted therapy such as Imatinib (Gleevec) (add reference).


editv4:Chromosomal Rearrangements (Gene Fusions)
The content below was from the old template. Please incorporate above.

No single or specific genetic abnormality has been identified in CEL, NOS. Rearrangement of PDGFRA, PDGFRB, or FGFR1 excludes the diagnosis of CEL, NOS. PCM1-JAK2, ETV6-JAK2, or BCR-JAK2 are also specifically excluded. [1] Three unique cases of myeloid/lymphoid neoplasm with eosinophilia have shown FLT3 rearrangement: one with t(13;14)(q12;q32)/TRIP11-FLT3 rearrangement, and two with ETV6-FLT3. Eosinophilia and FLT3 rearrangement typically shows myeloproliferative neoplasms, most frequently CEL, NOS, and T-ALL. [5] A unique case of CEL,NOS with a novel fusion gene between exon 22 of GCC2 and exon 12 of PDGFRB was detected and confirmed by PCR in a 54 year old man presenting with cough and dyspnea. [6]

Chromosomal Rearrangement Genes in Fusion (5’ or 3’ Segments) Pathogenic Derivative Prevalence
EXAMPLE t(9;22)(q34;q11.2) EXAMPLE 3'ABL1 / 5'BCR EXAMPLE der(22) EXAMPLE 5%
EXAMPLE t(8;21)(q22;q22) EXAMPLE 5'RUNX1 / 3'RUNXT1 EXAMPLE der(8) EXAMPLE 5%


editv4:Clinical Significance (Diagnosis, Prognosis and Therapeutic Implications).
Please incorporate this section into the relevant tables found in:
  • Chromosomal Rearrangements (Gene Fusions)
  • Individual Region Genomic Gain/Loss/LOH
  • Characteristic Chromosomal Patterns
  • Gene Mutations (SNV/INDEL)

Although survival is variable, acute transformation is common and prognosis is typically poor. The median survival time in one small series was 22.2 months. Response to imatinib has been reported but is uncommon. Treatment with Interferon alpha leading to cytogenic remission has been reported in 3 cases with translocations with a 5q31-33 breakpoint. Complete hematologic response has been achieved in one reported case of CEL with ETV6/FLT3 fusion, with the off-label use of FLT3-inhibitor, sorafenib.[2] Unfavorable prognostic findings include marked splenomegaly, blasts in the blood or increased blasts in bone marrow, cytogenetic abnormalities, and dysplastic features in other myeloid lineages. [1] According to a study of 17 cases defined by WHO-2016 standards, univariate survival analysis showed predictors of inferior survival included megakaryocytic atypia (P = .01), peripheral blood eosinophilic atypia (P = .024), LDH (P = .046) and abnormal karyotype (P = .020). [4] Of these patients, the most frequently utilized first line agents were hydroxyurea as a single agent or in combination with steroids, steroids as a single agent, or in combination. Half of patients treated with hydroxyurea based regimens responded with a persistent decline in eosinophil count over 17.5 months. Approximately one third of patients demonstrated response to steroids for a median duration of 13 months. Three of these patients were treated with imatinib of which two had normalization of eosinophil count. [4] Imatinib was successful in treatment of the individual case of CEL,NOS with novel fusion gene involving PDGFRB and GCC2 with disappearance of the fusion gene in bone marrow after three months. [6]

Individual Region Genomic Gain / Loss / LOH

Put your text here and fill in the table (Instructions: Includes aberrations not involving gene fusions. Can include references in the table. Can refer to CGC workgroup tables as linked on the homepage if applicable.)

Chr # Gain / Loss / Amp / LOH Minimal Region Genomic Coordinates [Genome Build] Minimal Region Cytoband Diagnostic Significance (Yes, No or Unknown) Prognostic Significance (Yes, No or Unknown) Therapeutic Significance (Yes, No or Unknown) Notes
EXAMPLE

7

EXAMPLE Loss EXAMPLE

chr7:1- 159,335,973 [hg38]

EXAMPLE

chr7

Yes Yes No EXAMPLE

Presence of monosomy 7 (or 7q deletion) is sufficient for a diagnosis of AML with MDS-related changes when there is ≥20% blasts and no prior therapy (add reference).  Monosomy 7/7q deletion is associated with a poor prognosis in AML (add reference).

EXAMPLE

8

EXAMPLE Gain EXAMPLE

chr8:1-145,138,636 [hg38]

EXAMPLE

chr8

No No No EXAMPLE

Common recurrent secondary finding for t(8;21) (add reference).

editv4:Genomic Gain/Loss/LOH
The content below was from the old template. Please incorporate above.
Chromosome Number Gain/Loss/Amp/LOH Region
EXAMPLE 8 EXAMPLE Gain EXAMPLE chr8:0-1000000
EXAMPLE 7 EXAMPLE Loss EXAMPLE chr7:0-1000000

Characteristic Chromosomal Patterns

Put your text here (EXAMPLE PATTERNS: hyperdiploid; gain of odd number chromosomes including typically chromosome 1, 3, 5, 7, 11, and 17; co-deletion of 1p and 19q; complex karyotypes without characteristic genetic findings; chromothripsis)

Chromosomal Pattern Diagnostic Significance (Yes, No or Unknown) Prognostic Significance (Yes, No or Unknown) Therapeutic Significance (Yes, No or Unknown) Notes
EXAMPLE

Co-deletion of 1p and 18q

Yes No No EXAMPLE:

See chromosomal rearrangements table as this pattern is due to an unbalanced derivative translocation associated with oligodendroglioma (add reference).

editv4:Characteristic Chromosomal Aberrations / Patterns
The content below was from the old template. Please incorporate above.

A recurrent karyotypic abnormality typically observed in myeloid disorders such as gain of chromosome 8, loss of chromosome 7 or isochromosome 17q supports a diagnosis as well as the presence of a translocation. [1] [1] Morsia et al demonstrated cytogenetic abnormalities in 15 of 17 (88.2%) patients diagnosed with CEL, NOS including trisomy 8 (n = 4), and complex karyotype (n = 3). [4]

Gene Mutations (SNV / INDEL)

Put your text here and fill in the table (Instructions: This table is not meant to be an exhaustive list; please include only genes/alterations that are recurrent and common as well either disease defining and/or clinically significant. Can include references in the table. For clinical significance, denote associations with FDA-approved therapy (not an extensive list of applicable drugs) and NCCN or other national guidelines if applicable; Can also refer to CGC workgroup tables as linked on the homepage if applicable as well as any high impact papers or reviews of gene mutations in this entity.)

Gene; Genetic Alteration Presumed Mechanism (Tumor Suppressor Gene [TSG] / Oncogene / Other) Prevalence (COSMIC / TCGA / Other) Concomitant Mutations Mutually Exclusive Mutations Diagnostic Significance (Yes, No or Unknown) Prognostic Significance (Yes, No or Unknown) Therapeutic Significance (Yes, No or Unknown) Notes
EXAMPLE: TP53; Variable LOF mutations

EXAMPLE:

EGFR; Exon 20 mutations

EXAMPLE: BRAF; Activating mutations

EXAMPLE: TSG EXAMPLE: 20% (COSMIC)

EXAMPLE: 30% (add Reference)

EXAMPLE: IDH1 R123H EXAMPLE: EGFR amplification EXAMPLE:  Excludes hairy cell leukemia (HCL) (add reference).


Note: A more extensive list of mutations can be found in cBioportal (https://www.cbioportal.org/), COSMIC (https://cancer.sanger.ac.uk/cosmic), ICGC (https://dcc.icgc.org/) and/or other databases. When applicable, gene-specific pages within the CCGA site directly link to pertinent external content.


editv4:Gene Mutations (SNV/INDEL)
The content below was from the old template. Please incorporate above.

JAK2 mutations have been identified, however mutations in ASXL1, TET2 and EZH2 appear to be common. [1] The study by Morsia et al. of 17 CEL patients demonstrated two patients each with 13q, 20q deletion, and chromosome 1 abnormalities, one patient with monosomy 7 and one with 3q deletion. All seven patients with NGS studies harbored one or more mutations; ASXL1 (42.9%); IDH1 (28.6%), and one each (14.3%) with TP53, SRSF2, SH2B3, STAT5B, KDM6A and NF1 mutations. [4] A novel JAK2 exon 13 insertion/deletion mutant has been identified and described by Patel et al. in a patient fulfilling diagnostic criteria for both PV and CEL. This study demonstrated that JAK2ex13InDel bears mechanistic resemblance to JAK2V617F but can activate STAT5 in the absence of βc family cytokines IL-3, IL-5, and GM-CSF, potentially promoting eosinophilic differentiation. [7] Keleman et al. discussed STAT5B mutations as reported in four cases: two cases of CEL, NOS; one case of CMML with eosinophilia; and one case of MDS with eosinophilia, respectively. While the presence of a STAT5B N642H mutation may be a potential marker of chronic eosinophilic neoplasms, similar mutations have been described in nonclonal HE and may not be independently sufficient to establish a diagnosis of CEL, NOS. [8]

Gene Mutation Oncogene/Tumor Suppressor/Other Presumed Mechanism (LOF/GOF/Other; Driver/Passenger) Prevalence (COSMIC/TCGA/Other)
EXAMPLE TP53 EXAMPLE R273H EXAMPLE Tumor Suppressor EXAMPLE LOF EXAMPLE 20%

Other Mutations

Type Gene/Region/Other
Concomitant Mutations EXAMPLE IDH1 R123H
Secondary Mutations EXAMPLE Trisomy 7
Mutually Exclusive EXAMPLE EGFR Amplification

Epigenomic Alterations

Genes and Main Pathways Involved

Put your text here and fill in the table (Instructions: Can include references in the table.)

Gene; Genetic Alteration Pathway Pathophysiologic Outcome
EXAMPLE: BRAF and MAP2K1; Activating mutations EXAMPLE: MAPK signaling EXAMPLE: Increased cell growth and proliferation
EXAMPLE: CDKN2A; Inactivating mutations EXAMPLE: Cell cycle regulation EXAMPLE: Unregulated cell division
EXAMPLE:  KMT2C and ARID1A; Inactivating mutations EXAMPLE:  Histone modification, chromatin remodeling EXAMPLE:  Abnormal gene expression program
editv4:Genes and Main Pathways Involved
The content below was from the old template. Please incorporate above.

Genetic Diagnostic Testing Methods

A detailed history, physical exam, blood count and examination of blood smear are key diagnostic measures. The process of making a diagnosis heavily relies on the exclusion of reactive eosinophilia, and myeloid neoplasms with the previously mentioned rearrangements or fusions. [1] Wang et al. demonstrated that targeted next generation sequencing helps to establish clonality in a portion of patients with hypereosinophilia that would otherwise be diagnosed with idiopathic hypereosinophilic syndrome. [3]

Familial Forms

Additional Information

Links

https://ccga.io/index.php/Chronic_Eosinophilic_Leukemia,_Not_Otherwise_Specified?veaction=edit&section=2

References

  1. Bain B.J, et al., (2017). Chronic eosinophilic leukemia, NOS, in World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues, Revised 4th edition. Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, Arber DA, Hasserjian RP, Le Beau MM, Orazi A, and Siebert R, Editors. IARC Press: Lyon, France, p54-56
  2. Ricci, F., Balducci, S., Guerrini, F., Grassi, S., Ciabatti, E., Baratè, C., . . . Galimberti, S. (2020). Sorafenib Induced Complete Cytogenetic and Molecular Response in a Chronic Eosinophilic Leukemia Case with t(12;13) Translocation. Clinical Hematology International, 2. doi:10.2991/chi.k.200714.001
  3. Wang SA, Tam W, Tsai AG, Arber DA, Hasserjian RP, Geyer JT, George TI, Czuchlewski DR, Foucar K, Rogers HJ, Hsi ED, Bryan Rea B, Bagg A, Dal Cin P, Zhao C, Kelley TW, Verstovsek S, Bueso-Ramos C, Orazi A. Targeted next-generation sequencing identifies a subset of idiopathic hypereosinophilic syndrome with features similar to chronic eosinophilic leukemia, not otherwise specified. Mod Pathol. 2016 Aug;29(8):854-64. doi: 10.1038/modpathol.2016.75. Epub 2016 May 13. PMID: 27174585.
  4. Morsia, E., Reichard, K., Pardanani, A., Tefferi, A., & Gangat, N. (2020). WHO defined chronic eosinophilic leukemia, not otherwise specified ( CEL , NOS ): A contemporary series from the Mayo Clinic. American Journal of Hematology, 95(7). doi:10.1002/ajh.25811
  5. Shao, H., Wang, W., Song, J., Tang, G., Zhang, X., Tang, Z., . . . Zhang, L. (2020). Myeloid/lymphoid neoplasms with eosinophilia and FLT3 rearrangement. Leukemia Research, 99, 106460. doi:10.1016/j.leukres.2020.106460
  6. Iriyama N, Takahashi H, Naruse H, Miura K, Uchino Y, Nakagawa M, Iizuka K, Hamada T, Hatta Y, Nakayama T, Takei M. A novel fusion gene involving PDGFRB and GCC2 in a chronic eosinophilic leukemia patient harboring t(2;5)(q37;q31). Mol Genet Genomic Med. 2019 Apr;7(4):e00591. doi: 10.1002/mgg3.591. Epub 2019 Jan 29. PMID: 30697976; PMCID: PMC6465652.
  7. Patel AB, Franzini A, Leroy E, Kim SJ, Pomicter AD, Genet L, Xiao M, Yan D, Ahmann JM, Agarwal AM, Clair P, Addada J, Lambert J, Salmon M, Gleich GJ, Cross NCP, Constantinescu SN, O'Hare T, Prchal JT, Deininger MW. JAK2 ex13InDel drives oncogenic transformation and is associated with chronic eosinophilic leukemia and polycythemia vera. Blood. 2019 Dec 26;134(26):2388-2398. doi: 10.1182/blood.2019001385. PMID: 31697804; PMCID: PMC6933291.
  8. Katalin Kelemen, MD, PhD, Leonie Saft, MD, Fiona E Craig, MD, Attilio Orazi, MD, Megan Nakashima, MD, Gerald B Wertheim, MD, PhD, Tracy I George, MD, Hans-Peter Horny, MD, Rebecca L King, MD, Leticia Quintanilla-Martinez, MD, Sa A Wang, MD, Lisa M Rimsza, MD, Kaaren K Reichard, MD, Eosinophilia/Hypereosinophilia in the Setting of Reactive and Idiopathic Causes, Well-Defined Myeloid or Lymphoid Leukemias, or Germline Disorders: Report of the 2019 Society for Hematopathology/European Association for Haematopathology Workshop, American Journal of Clinical Pathology, , aqaa244, https://doi.org/10.1093/ajcp/aqaa244


Notes

*Primary authors will typically be those that initially create and complete the content of a page.  If a subsequent user modifies the content and feels the effort put forth is of high enough significance to warrant listing in the authorship section, please contact the CCGA coordinators (contact information provided on the homepage).  Additional global feedback or concerns are also welcome. *Citation of this Page: “Chronic eosinophilic leukaemia”. Compendium of Cancer Genome Aberrations (CCGA), Cancer Genomics Consortium (CGC), updated 12/13/2023, https://ccga.io/index.php/HAEM5:Chronic_eosinophilic_leukaemia.