Blastic plasmacytoid dendritic cell neoplasm

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:Blastic Plasmacytoid Dendritic Cell Neoplasm.

(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)*

Hao Liu, MD and Daynna J. Wolff, PhD

Cancer Category / Type

Myeloid neoplasms/Acute myeloid leukemia

Cancer Sub-Classification / Subtype

Blastic plasmacytoid dendritic cell neoplasm (BPDCN)

Definition / Description of Disease

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare, clinically aggressive hematologic malignancy that derives from precursors of plasmacytoid dendritic cells (pDCs)[1][2].

Synonyms / Terminology

  • Agranular CD4+ NK cell leukaemia (obsolete)[1]
  • Blastic NK leukaemia/lymphoma (obsolete)[1]
  • Agranular CD4+ CD56+ hematodermic neoplasm/tumor[1][2]

Epidemiology / Prevalence

  • BPDCN is rare, estimated to represent < 1% of all hematologic malignancies[1].
  • The incidence of BPDCN in the USA: 0.04 cases per 100,000 individuals[3].
  • BPDCN has no known racial or ethnic predilection.
  • Though BPDCN can occur at any age, it more commonly occurs in elderly patients with a mean/median patient age at diagnosis of 61-67 years[1][4].
  • It most often affects males, with a male-to-female ratio of 3.3:1[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.

Typical BPDCN patients may have two stages[1]:

o  First stage: affects the skin , usually contained or indolent

o  Second stage:  rapid leukemic spread and multi-organ involvement that eventually leads to death[1].

Sites of Involvement

  • Multiple sites are frequently involved by BPDCN. The three most common are the skin (in 60–100% cases), followed by the bone-marrow and peripheral blood (in 60–90% of cases), and thirdly the lymph nodes (in 40–50% of cases)[1].
  • Upon diagnosis, the central nervous system (CNS) is also frequently found to be involved, and up to one third of patients have CNS involvement at relapse[3].

Morphologic Features

  • BPDCN is commonly characterized by a diffuse, monomorphous infiltrate of small or medium-sized blasts[1].
  • The morphology of the neoplastic cells is similar to lymphoblasts or myeloblasts:  high N: C ratio, eccentrically located nucleus, fine chromatin, a prominent nucleolus and scant amphophilic cytoplasm[1].
  • Mitoses are variable in number, and the Ki-67 rate ranges from 20 to 80%[1].
  • Necrosis may present.

Immunophenotype

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

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


editv4:Immunophenotype
The content below was from the old template. Please incorporate above.
  • BPDCN cells express CD4, CD43, CD45RA, CD56, and the pDC associated antigens, including CD123 (IL3 α chain receptor), CD303, TCL1A, CD2AP, and TCF4[1][2][3].
  • BPDCN is characterized by high expression levels of CD123 and weak expression of CD45[1].
  • BPDCN cells are negative for lineage-specific markers including CD3, CD19, and myeloperoxidase[1][3].
  • BPDCN cells also do not express myeloid cell nuclear differentiation antigen (MNDA)[3].
  • BPDCN cells in some cases variably express CD2, CD5, CD7, CD33, CD38, CD68, CD117, HLA-DR, and TdT[1][3].

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.

A recurrent balanced translocation t(6;8)(p21;q24) involving the MYC locus was exclusively identified in BPDCN[5][6][7][8][9]. The prevalence of MYC translocation in BPDCN is 5% -12%[6]. Rearrangements involving the MYC locus on 8q24 are associated with MYC protein overexpression and specific clinical features, including older onset age and shorter median survival[6]. RUNX2, located on chromosome 6p21, is strongly expressed in pDCs and BPDCN cells. The t(6,8) generates mutant-allele super-enhancer of RUNX2 which may increase the expression of MYC and lead to the development of BPDCN[5]. SUPT3H, a TATA-binding protein-associated factors (TAF)-associated protein, was identified as a novel 8q24/MYC partner in BPDCN[7].

Chromosomal Rearrangement Genes in Fusion (5’ or 3’ Segments) Pathogenic Derivative Prevalence
t(6;8)(p21;q24) 5'RUNX2 super enhancer / 5'MYC increased expression MYC 5-12%[5][6][7][8][9][10]
t(8;var)(q24;var) ? / 5'MYC ?increased expression MYC


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)
  • BPDCN is extremely aggressive, with a median survival of 10-19.8 months[1].
  • Age is an adverse impact factor for prognosis[1].
  • Diagnosis is usually established through skin biopsy with immunohistochemistry or flow cytometry[4].
  • Traditional therapeutic approaches include multi-agent chemotherapy, such as CHOP, hyper-CVAD[2][3][4]. However, the traditional chemotherapy is associated with high relapse rate and death[11].
  • A new targeted therapy, Tagraxofusp (SL-401, ELZONRIS) was recently approved.  This agent is a CD123-directed cytotoxin consisting of recombinant human interleukin-3 fused to a truncated diphtheria toxin[2][4][11].
  • For the patients in first complete remission after induction therapy, allogeneic hematopoietic stem cell transplantation (HSCT) is recommended to achieve long-term survival[1][4].

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.

1).Deletion of the 9p21.3 locus[12]:

·        Most recurrent event in cases of BPDCN

·       Associated with a poor prognosis when biallelic

2).  12p13/ETV6 deletions[13]:

·       Monoallelic or biallelic

·        May represent early clonal events

3). Del(5q), del(7q), del(9q), del(11q), del(12p) and del(13q) are frequently identified in BPDCN patients with myelodysplastic syndrome or acute myeloid leukemia with myelodysplasia-related changes[3].

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.
  • Chromosomal abnormalities are identified in the majority of BPDCN cases; about two thirds of BPDCN patients have an abnormal karyotype[1].
  • About 75% of BPDCN patients have a complex karyotype, which is defined by three or more abnormalities, including at least one structural abnormality[3].
  • Abnormalities involving the short arm of chromosome 12, the 12p13 locus which contains ETV6 gene, are the one of the most frequent findings in BPDCN (in 64% patients)[1][3].
  • Chromosome 6 (6q23-qter, in 50% patients) and chromosome 13 (13q13-21, in 64% patients) are also frequently involved[1][3].
  • Six major recurrent chromosomal targets were defined in one study[10]. These were 5q, 12p, 13q, 6q, 15q, and 9, which were involved in 72% (5q), 64% (12p and 13q), 50% (6q), 43% (15q), and 28% (monosomy 9) of cases, respectively.

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.
  • Common gene mutations in BPDCN: TET2, ASXL1, NRAS, ATM, and NPM1[1][3][6][14].
  • Less common mutations in BPDCN: APC, BRAF, IDH2, KIT, KRAS, MET, MLH1, RB1, RET, TP53, and VHL[3].

Other Mutations

Epigenomic Alterations

  • Multiple mutated epigenetic modifier genes have been identified in BPDCN[12][14], which include those participate in:

o  DNA methylation: TET2, IDH2

o  Chromatin accessibility: ARID1a, CHD8, SMARCA1

o  Histone modification: methylation (ASXL1, SUZ12, MLL), demethylation (KDM4D), acetylation (EP300, EP400), ubiquitination (PHC1, PHC2), dephosphorylation (EYA2) and exchange (SRCAP)[12][14]

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.

·        BCL-2 and NF-ĸB pathways[12][15]

Genetic Diagnostic Testing Methods

  • A dual-color TCF4/CD123 immunohistochemistry stain has been reported to have both high sensitivity and specificity for the diagnosis[16].
  • Immunophenotyping showing expression of pDC antigens and no lineage specific markers is relatively specific for this entity.
  • Chromosome analysis and/or copy number assessment by whole genome microarray or other technology can identify recurrent aberrations.

Familial Forms

Additional Information

Links

Put your links here

References

(use the "Cite" icon at the top of the page) (Instructions: Add each reference into the text above by clicking on where you want to insert the reference, selecting the “Cite” icon at the top of the page, and using the “Automatic” tab option to search such as by PMID to select the reference to insert. The reference list in this section will be automatically generated and sorted. If a PMID is not available, such as for a book, please use the “Cite” icon, select “Manual” and then “Basic Form”, and include the entire reference.)

  1. 1.00 1.01 1.02 1.03 1.04 1.05 1.06 1.07 1.08 1.09 1.10 1.11 1.12 1.13 1.14 1.15 1.16 1.17 1.18 1.19 1.20 1.21 1.22 1.23 Fachetti F, et al., (2017). Blastic plasmacytoid dendritic cell neoplasm, 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, p174-177.
  2. 2.0 2.1 2.2 2.3 2.4 Sapienza, Maria Rosaria; et al. (2019). "Blastic Plasmacytoid Dendritic Cell Neoplasm: State of the Art and Prospects". Cancers. 11 (5). doi:10.3390/cancers11050595. ISSN 2072-6694. PMC 6562663. PMID 31035408.
  3. 3.00 3.01 3.02 3.03 3.04 3.05 3.06 3.07 3.08 3.09 3.10 3.11 3.12 Khoury, Joseph D. (2018). "Blastic Plasmacytoid Dendritic Cell Neoplasm". Current Hematologic Malignancy Reports. 13 (6): 477–483. doi:10.1007/s11899-018-0489-z. ISSN 1558-822X. PMID 30350260.
  4. 4.0 4.1 4.2 4.3 4.4 Kerr, Daniel; et al. (2019). "Blastic Plasmacytoid Dendritic Cell Neoplasm". Current Treatment Options in Oncology. 20 (1): 9. doi:10.1007/s11864-019-0605-x. ISSN 1534-6277. PMID 30715612.
  5. 5.0 5.1 5.2 Kubota, Sho; et al. (2019). "Lineage-specific RUNX2 super-enhancer activates MYC and promotes the development of blastic plasmacytoid dendritic cell neoplasm". Nature Communications. 10 (1): 1653. doi:10.1038/s41467-019-09710-z. ISSN 2041-1723. PMC 6458132. PMID 30971697.
  6. 6.0 6.1 6.2 6.3 6.4 Sumarriva Lezama, Lhara; et al. (2018). "An analysis of blastic plasmacytoid dendritic cell neoplasm with translocations involving the MYC locus identifies t(6;8)(p21;q24) as a recurrent cytogenetic abnormality". Histopathology. 73 (5): 767–776. doi:10.1111/his.13668. ISSN 1365-2559. PMID 29884995.
  7. 7.0 7.1 7.2 Nakamura, Y.; et al. (2015). "Identification of SUPT3H as a novel 8q24/MYC partner in blastic plasmacytoid dendritic cell neoplasm with t(6;8)(p21;q24) translocation". Blood Cancer Journal. 5: e301. doi:10.1038/bcj.2015.26. ISSN 2044-5385. PMC 4450326. PMID 25860292.
  8. 8.0 8.1 Sakamoto, Kana; et al. (2018). "Recurrent 8q24 rearrangement in blastic plasmacytoid dendritic cell neoplasm: association with immunoblastoid cytomorphology, MYC expression, and drug response". Leukemia. 32 (12): 2590–2603. doi:10.1038/s41375-018-0154-5. ISSN 1476-5551. PMID 29795241.
  9. 9.0 9.1 Boddu, Prajwal C.; et al. (2018). "8q24/MYC rearrangement is a recurrent cytogenetic abnormality in blastic plasmacytoid dendritic cell neoplasms". Leukemia Research. 66: 73–78. doi:10.1016/j.leukres.2018.01.013. ISSN 1873-5835. PMID 29407586.
  10. 10.0 10.1 Leroux, Dominique; et al. (2002). "CD4(+), CD56(+) DC2 acute leukemia is characterized by recurrent clonal chromosomal changes affecting 6 major targets: a study of 21 cases by the Groupe Français de Cytogénétique Hématologique". Blood. 99 (11): 4154–4159. doi:10.1182/blood.v99.11.4154. ISSN 0006-4971. PMID 12010820.
  11. 11.0 11.1 Pemmaraju, Naveen; et al. (2019). "Tagraxofusp in Blastic Plasmacytoid Dendritic-Cell Neoplasm". The New England Journal of Medicine. 380 (17): 1628–1637. doi:10.1056/NEJMoa1815105. ISSN 1533-4406. PMID 31018069.
  12. 12.0 12.1 12.2 12.3 Lezama, Lhara; et al. (2019). "Expounding on the essence of epigenetic and genetic abnormalities in blastic plasmacytoid dendritic cell neoplasms". Haematologica. 104 (4): 642–643. doi:10.3324/haematol.2018.211557. ISSN 1592-8721. PMC 6442968. PMID 30930334.
  13. Tang, Zhenya; et al. (2018). "Genomic aberrations involving 12p/ETV6 are highly prevalent in blastic plasmacytoid dendritic cell neoplasms and might represent early clonal events". Leukemia Research. 73: 86–94. doi:10.1016/j.leukres.2018.09.006. ISSN 1873-5835. PMID 30248580.
  14. 14.0 14.1 14.2 Sapienza, Maria Rosaria; et al. (2019). "Blastic plasmacytoid dendritic cell neoplasm: genomics mark epigenetic dysregulation as a primary therapeutic target". Haematologica. 104 (4): 729–737. doi:10.3324/haematol.2018.202093. ISSN 1592-8721. PMC 6442957. PMID 30381297.
  15. Chang, Kung-Chao; et al. (2019). "Blastic plasmacytoid dendritic cell neoplasm with immunoblastoid morphology and MYC rearrangement and overexpression". Pathology. 51 (1): 100–102. doi:10.1016/j.pathol.2018.09.058. ISSN 1465-3931. PMID 30482401.
  16. Sukswai, Narittee; et al. (2019). "Dual Expression of TCF4 and CD123 Is Highly Sensitive and Specific For Blastic Plasmacytoid Dendritic Cell Neoplasm". The American Journal of Surgical Pathology. 43 (10): 1429–1437. doi:10.1097/PAS.0000000000001316. ISSN 1532-0979. PMID 31261288.

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: “Blastic plasmacytoid dendritic cell neoplasm”. Compendium of Cancer Genome Aberrations (CCGA), Cancer Genomics Consortium (CGC), updated 12/13/2023, https://ccga.io/index.php/HAEM5:Blastic_plasmacytoid_dendritic_cell_neoplasm.