Twenty-one cases of blastic plasmacytoid dendritic cell neoplasm: focus on biallelic locus 9p21.3 deletion

Blood ◽  
2011 ◽  
Vol 118 (17) ◽  
pp. 4591-4594 ◽  
Author(s):  
Marco Lucioni ◽  
Francesca Novara ◽  
Giacomo Fiandrino ◽  
Roberta Riboni ◽  
Daniele Fanoni ◽  
...  

Abstract Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare and aggressive malignancy derived from precursors of plasmacytoid dendritic cells. We analyzed 21 cases with array-based comparative genomic hybridization (aCGH). Complete or partial chromosomal losses largely outnumbered the gains, with common deleted regions involving 9p21.3 (CDKN2A/CDKN2B), 13q13.1-q14.3 (RB1), 12p13.2-p13.1 (CDKN1B), 13q11-q12 (LATS2), and 7p12.2 (IKZF1) regions. CDKN2A/CDKN2B deletion was confirmed by FISH. This scenario argues for disruption of cell cycle at G1/S transition, representing a genetic landmark of BPDCN, and possibly contributing to its pathogenesis. Statistical analysis of overall survival in our series highlighted an association of poor outcome with biallelic loss of locus 9p21.3. We suggest that, in the absence of reliable parameters for predicting prognosis in BPDCN other than age, tumor stage, and/or clinical presentation, simple methods, such as FISH for CDKN2A/CDKN2B, could help to identify the most aggressive cases.

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 456-456 ◽  
Author(s):  
Eve Poret ◽  
Chrystelle Vidal ◽  
Yohan Desbrosses ◽  
Fanny Angelot Delettre ◽  
Maider Pagadoy ◽  
...  

Abstract Blastic plasmacytoid dendritic cell neoplasm is a rare and aggressive neoplasm for which there is still no current consensus on the best therapeutic approach. Most patients respond to intensive chemotherapy, but relapses are almost inevitable with median overall survival (OS) in the largest patient series ranging from 8 to 12 months except for patients who could benefit from allogenic hematopoietic stem cell transplantation (allo-HSCT). We present results of the first line treatments used in France between 2000 and 2013 for 86 patients recruited in the French network of BPDCN (abstract ASH 2015 N°78460). Seventeen patients were treated with acute lymphoid leukemia (ALL)-like therapy (median age : 63 yo) , 19 with acute myeloid leukemia (AML)-like therapy (median age : 40 yo), 16 patients with CHOP-like therapy (median age : 72 yo), 16 patients with NK/T-like therapy (based on high-dose methotrexate and L-asparaginase, ± dexamethasone, median age: 59 yo), and 12 patients received "other treatments" (OT, means variable drugs, median age : 82 yo). Thirty four patients obtained a complete remission (CR) and received HSCT (autologous n=4, or allogeneic n=30). The response rates for CHOP-like and OT groups were 31.3% and 25.0% respectively. For ALL-like, AML-like, and NK/T-like groups, response rates reached 70.6%, 78.9%, and 62.5% respectively (no statistic difference). Relapse rates among responders for CHOP-like and OT groups were 60% and 33.3% whereas there were only 25%, 26.7%, and 20% in ALL-like, AML-like, and NK/T-like groups respectively. For patients who obtained remission, the median of remission duration was 8.0 and 14.0 months for patients who received CHOP-like treatments (n=5) and OT (n=3) respectively and 10.0, 10.0, and 9.0 months for ALL-like (n=11), AML-like (n=14), and NK/T-like groups (n=9) respectively (p = 0.6339). In preclinical studies, we have shown that BPDCN cells are sensitive in vitro to idarubicine (Angelot Delettre F et al, 2015) so we studied patients receiving idarubicine in first line therapy in our series (n=9). From these 9 patients, 7 obtained CR and only one relapsed after 10 months. The 6 patients in continuous CR without any relapse have received HSCT (allo, n=5 or auto, n=1). Two out of those 6 patients are alive at the time of data collection with a follow-up of 40 and 87 months; the other 4 patients died after the graft, one relapsed after auto-HSCT, and 3 died of infectious complications after allo-HSCT. The median OS for patients who received HSCT, auto or allo (n=34) and other patients (n = 52) is respectively 49 and 8 months (p < 0.0001, Figure 1). The beneficial effect of HSCT persists independently of age in multivariate analysis. These results suggest that NK/T-like, AML-like, and ALL-like groups give better results than CHOP-like and OT groups. However, there is no significant statistical difference between AML-like, ALL-like, and NK/T-like groups. Thus it seems to be wise to combine "lymphoid" drugs like methotrexate, L-asparaginase and dexamethasone with "myeloid" drug such as idarubicine. The importance of allogenic stem cell transplantation to sustain remission is clear in this study and other one (Roos-Weil et al, 2013). We also observed a prolonged CR in one patient after auto-HSCT. Based on our results, we will propose the first prospective, multicentric, phase II trial in BPDCN, testing a combination of 3 cycles of methotrexate, L-asparaginase, idarubicine and dexamethasone followed by an allo-HSCT in first clinical remission for all eligible patients or repeated cycle of these drugs for unfit patients with auto-HSCT if possible. Kaplan-Meier overall survival curves compared by the Log-Rank test in the cohort of 34 HSCT patients (auto and allo, blue line) and 52 non HSCT patients (red line) (p<0.0001). Censured patients are patient's alive or lost (+). OS of HSCT patients is still statistically significative with adjustment of age in multivariate analysis (Cox multivariate). Figure 1. Overall survival of HSCT patients and non HSCT patients. Figure 1. Overall survival of HSCT patients and non HSCT patients. Disclosures Recher: Celgene; Amgen; Chugai: Research Funding; Janssen; Novartis; Amgen: Other: Travel, accommodations, expenses; Sunesis; Celgene: Consultancy. Deconinck:CHUGAI: Other: Travel for international congress; NOVARTIS: Other: Travel for international congress; ALEXION: Other: Travel for international congress; LFB loboratory: Consultancy; JANSSEN: Other: Travel for international congress; PFIZER: Research Funding; ROCHE: Research Funding.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4703-4703
Author(s):  
Paola Carluccio ◽  
Mario Delia ◽  
Anna Mestice ◽  
Domenico Pastore ◽  
Alessandra Ricco ◽  
...  

Abstract Abstract 4703 The World Health Organization (WHO) recently published a revised, updated edition of the WHO Classification of Tumours of the Haematopoietic and Lymphoid Tissues, including new criteria for the recognition of some previously described neoplasms as well as clarification and refinement of the defining criteria for others. It also adds entities – some defined mainly by genetic and immunophenotypic features – that have only recently been characterized. Particularly, the diagnosis and classification of acute leukemias of ambiguous lineage is debated; among these: “blastic NK-cell lymphoma” and “agranular CD4+/CD56+ hematodermic neoplasms”. Both of them are now known to be, in virtually all cases, a tumor derived from precursors of a specialized subset of dendritic cells, plasmacytoid dendritic cells, and so are myeloid-related neoplasms defined as blastic plasmacytoid dendritic cell neoplasm (BPDCN). This is a clinically aggressive neoplasm that is usually characterized at onset by solitary or multiple skin lesions, often with associated regional lymphadenopathy, and frequently by involvement of the PB and BM. Leukemic cells show submembranous cytoplasmic vacuoles and pseudopodia-like extensions of agranular cytoplasm. The blasts in such cases do not express myeloperoxidase or nonspecific esterase, and are characterized by the expression of CD4, CD43, CD56, CD123, BDCA-2/CD303, TCL1, and CLA; CD7 and CD33 are not uncommonly expressed as well, and TdT is expressed in about 30% of cases. There is no expression of CD34 or CD117. Here we report three cases with clinical data, cytological and immunophenotypic findings strongly suggesting the diagnosis of BPDCN. Case 1 An 80 year-old-man was admitted to our institution on December 2006. He referred the occurrence of skin lesions since January 2005, when a diagnosis of extranodal B-cell non-Hodgkin lymphoma was made and treatment with conventional chemotherapy was performed, but without achieving any response. At our evaluation he presented leukocytosis (144 × 109/L) associated with purplish, firm nodules on the trunk, arms and face. Peripheral blood and bone marrow aspirate showed the presence of blast cells with a lymphoid appearance, granular periodic acid-Schiff (PAS) positivity and a high expression of CD33, CD4, and CD56. He was treated with AML-like therapy, but died of disease progression. Case 2 A 79-year old woman was admitted in December 2006 with a 2-month history of anemia, splenomegaly, and weight loss of 10 kg in the last year. Laboratory tests were as follows: Hb, 41 g/L; leukocytes, 2.5 × 109/L (with 10% of blast cells); platelets, 43 × 109/L. No lymphadenopathy or skin lesions were present. Bone marrow examination revealed 41% of small to medium-sized blast cells without Auer rods or granula and negative reactivity to myeloperoxidase, esterase and PAS. She was treated with an AML-like protocol; she achieved partial response, but died after three months, of disease progression. Case 3 A 69-year-old man was admitted to our Institution for cytopenia in June 2009. He referred the occurrence of brownish-purple firm nodules on the trunk since April 2009. At our evaluation he presented pancytopenia; bone marrow aspiration was performed and revealed infiltration by 65% of blasts with reticulated chromatin, evident nucleoli, a vacuolated cytoplasm and pseudopodia-like expansions. The blasts were negative for myeloperoxidase, monocyte esterase and PAS staining. Skin biopsy revealed a dermal infiltration by the same blastic-cell BM population. He underwent AML-like therapy and, although the skin lesions disappeared, 30% blastic bone marrow infiltration persisted. Morphological revision of these cases, selected for their peculiar immunophenotype reported in the following Table, revealed the same cytological features and cytochemical reactivity in cases 2 and 3; case 1 had a lymphoblastic-like morphology and showed PAS positivity, but the lack of cCD3 was not consistent with the diagnosis of ALL. All the cases were FLT3-ITD+. We suggest that a correct modern panel of MoAb with a careful morphological examination could help to pose the diagnosis of BPDCN, which typically affects older patients and is characterized by poor prognosis. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1486-1486
Author(s):  
Yohei Osaki ◽  
Akihiko Yokohama ◽  
Akio Saitoh ◽  
Kenichi Tahara ◽  
Kunio Yanagisawa ◽  
...  

Abstract Abstract 1486 Introduction: Dendritic cells (DCs) play critical roles in the induction and regulation of the innate and adaptive immune responses. Human blood DCs can be classified into plasmacytoid dendritic cell (pDC) and myeloid dendritic cell (mDC). In general, pDC is defined as lineage (Lin)-HLA-DR (DR)+CD123+CD11c-, and mDC is defined as Lin-DR+CD123+CD11c+. PDCs are a specific type of dendritic cells that is found in an immature form in the peripheral blood and that is the major interferon-alpha producing cell in response to viruses. Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare hematological malignancy that has a putative plasmacytoid dendritic cells origin. Unlike blood pDCs, the specific feature of BPDCN is the positive expression of CD56. In addition to these markers, BPDCNs can express various antigens, such as CD2, CD10, CD13, CD33 and even CD11c, that cause immunophenotypical diversity among cases. The goal of this study was to clarify the normal counterpart of BPDCN by analyzing the characteristics of CD56-positive blood Dendritic-like Cells (DLCs). Material and Methods: Human peripheral blood mononuclear cells (PBMNCs) were isolated by gradient centrifugation from healthy volunteers, and CD3, CD14, CD16 and CD19 antibodies were used as a lineage cocktail. We defined CD56+pDC-like cells (pDLCs) as Lin-DR+CD56+CD123+ cells, CD56+mDC-like Cells (mDLCs) as Lin-DR+CD56+CD123-CD11c+ cells, pDCs as Lin-DR+CD56-CD123+CD11c-cells and mDCs as Lin-DR+CD56-CD123+CD11c+cells. In some experiments, cells were purified from PBMNCs using a cell sorter. Sorted cells were analyzed for mRNA levels of toll-like receptors (TLRs), cytokines and transcriptional factors. Phagocytic activity and mixed lymphocyte reactions were analyzed by flow cytometry. Sorted cells were also analyzed after 4–6 days of culture with Fms-like tyrosine kinase 3 ligands (Flt3-L) and granulocyte macrophage colony-stimulating Factor (GM-CSF). Results: PBMCs comprised a small population of each cell type: 0.03% of CD56+pDLCs, 0.35% of CD56+mDLC, 0.93% of pDC 0.93%, and 0.60% of mDC. CD56+pDLCs had oval or U-shaped nuclei with condensed chromatin, and perinuclear halo, which is feature of pDC, was clearly observed in the cytoplasm. CD11c expression in CD56+pDLCs was lower than that in mDCs but higher than that in pDCs. CD56+pDLCs were not Natural Killer (NK) cells, as there was no expression of CD122 or other NK-specific antigens. Meanwhile, CD56+pDLCs had clear expression of BDCA2 and BDCA4, suggesting that this population was closely related to pDCs. Real-time quantitative (RQ) PCR assay revealed that TLRs were expressed in an intermediate level between pDCs and mDCs in CD56+pDLCs (CD56+pDLC vs. pDC vs. mDC: TLR2, 0.17 vs. 0.09 vs. 1.13; TLR4, 0.14 vs. 0.06 vs. 0.53; TLR7, 0.67 vs. 16.70 vs. 0.30; TLR9, 3.73 vs. 72.41 vs. 0.18). Expression of the transcription factors, E2-2, Irf8 and SpiB, in pDCs was higher than that in CD56+pDLCs, but lower than that in mDCs (CD56+pDLC vs. pDC vs. mDC: E2-2, 16.78 vs. 118.69 vs. 1.45; Irf8, 1.73 vs. 9.07 vs. 0.55; SpiB, 0.14 vs. 0.52 vs. 0.02). RQ−PCR after CpG stimulation revealed that CD56+pDLCs had lower interferon–alpha production when compared with pDCs (5.7405 vs. 360.881). Phagocytic capacity of CD56+pDLCs was lower than that of mDC or pDC (1.96% vs. 4.32 % vs. 52.6% for FITC-dextran positive cells in CD56+pDLCs vs. pDCs vs. mDCs). Allogeneic T cells proliferated less efficiently after culture with CD56+pDLCs than they did after culture with pDC. After in vitro culture with Flt3L and GM-CSF, the percentage of BDCA1-positive cells increased from 2.75% to 62.9%. Discussion: CD56+pDLCs were rare population in PBMNCs. Their phenotype and function were similar to pDCs, in part, but they expressed myeloid antigens and had lower function of phagocytosis and cytokine production than pDCs. In vitro culture suggested plasticity in the immunophenotype of CD56+pDLCs when compared with pDC and mDC. Collectively, these data suggest that CD56+pDLCs is a distinct new population of DCs that possesses a high degree of plasticity. These immunophenotypic characteristics and plasticity may influence the immunophenotypic diversity of BPDCNs. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2365-2365 ◽  
Author(s):  
Uday Deotare ◽  
Elizabeth Hyjek ◽  
Anna Porwit ◽  
Rumina Musani ◽  
David Barth ◽  
...  

Abstract Background: Although classified by WHO 2008 as belonging to the category “Acute myeloid leukemia and related precursor neoplasms”, Blastic Plasmacytoid Dendritic Cell Neoplasm (BPDCN) presents as an acute leukemia (AL) only in a minority of cases. There are only few studies describing the comprehensive immunophenotypic pattern of BPDCN in the bone marrow. Furthermore, given the rarity of this hematologic malignancy optimal frontline therapy is unclear. Patients and Methods: This retrospective analysis evaluates the diagnostic flow cytometry pattern and outcome of 9 patients who were diagnosed with BPDCN at the Princess Margaret Cancer Centre between December 2008 and June 2014. A four tube 10-color flow cytometry (FCM) panel has been used to correctly make the diagnosis of BPDCN in 6 patients, whereas a 5-colour panel was used in the remaining patients in conjunction with immunohistochemistry. The following markers were included in the10-color panel: Tube 1: CD65 FITC, CD13 PE, CD14 ECD, CD33 PC5.5, CD34 PC7, CD117 APC, CD7 A700, CD11b A750, CD16 PB, and CD45 KO; Tube 2: CD36 FITC, CD64 PE, CD56 ECD, CD33 PC5.5, CD34 PC7, CD123 APC, CD19 A700, CD38 A750, HLA-DR PB, and CD45 KO; Tube 3: CD71 FITC, CD11c PE, CD4 ECD, CD33 PC5.5, CD34 PC7, CD2 APC, CD10 A700, CD235a A750, CD15 PB, and CD45 KO; Tube 4:nuclear (n) TdT FITC, cytoplasmic (cyt.) MPO PE, CD14 ECD, CD33 PC5.5, CD34 PC7, cyt.CD79a APC, cyt.CD22 A700, CD19 A750, cyt.CD3 PB, and CD45 KO. Results: Median age was 66 years (range, 25 to 91 years); 3 patients were over the age of 70 years. Fifty-six percent were males. All presented with skin lesions and 78% presented each with lymphadenopathy and bone marrow involvement. Cytogenetics were poor-risk in 2 patients, intermediate-risk in 3 and unknown or inconclusive in 4. By 10-color FCM, leukemic cells were in the blast gate (CD45dim/low SSC) and were positive for CD4(bright), CD33(dim), CD56(heterogenous), CD123(bright), CD36, CD38, HLA-DR, CD71, but negative for CD10, CD11b, CD13, CD14, CD15, CD16, CD19, CD34, CD64, CD65, CD235a. Other markers, such as cyt.MPO, cyt.CD3, cyt.CD22 and nTdT were negative, while dim cyt.CD79a was seen in 3 cases. CD7 expression was found in 5 cases, whereas CD2 and CD117 were found in single cases only. BM involvement by BPDCN leukemic cells ranged from 27% to 92% of the marrow cellularity. Skin involvement showed dense infiltrate of cells with blastoid morphology and characteristic grenz zone. Seven patients received front-line induction therapy with HyperCVAD with an overall response rate of 86% (4 complete remissions (CR), 2 unconfirmed CRs). One patient died of multi-organ failure during induction. Three of 6 responders underwent planned allogeneic hematopoietic cell transplantation (HCT); 1 patient has since died of acute graft versus host disease (GVHD), whereas 2 are alive in remission with chronic GVHD, 12 and 14 months post transplant with complete donor chimerism. One transplant ineligible patient relapsed 22 months after achievement of CR1. Median follow-up of all patients was 12 months with a overall survival at 1 year of 59.3% for the entire group. Patients who underwent allogeneic HCT had overall survival at 1 year of 66.7% and for the chemotherapy group was 27.8% at 1 year.(p=0.34). Conclusion: An accurate diagnosis of BPDCN can be made by 10-colour FCM using a 4-tube acute leukemia panel. BPDCN demonstrates a characteristic pattern of antigen expression . Although front-line induction chemotherapy with HyperCVAD can yield high CR rates, allogeneic HCT should be performed in first CR for transplant eligible patients, as this appears to be required for long term durable remissions. For transplant ineligible or relapsed BPDCN patients, there is an unmet need for novel therapeutic agents. Disclosures Porwit: Beckman-Coulter: Speakers Bureau. Gupta:Novartis: Consultancy, Honoraria, Research Funding; Incyte Corporation: Consultancy, Research Funding.


Cancers ◽  
2021 ◽  
Vol 13 (23) ◽  
pp. 5888
Author(s):  
C. Cameron Yin ◽  
Naveen Pemmaraju ◽  
M. James You ◽  
Shaoying Li ◽  
Jie Xu ◽  
...  

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare, aggressive neoplasm derived from plasmacytoid dendritic cells. While advances in understanding the pathophysiology of the disease have been made, integrated systematic analyses of the spectrum of immunophenotypic and molecular alterations in real-world clinical cases remain limited. We performed mutation profiling of 50 BPDCN cases and assessed our findings in the context of disease immunophenotype, cytogenetics, and clinical characteristics. Patients included 42 men and 8 women, with a median age of 68 years (range, 14–84) at diagnosis. Forty-two (84%) patients had at least one mutation, and 23 (46%) patients had ≥3 mutations. The most common mutations involved TET2 and ASXL1, detected in 28 (56%) and 23 (46%) patients, respectively. Co-existing TET2 and ASXL1 mutations were present in 17 (34%) patients. Other recurrent mutations included ZRSR2 (16%), ETV6 (13%), DNMT3A (10%), NRAS (10%), IKZF1 (9%), SRSF2 (9%), IDH2 (8%), JAK2 (6%), KRAS (4%), NOTCH1 (4%), and TP53 (4%). We also identified mutations that have not been reported previously, including ETNK1, HNRNPK, HRAS, KDM6A, RAD21, SF3A1, and SH2B3. All patients received chemotherapy, and 20 patients additionally received stem cell transplantation. With a median follow-up of 10.5 months (range, 1–71), 21 patients achieved complete remission, 4 had persistent disease, and 24 died. Patients younger than 65 years had longer overall survival compared to those who were ≥65 years (p = 0.0022). Patients who had ≥3 mutations or mutations in the DNA methylation pathway genes had shorter overall survival (p = 0.0119 and p = 0.0126, respectively). Stem cell transplantation significantly prolonged overall survival regardless of mutation status. In conclusion, the majority of patients with BPDCN have somatic mutations involving epigenetic regulators and RNA splicing factors, in addition to ETV6 and IKZF1, which are also frequently mutated. Older age, multiple mutations, and mutations in the DNA methylation pathway are poor prognostic factors.


2015 ◽  
Vol 6 (4) ◽  
pp. 297-306 ◽  
Author(s):  
Haisheng Yu ◽  
Peng Zhang ◽  
Xiangyun Yin ◽  
Zhao Yin ◽  
Quanxing Shi ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document