You are on page 1of 10

Review

Blastic Plasmacytoid Dendritic Cell


NeoplasmeCurrent Insights
Sangeetha Venugopal,1 Selena Zhou,2 Siraj M. El Jamal,3 Andrew A. Lane,4
John Mascarenhas1
Abstract
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare clonal hematologic malignancy of plasmacytoid
dendritic cell precursors. The presentation and clinical course of BPDCN is widely heterogeneous and was most
recently categorized as a distinct clinical entity by the World Health Organization in 2016. The expanded under-
standing of the pathobiology of BPDCN has improved diagnostic accuracy and informed novel targeted therapeutic
options. The United States Food and Drug Administration-approval of tagraxofusp (SL-401) in December 2018 has
focused attention on this leukemia frequently associated with skin involvement. Herein, we aim to: (1) review etiology;
(2) summarize diagnostic criteria; and (3) discuss historic treatments and novel therapies for BPDCN.

Clinical Lymphoma, Myeloma & Leukemia, Vol. -, No. -, --- ª 2019 Elsevier Inc. All rights reserved.
Keywords: Acute leukemia, CD123, Myeloid neoplasms, SL-401, Tagraxofusp

Introduction Epidemiology
Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a BPDCN is a rare disease, with an overall incidence rate of 0.04
hematopoietic clonal neoplasm originating from plasmacytoid cases per 100,000 people.8 It is generally a disease of older males,
dendritic cell (pDC) precursors. BPDCN was first proposed as a with a median age of 53 to 68 years and a 2.0 to 3.3:1 male to
distinct entity in 1998 when Kameoka et al described 2 cases of female ratio.8-10 Recently, it was found to have a bimodal incidence
cutaneous agranular CD2-/CD4þ/CD56þ “lymphoma.”1 Since pattern, with higher incidences at younger than 20 years and older
that time, its rarity has led to a rich history of misdiagnosis and than 60 years of age.8,10,11 Ethnic predilection is not clear, with
irregular nomenclature, including CD56þ/TdTþ blastic NK cell conflicting published reports.8
tumor and CD4þ/CD56þ hematodermic neoplasm.2,3 Lucio et al
were the first to note a shared pattern of high CD123 expression
and propose pDCs as the origin of BPDCN,4 which was validated The Genesis of BPDCN
by a subsequent study that demonstrated immune function in BPDCN evolves from a progenitor pDC, and normal pDCs known
leukemic cells similar to pDCs.5 This understanding of the biology for their senescence comprise less than 0.5% of circulating mono-
and origin of this rare neoplasm led the World Health Organization nuclear cells. pDCs are commonly found in lymph nodes and tonsils.
to establish the term BPDCN in 2008 and classify it as a distinct They are rare in the thymus, bone marrow, spleen, and mucosa-
entity in 2016.6,7 associated lymphoid tissue. pDCs accumulate in lymph nodes12 on
exposure to antigenic stimuli, may it be viral infection or autoimmune
disease, such as systemic lupus erythematosus and psoriasis.13,14 In
1
Tisch Cancer Institute, Division of Hematology/Oncology response to foreign viral nucleic acids, pDCs secrete massive amounts of
2
3
Icahn School of Medicine at Mount Sinai, New York, NY type I interferons (IFNa and IFNb) and other proinflammatory cy-
Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY
4
Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical tokines (interleukin [IL]-6, IL-8, IL-12, and tumor necrosis factor)
School, Boston, MA through activation of toll-like receptor 7/9-MyD88-IRF7 pathway.
Submitted: Apr 9, 2019; Revised: May 14, 2019; Accepted: Jun 4, 2019 Coupled with their antigen presentation capacity, pDCs orchestrate
innate and adaptive immune responses, thus cementing their role as an
Address for correspondence: John Mascarenhas, MD, Director, Adult Leukemia Pro- arbitrator of innate and adaptive immune system.12 Additionally,
gram, Leader, Myeloproliferative Disorders Clinical Research Program, Associate Pro- mature pDC proliferation (MPDCP) have been associated with
fessor of Medicine, Tisch Cancer Institute, Division of Hematology/Oncology, Icahn
School of Medicine at Mount Sinai, One Gustave L. Levy P, Box 1079, New York, NY myeloid neoplasms, although data does not support MPDCP as a
10029 precursor lesion to BPDCN.15,16 Detailed evaluation failed to detect
E-mail contact: john.mascarenhas@mssm.edu
any viral markers (Epstein-Barr virus or human herpesvirus-6) in

2152-2650/$ - see frontmatter ª 2019 Elsevier Inc. All rights reserved.


https://doi.org/10.1016/j.clml.2019.06.002 Clinical Lymphoma, Myeloma & Leukemia Month 2019 -1
BPDCNeCurrent Insights
BPDCN,3 thus vetoing the role of latent viral infection in the patho- these patients progress to a leukemic phase eventually, a clonal
genesis of this disease. Elucidation of the pathogenesis of BPDCN is still evolution model was recently proposed. In this model, the initial
a work in progress, and ongoing endeavors hope to shed light on the malignant transformation of a pDC occurs in the skin followed by
precise biology of this rare hematologic neoplasm. dissemination to the bone marrow.27 Griffin et al studied the
developmental ontogeny of BPDCN through serial DNA
Molecular Pathogenesis sequencing of bone marrow and skin biopsies in 10 patients. All
Although 50% to 66% of patients with BPDCN exhibit karyotypic patients with “skin-only” disease and negative bone marrow
abnormalities, none is inherent to BPDCN.9,17 Cytogenetic abnor- assessment at diagnosis harbored known pathogenic gene variants
malities seen in patients with BPDCN predominantly involve recurrently mutated in BPDCN and related myeloid malignancies
genomic losses rather than gene-specific rearrangements; the 6 key in the bone marrow, including ASXL1, TET2, SF3B1, ZRSR2,
recurrent chromosomal abnormalities include 5q, 12p, 17p, 13q, 6q, CUX1, and EZH2, all consistent with clonal hematopoiesis. Post-
15q, and 9 (monosomy).17,18 Furthermore, genome wide array-based therapy analysis in patients with minimal to extensive marrow
comparative genomic hybridization analysis (n ¼ 21) identified involvement also showed persistent pathogenic mutations. Addi-
commonly deleted chromosome regions, namely 9p21.3 (CDKN2A/ tionally, most mutations within bone marrow cells were also
CDKN2B), 13q13.1-q14.3 (RB1), 12p13.2-p13.1 (CDKN1B), detected in paired skin biopsies, suggesting clonal evolution from a
13q11-q12 (LATS2), and 7p12.2 (IKZF1).19 It is interesting to note common pre-malignant hematopoietic precursor clone. Evidence of
that most of the deleted regions represent tumor suppressor genes clonality was further substantiated (through WES) in a patient with
involved in cell cycle regulation. Additionally, a molecular cytogenetic “skin-only” disease who showed identical ASXL1 and TET2 mu-
analytic series (n ¼ 47) identified a monoallelic deletion of NR3C1 tations in high variant allele frequency in the skin and bone marrow
(5q31), encoding the glucocorticoid receptor (a ligand-dependent at time of diagnosis. Interestingly, WES demonstrated that 87% of
transcription factor of the nuclear hormone receptor family tumor all somatic single nucleotide variants (SNVs) were skin-specific and
suppressor) in 13 (28%) of 47 patients with BPDCN. Subsequent 11% of SNVs were bone marrow-specific, with only 2% common
elaborate analysis of the t(3;5)(q21;q31) region unveiled the fusion of SNVs, thus proposing a model of branching pre-malignant clonal
NR3C1 to a long noncoding RNA gene (lincRNA-3q) that encodes a evolution in the bone marrow, with a sub-clone seeding the skin
novel, nuclear, noncoding RNA involved in G1/S cell cycle transition and then acquiring additional mutations during neoplastic
via E2F.20 Moreover, MYC rearrangements on 8q24 with resultant evolution.27
MYC protein overexpression was reported in 38% of BPDCN cases.
Given that MYC targets the transcription factor E2F1, it is suggested Prognosis
that MYC overexpression may potentially reinforce G1/S progres- BPDCN is a rare and aggressive hematopoietic neoplasm with a
sion.21,22 Taken together, loss of multiple cell cycle checkpoints median survival of less than 2 years (95% confidence interval [CI],
leading to altered G1/S transition appears to be substantial in the 17-34 months).28 A population-based analysis from the Surveil-
molecular pathogenesis of BPDCN. lance, Epidemiology, and End Results database identified that
Apart from the loss of tumor suppressor genes owing to various young age (age less than 60 years) and early hematopoietic stem cell
chromosomal deletions, patients with BPDCN share a mutational transplantation (HSCT) were predictive of superior outcomes.28
profile comparable to other myeloid malignancies, including myelo- Several small studies have also shown that the presence of isolated
dysplastic syndrome. Stenzinger et al used targeted deep sequencing skin involvement and a high proliferative index are associated with
(n ¼ 33 BPDCNs) to delineate recurrent mutations in 4 genes, NRAS, better outcomes.17,19 Moreover, TdT expression appears to corre-
IDH2, APC, and ATM, that implicates the predominance of activated late with longer survival, suggesting that differentiation state may
RAS signaling in BPDCN.23 Menezes et al employed a combined correlate with sensitivity to therapy.29 Furthermore, biallelic loss of
approach of whole exome sequencing (WES) in 3 cases and targeted the 9p21.3 locus and NR3C1 haploinsufficiency was associated with
resequencing (n ¼ 25 BPDCNs) to elucidate mutations involving poor outcome, as were mutations involving the DNA methylation
DNA methylation (TET2),24 transcriptional regulation (ASXL1), and machinery.19,25 Prognostication using clinical and molecular data in
transcription factors (IKZF1-3, ZEB2). NPM1 and FLT3 ITD muta- BPDCN remains a challenge owing to its rarity and the lack of a
tions, although common in acute myeloid leukemia (AML), were characteristic clinical/molecular profile.
infrequently identified in this series. Furthermore, it was observed that
the mutations involved in aberrant methylation patterns were associ- Diagnosis
ated with poor overall survival (OS) (median, 11 months vs. 79 months; Clinical Presentation
P ¼ .047).25 Additionally, a gene expression analysis identified high BDPCN is an aggressive hematologic malignancy typified by its
expression of BCL2 in neoplastic pDCs compared with normal proclivity for cutaneous tropism and/or a leukemic phase30,31
pDCs.26 Collectively, the shared genetic profile between BPDCN and (Figure 1). Ninety percent of patients initially present with cuta-
related myeloid malignancies resolutely endorse a genetic link between neous lesions (Figure 2A and 2B) and 10% with overt acute leu-
the 2 entities and the potential for comparable therapeutic options. kemia.34 The cutaneous presentation of BPDCN is heterogeneous,
ranging between maculo-nodular lesions and dystropic xanthoma-
Proposed Clonal Evolution Model of tosis.10,35,36 However, bruise-like patches in the upper body appear
BPDCN to be the most common presenting sign in BPDCN limited to
Given that most patients present with skin-only disease without skin.37 Bone marrow involvement resulting in cytopenias (particu-
conspicuous bone marrow involvement and that the majority of larly thrombocytopenia, 78% of patients), hepatosplenomegaly, and

2 - Clinical Lymphoma, Myeloma & Leukemia Month 2019


Sangeetha Venugopal et al
Figure 1 Patterns of BPDCN Involvement

Abbreviation: BPDCN ¼ blastic plasmacytoid dendritic cell neoplasm.


(Adapted From10,11,32,33)

lymphadenopathy were also noted in varying frequencies.38,39


Figure 2 A, Photograph Showing a Central Violaceous Plaque Given that central nervous system (CNS) involvement occurs in
Surrounded by Numerous Erythematous Papules on up to 30% of cases, flow cytometry evaluation of cerebrospinal fluid
the Shoulder. B, Violaceous Tumor With Surrounding
is suggested in newly suspected cases of BPDCN.9,40 Other re-
Petechiae on the Lower Back
ported sites of involvement include lung, breast, tongue, gall-
bladder, and the paranasal sinuses.9,35,41,42 Given the heterogeneity
of affected organs, computed tomography or positron emission to-
mography should be included in the initial evaluation of a patient
with suspected BPDCN.
A diagnosis of BPDCN is established by histopathologic assess-
ment of the involved tissue coupled with germane immunopheno-
typic markers (Figure 3).

Histopathology
BPDCN lesions demonstrate similar microscopic morphology
across all sites of involvement, may it be skin, lymph nodes, or bone
marrow. The neoplastic cells form dense monomorphous atypical
blastic cells resembling myeloid sarcoma. The neoplastic cells are
medium- to large-sized with scant cytoplasm, prominent single or
multiple nucleoli with irregular nuclear contours, and immature,
fine chromatin. Mitotic figures are common.2,31 The skin lesions in
BPDCN exhibit a nodular-diffuse growth pattern in the dermis,
with a predilection for perivascular/periadnexal distribution and
intra-tumoral hemorrhage. Additionally, there is a well-demarcated
dermal grenz zone, a narrow uninvolved zone of papillary dermis
between the epidermis and the underlying dendritic neoplastic cell
infiltrate.32 The epidermis and adnexa are usually spared. Further-
more, the evident absence of coagulation necrosis and angioinvasion
represent a distinctive diagnostic feature of BPDCN.30 Hematologic
involvement is remarkable for cytopenias owing to bone marrow
infiltration by the neoplastic dendritic cells.2,24,38 In aspirate smears,

Clinical Lymphoma, Myeloma & Leukemia Month 2019 -3


BPDCNeCurrent Insights
Figure 3 Skin Biopsy Shows Blastic Plasmacytoid Dendritic Cell Neoplasm. Atypical Immature Blastic Cell Infiltration Extends Deep in
the Dermis (A; 3100). The Cells are Medium-Sized With Irregular Contours, Prominent Nucleoli, and Fine Chromatic. Mitotic
figures are Readily Found (B; 3400). The Cells are Positive for CD4, CD56, and CD123; and Negative for Other Lineage
Specific Markers Like CD3, CD19, and MPO

these cells display a distinctive cytoplasmic vacuolation pattern akin specific to BPDCN,46 as it is also expressed in a wide spectrum of B
to a “pearl necklace” and cytoplasmic pseudopod extensions.10,32,38 and T cell lymphomas and leukemias.54 BPDCN is also positive for
granzyme B, but not perforin and TIA-1. Additionally, BCL11A
Immunophenotyping and SPIB are transcription factors involved in pDC develop-
Immunophenotyping is indispensable in establishing the diag- ment49,55; however, their expression in the case of BPDCN needs to
nosis of BPDCN. The immunophenotype of BPDCN resembles be interpreted in conjunction with other phenotypic markers.
that of pDC subsets; however, with atypical CD56 expression. Terminal deoxynucleotidyl transferase (TdT) and CD68 expression
BPDCN is characterized by the expression of CD4 and CD56 in are variably expressed in 40-50% of cases.32,44 Neither CD34
the absence of lineage-specific markers for either myeloid, T-lym- expression, nor Epstein-Barr virus-encoded RNA is observed in
phocytes, B-lymphocytes, or NK cells. CD5, CD7, CD68, and BPDCN. Overall, there is a distinct lack of unique BPDCN
CD33 may be expressed in some cases.3,10,32,43,44 The expression of markers, and there is no consensus on the minimal phenotype to
CD56 in BPDCN, but not in normal pDC, is an important feature establish an immunophenotypic diagnosis of BPDCN. It has been
and likely indicates the neoplastic transformation of these cells. proposed that BPDCN may be confidently diagnosed when the
BDCA-2/CD303, IL-3Ra/CD123, CD2AP, TCL1, BCL11A, and blastic dendritic cells pDCs express 4 of the 5 principal markers
SPIB are pDC-associated markers that may aid in the diagnosis of (CD4, CD56, CD123, TCL1, and CD303).10,24,50 Cases with
BPDCN.3,10,11,45-50 Although the interleukin-3 receptor alpha immunophenotype that is short of the 4 antigens should be classi-
chain (IL-3Ra) CD123 is expressed in the majority of BPDCN, it is fied as “acute leukemia of ambiguous lineage.”56
not specific as it is seen in other hematologic malignancies including
AML.3,10,31,51 CD303 (also known as blood dendritic cell antigen 2 BPDCN Mimics
- BDCA2), a pDC-specific type II C-type lectin receptor involved in Given BPDCN’s tendency for cutaneous predilection, typical skin
antigen capture and presentation,50 considered the most specific lesions should primarily alert the clinician to the possibility of
marker for plasmacytoid dendritic cells, may be aberrantly lost in BPDCN. However, given that 10% of patients initially present with
BPDCN, thus limiting its sensitivity.52 The CD2-associated pro- aggressive leukemia sans skin involvement,34 the diagnosis of
tein, an adaptor protein involved in T-cell signaling, is present in BPDCN should be entertained in any patient presenting with poorly
normal and neoplastic pDCs.48,53 Its expression in BPDCN is differentiated leukemia with an ambivalent immunophenotype.
heterogeneous and is also expressed in diffuse large B cell lym- MPDCP commonly occurs in the setting of other myeloid neo-
phomas.53 The T-cell leukemia/lymphoma protein 1 (TCL1), plasms like chronic myelomonocytic leukemia, AML, and myelo-
encoded by the proto-oncogene TCL1, is also present but not dysplastic syndrome,57,58 demonstrating mature pDC morphology

4 - Clinical Lymphoma, Myeloma & Leukemia Month 2019


Sangeetha Venugopal et al
and classical myeloid immunophenotype in the absence of CD56 Tagraxofusp
expression. Appreciable success of targeted therapies in related hematopoietic
Myeloid sarcoma (MS), another condition that can be indistin- neoplasms and the lack of effective therapeutic response with con-
guishable from BPDCN based on morphology, may show a CD4- ventional chemotherapeutic regimens in BPDCN provided the
positive/CD56-positive immunophenotype. Additionally, a large impetus to identify an appropriate actionable target. IL3R-a
subset of MS is CD34-negative/MPO-negative, an imminopheno- (CD123) was deemed a potential actionable therapeutic target,
type that is shared with BPDCN. However, the expression of given its ubiquitous expression in BPDCN and the neoplastic
lysozyme, CD68, or CD163 is more consistent with MS. Sangle dendritic blasts requirement of IL-3 supplementation for growth
et al identified a 7-antibody panel (CD4, CD56, CD123, TCL1, and survival.5,61,62
myxovirus A (MxA), lysozyme, and myeloperoxidase) with a pre- Tagraxofusp (SL-401) is a CD123 directed recombinant fusion
dictive value of  90% and employed these markers using specific protein composed of IL-3 conjugated to modified diphtheria toxin
staining criteria to reasonably distinguish between MS and via amide linkage. Binding of the IL-3 domain of SL-401 to its
BPDCN. In summary, BPDCN was associated with positive natural receptor with subsequent internalization leads to trans-
staining for CD56, TdT, or TCL1 or negative staining for lyso- location of the diphtheria toxin fragment to the cytosol, followed by
zyme. MS was associated with positive staining for lysozyme or ADP ribosylation of elongation factor 2, with resultant cessation of
myeloperoxidase, or negative staining for CD56, CD123, myxo- protein synthesis and cell death.63 Preclinical studies demonstrated
virus, or TCL1.59 Myeloid cell nuclear differentiation antigen, the exquisite cytotoxicity of SL-401 in cell lines derived from pa-
which is expressed in the majority of MS but not BPDCN, is also tients with BPDCN (CAL-1 and GEN2.2) and primary BPDCN
useful and may be included in the diagnostic evaluation of blastic cells with femtomolar IC50 values. Additionally, SL-401 meaning-
hematopoietic infiltrates.60 fully expanded the median OS of a BPDCN xenograft model
Given the cutaneous tropism of BPDCN, it should be differen- following a single cycle of treatment (53 days vs. 15 days in un-
tiated from other CD56þ hematopoietic neoplasms with skin treated mice).64
involvement. The potential diagnostic pitfalls include CD56þ Based on the encouraging preclinical enabling studies, a phase I/
AML, extranodal NK/T cell lymphoma, and classic cutaneous T cell II study of SL-401 was conducted in 11 patients, including those
lymphoma.44 These distinct entities must be carefully distinguished with recurrent/refractory BPDCN or who were ineligible for stan-
utilizing morphology and immunohistochemistry, given the signif- dard chemotherapy. Seven (78%; n ¼ 11) of 9 evaluable patients
icant prognostic and therapeutic implications (Table 1). Ultimately, attained a major clinical response (complete response [CR], 5;
the expression of CD303 essentially confirms the pDC origin and partial response [PR], 2) after a single course of SL-401. The me-
excludes any non-pDC hematopoietic lesions.50 dian response duration was 5 months (range, 1-20þ months). Only
5 of 11 patients experienced infusion related fever and chills, all of
Management which resolved with supportive management. Almost all patients
BPDCN is an aggressive disease with a median OS of less than a had  1 adverse event, including hypoalbuminemia, edema,
year if left untreated.7 Given the elusive nature of the disease, there hyponatremia, hypocalcemia, uremia evocative of capillary leak
has been no standard of care until the most recent United States syndrome, although the severity was mitigated by early adminis-
Food and Drug Administration-approved targeted therapy tagrax- tration of parenteral albumin and diuretics. Generally, SL-401 was
ofusp (SL-401). When tagraxofusp is not an option, acute well-tolerated with no treatment related mortality at the recom-
lymphoblastic leukemia (ALL)-inspired intensive chemotherapy mended phase 2 dose (RP2D) of 12.5 mg/kg/day.62 The subsequent
regimen with CNS prophylaxis is an acceptable alternative to induce multicenter, multicohort open label, non-randomized, single-arm
remission, followed by HSCT for consolidation and potential cure. phase II clinical trial evaluated efficacy of tagraxofusp in patients
Low-intensity therapies may be a viable option in patients ineligible with treatment-naive or relapsed/refractory (R/R) BPDCN. The
for more intensive chemotherapy or HSCT (Figure 4). trial consisted of 3 stages: stage 1 (lead-in, dose escalation), stage 2

Table 1 Differential Diagnosis of CD56D Hematopoietic Neoplasms With Skin Involvement by Immunohistochemistry

Myelomonoytic
Markers (CD13, T Cell Markers
CD15, Lysozyme, Markers of EBV (CD3, CD2, CD5,
Disease CD4 CD56 CD123 CD117, or MPO) Infection (EBNA-1) CD7, CD8)
BPDCN þ þ þ   
AML/myeloid sarcoma þ/ þ þ/ þ  
Extra nodal NK/T cell  þ   þ þ
lymphoma
Classic primary  þ    þ
cutaneous T cell
lymphoma

Abbreviations: AML ¼ acute myeloid leukemia; BPDCN ¼ blastic plasmacytoid dendritic cell neoplasm; EBNA ¼ Epstein Barr nuclear antigen; EBV ¼ Epstein Barr virus; MPO ¼ myeloperoxidase.
Adapted from reference.44

Clinical Lymphoma, Myeloma & Leukemia Month 2019 -5


BPDCNeCurrent Insights
Figure 4 Proposed Treatment Algorithm for BPDCN

Abbreviations: ALL ¼ acute lymphocytic leukemia; AML ¼ acute myeloid leukemia; BPDCN ¼ blastic plasmacytoid dendritic cell neoplasm.

(expansion), and stage 3 (pivotal, confirmatory). Stage 1 and 2 AML-based regimens. One study of 15 patients treated with ALL-
enrolled patients who were treatment-naive and patients with R/R based regimens and 26 patients treated with AML-based regimens
BPDCN, whereas the stage 3 pivotal cohort enrolled only found significant improvement in both CR rates (66% vs. 26%;
treatment-naive patients. Patients treated within the stage 2 and 3 P ¼ .02) and OS benefit (12.3 vs. 7.1 months; P ¼ .02) with the
cohorts received the stage 1 recommended tagraxofusp dose of 12 ALL-based therapy group. However, 35% of treated patients who
mg/kg/day intravenously administered daily on days 1 to 5 of a 21- achieved complete remission subsequently relapsed, more frequently
day cycle. Seven of 13 patients in the treatment-naive stage 3 after ALL-type chemotherapy, at a median time of 9.1 months
BPDCN cohort achieved a sustained CR/clinical complete response (range, 5.8-19.8 months) after diagnosis.9 This was further
(CRc) (53.8%; 95% CI, 25.1%-80.8%). After a median follow-up corroborated by a study of 46 patients that found ALL-based reg-
of 11.5 months, the median response duration was not reached. imens and HSCT to have better outcomes than AML- and
Among 29 treatment-naive patients across all stages, the overall lymphoma-based regimens.67 Regardless of the chosen induction
response rate (ORR) was 90% (26/29) with a 72% (21/29) rate of chemotherapy, intrathecal therapy should be included as BPDCN
CR þ CRc þ CRi (CRi ¼ CR with incomplete hematologic re- has a predilection for involvement of the CNS, and intrathecal
covery), and 45% (13/29) of these patients successfully bridged to prophylaxis has been shown to reduce incidence of CNS disease and
HSCT (10 allogeneic þ 3 autologous). The median OS was not improve OS.40 Tagraxofusp clinical trials have not reported CNS
reached among 29 treatment-naive patients with a median follow- relapses, suggesting that novel agents may have improved activity in
up of 23.0 months (range, 0.2-41þ months). Capillary leak syn- the CNS. However, those trials excluded patients with active CNS
drome was the most serious but manageable treatment emergent disease at the time of enrollment, which indicates that additional
adverse event noted.65 This study led to the United States Food and studies focused on treatment of BPDCN with CNS involvement
Drug Administration approval of tagraxofusp on December 21, may be required to define optimal clinical practice in this setting.
2018 for the treatment of both treatment-naive and previously A meta-analysis of 97 patients with a purported diagnosis of
treated BPDCN in adults and pediatric patients 2 years and older.66 BPDCN evaluated the impact of different therapeutic approaches
on outcome. Patients were divided into groups based on the in-
Induction Chemotherapy tensity of therapy: A, less aggressive than CHOP; B, moderately
The archival therapies for BPDCN include aggressive non- intensive CHOP or CHOP-like regimens; C, intensive leukemia-
Hodgkin lymphoma regimens such as CHOP (cyclophosphamide, inspired regimens (lymphoblastic and myeloblastic); and D, mye-
doxorubicin, vincristine, and prednisone) or CHOP-inspired, ALL loablative therapy followed by stem cell rescue (n ¼ 10; autologous
regimens such as hyper-CVAD (hyperfractionated cyclophospha- HSCT, 4; allogeneic HSCT, 6). Myeloablative conditioning
mide, vincristine, doxorubicin, and dexamethasone) alternating with regimen included total body irradiation and high-dose cyclophos-
methotrexate and cytarabine, or AML induction regimens (eg, phamide in this cohort. Moderately intensive CHOP or CHOP-like
MICE [mitoxantrone, idarubicin, cytarabine, and etoposide]; regimens did not result in better survival rates or sustained CR when
cytarabine and an anthracycline [7 þ 3]; or FLAG-IDA [fludar- compared with less aggressive therapies. However, leukemia-
abine, cytarabine, granulocyte colony stimulating factor, and inspired regimens demonstrated a CR rate of 94% with approxi-
idarubicin]). mately 40% sustained CR. Although the entire group D cohort
Of the aggressive leukemia-inspired regimens for the treatment of enjoyed a median survival of 31.5 months, allogeneic HSCT re-
BPDCN, ALL-based regimens appear to be more successful than cipients had a survival benefit over autologous HSCT recipients

6 - Clinical Lymphoma, Myeloma & Leukemia Month 2019


Sangeetha Venugopal et al
(38.5 vs. 16.5 months). Taken together, aggressive therapies appear shown that myeloablative conditioning resulted in lower relapse
to be superior to less aggressive treatment modalities in maintaining rates when compared with reduced intensity conditioning (18% vs.
CR (group A, 7%; group B, 3%; group C, 35%; group D, 50%) 40%), suggesting a possible benefit linked to the intensity of con-
along with survival benefit.68 ditioning regimens in BPDCN. Given the evolving long-term
outcome data of tagraxofusp, allogenic HSCT in CR1 is currently
HSCT regarded the appropriate post-remission consolidation strategy, and
HSCT for BPDCN is associated with improved OS and disease- CR at the time of HSCT is predictive of better outcome.71
free survival (DFS) compared with chemotherapy alone.9,67,68 Aoki
et al evaluated the clinical outcomes after transplantation in patients Low-intensity Regimens
with BPDCN from a Japanese transplant registry (allogeneic Low-intensity treatment regimens may be a reasonable alternative
HSCT, n ¼ 14; autologous HSCT, n ¼ 11). All 11 patients who in patients ineligible for a more intensive chemotherapy or HSCT
underwent autologous HSCT were in CR1. Among patients who approach. However, the data is primarily derived from small
underwent allogenic HSCT, 12 were in CR1, 2 were in CR2, and 2 retrospective studies, and robust prospective data is lacking. Several
were not in remission at time of HSCT. At a median follow-up of case reports have demonstrated the limited efficacy of azacitidine,
53.5 months, patients who underwent autologous HSCT in CR1 gemcitabine with docetaxel, pralatrexate, and bendamustine in this
enjoyed better OS and PFS than those that received an allogeneic population (Table 2).
HSCT in CR1 (OS, 82% vs. 69%; P ¼ .44 and PFS, 73% vs. 60%; The most compelling data is with gemcitabine in combination
P ¼ .43, respectively).69 However, a North American collaborative with docetaxel. A case series examined the outcomes of 3 patients
multicenter study demonstrated lack of efficacy with autologous with BPDCN who were heavily pre-treated (median, 4.6 prior
HSCT, but this study was limited by a small sample size to evaluate therapies), 2 of whom had relapsed after HSCT. The median OS
for autologous transplant (allogeneic HSCT ¼ 37 and autologous was 13.3 months with combination gemcitabine therapy, with 1
HSCT ¼ 8). Additionally, only 5 (63%) of 8 patients were in CR1, patient alive at the time of publication (the other 2 experienced
and the median age of the patient cohort was 10 years older than the progression of disease). All 3 patients attained a skin CR, and both
Japanese study.70 Taken together, autologous HSCT may have a patients with bone marrow involvement attained bone marrow
potential role in the management of patients with BPDCN in CR1, CR.73 A case report of gemcitabine monotherapy in a patient with
especially in those deemed ineligible for allogenic HSCT. BPDCN with skin involvement attained a partial response, with a
A recent meta-analysis of allogeneic HSCT in 128 patients with decreased number and intensity of skin lesions.72
BPDCN reported a pooled OS and PFS/DFS rates of 50% (95% Azacitidine has the most available data. Azacitidine first showed
CI, 41%-59%) and 44% (95% CI, 34%-53%), regardless of the rapid partial regression of cutaneous disease in 2 patients; however,
remission status at the time of allografting, suggesting that alloge- both developed neutropenia and died from sepsis at 8 and 9
neic HSCT is an effective treatment option in patients with months.74 The second study again showed initial partial regression
BPDCN. These pooled rates (OS and PFS/DFS) were determined of cutaneous lesions; however, all 3 patients had eventual disease
at a time point of 2 to 4 years after allograft. As most patients in the progression with survivals of 14, 17, and 25 months.75
4 studies included in the meta-analysis underwent allogeneic HSCT Pralatrexate was used as both first-line therapy (1 patient) and in
in CR1, a subgroup analysis of OS and PFS/DFS in patients who the setting of CHOP failure (2 patients). In all 3 patients, a cuta-
underwent HSCT in CR1 versus those who underwent allograft neous response was achieved. However, these patients experienced
beyond CR1 were performed. Predictably, patients who underwent disease progression.76,78
HSCT in CR1 had higher pooled rates of OS (67% vs. 7%) and The outcomes of patients with BPDCN treated with bend-
PFS/DFS (53% vs. 7%), thus supplementing the recommendation amustine was reviewed in a 5-patient case series. One patient ach-
of allogeneic HSCT in CR1 for BPDCN. Additionally, it was ieved a complete skin response for 7 months with eventual

Table 2 Low-intensity Therapy Regimens for BPDCN Treatment Reported in the Literature

Therapy Patients Mechanism Outcome Reference


Gemcitabine þ 1 (first-line, gemcitabine Pyrimidine antimetabolite Partial skin response, alive at time of publication 72
73
docetaxel monotherapy) Pyrimidine antimetabolite, Complete skin response, median PFS was 10.6 months
3 (all with 4þ prior microtubule inhibitor (1 alive at 15 months)
therapies)
74
Azacitidine 2 (first-line) DNA Methyltransferase Partial skin response, both developed neutropenia and sepsis,
75
3 (first-line) inhibitor median OS was 8 months
Partial skin response, 1 alive at time of publication, median OS
was 17 months (1 patient alive at 25 months)
76
Pralatrexate 1 (progression on CHOP) Antifolate Partial skin response, alive at 24 months
77
1 (first-line) Complete skin response, developed acute leukemia, OS 1 month
78
1 (progression on CHOP) Partial skin response, developed acute leukemia, OS 10 months
79
Bendamustine 5 (all with 1þ prior therapies) Alkylating and antimetabolite 1 complete skin response with progression of disease and OS
26 month, 1 developed tumor lysis syndrome at 34 months,
2 immediate progression of disease

Abbreviations: CHOP ¼ cyclophosphamide, doxorubicin, vincristine, and prednisone; OS ¼ overall survival; PFS ¼ progression-free survival.

Clinical Lymphoma, Myeloma & Leukemia Month 2019 -7


BPDCNeCurrent Insights
progression of disease, 1 died from tumor lysis syndrome, 2 were neurotoxicity, thus encouraging further evaluation of this immu-
primary therapy failure, and 1 was unevaluable owing to fatal notherapeutic strategy in both transplant-eligible and -ineligible
complications associated with an underlying myelodysplastic patients with BPDCN.88
syndrome.79 Montero et al reported BCL2 dependence of BPDCN through
Even in the face of initial treatment response, durability of dynamic BH3 profiling and demonstrated that BCL2 inhibitor
clinical remission and prolongation of survival is not reliably ob- (venetoclax) treatment elicited clinical response and prolonged
tained with all the lower intensity therapeutic options evaluated in survival in a xenograft model.89 Off-label use of venetoclax in 2
BPDCN. patients with R/R BPDCN who had progressed while receiving an
CD123 directed therapy attained a partial response at 4 weeks.
Skin-only BPDCN However, 1 patient died of intracranial hemorrhage secondary to
At least 34% of patients with BPDCN present with isolated skin thrombocytopenia (preceding venetoclax therapy), and the other
findings at the time of initial presentation.10 In a study of 14 pa- had disease progression at 12 weeks.89 Additionally, a recently
tients with isolated cutaneous BPDCN on presentation who published retrospective review of venetoclax combination therapy in
received first-line radiotherapy alone, 13 of 14 attained a response R/R myeloid malignancies, including 2 patients with BPDCN who
(CR, 8/13; PR, 5/13). However, the optimal dose of radiotherapy attained a cutaneous response with 1 patient attaining a positron
was not determined, and most patients eventually progressed.80 emission tomography/computed tomography response and > 50%
Radiation therapy may be an option in patients with isolated skin blast reduction in the bone marrow.90 Most recently, the clinical
lesions or in those ineligible for chemotherapy. Other treatment activity of venetoclax was demonstrated in 3 patients (n ¼ 3) who
options include ALL-based regimens and DeVIC (dexamethasone, received hyper-CVAD in combination with venetoclax. All 3 pa-
etoposide, ifosfamide, and carboplatin) with L-asparaginase.81 tients achieved complete remission, and 2 patients with R/R disease
were successfully bridged to HSCT.91 Based on these encouraging
Management of R/R Disease findings, a phase I study is underway to evaluate venetoclax mon-
R/R disease is associated with an extremely poor prognosis, lacking otherapy in patients with R/R BPDCN (NCT03485547).
effective treatment strategy. Clinical trial enrollment is the optimal Additionally, Sapienza et al, employing integrated bioinformatics,
management option for R/R disease, and in the absence of an demonstrated constitutive activation of the nuclear factor-kappa B
appropriate clinical trial option, prior therapy dictates the treatment (NF-kB) pathway in primary BPDCN cells and that the proteasome
choice. Patients who were previously treated with ALL-based inhibitor bortezomib was capable of inhibiting cell cycle progression
chemotherapy should be offered tagraxofusp and vice versa. In both in a BPDCN cell line (CAL-1) through NF-kB inhibition.26 These
cases, the treatment goal remains HSCT. Gemcitabine and docetaxel results were reinforced in 7 primary BPDCN samples where bor-
may be an effective treatment, given the evidence of its efficacy in a tezomib was associated with a decrease in RelA (NF-kB subunit)
heavily pre-treated population.72 Donor lymphocyte infusion has also expression and protracted the xenograft mouse survival.92
been used in patients who relapse after HSCT.34,82,83 Bromodomain and extra-terminal domain (BET) protein inhi-
bition is of therapeutic interest in BPDCN. Through gene expres-
Emerging Targeted Therapeutic sion profiling, Emadali et al demonstrated haploinsufficiency for
Options NR3C1 and a fusion between NR3C1 and a long noncoding RNA
Apart from tagraxofusp, there are several additional CD123- gene (lncRNA-3q). The overexpression of lncRNA-3q appeared to,
based targeted therapies under evaluation in phase I trials. in part, proffer sensitivity to BET inhibitor (BETi) in vitro.20 In
IMGN632 is a novel CD123-targeting antibody-drug conjugate another study, Ceribelli et al identified a BPDCN-specific tran-
composed of a humanized anti-CD123 antibody linked to a DNA scriptional network moderated by the E-box transcription factor
alkylating agent that has shown preclinical activity in CD123þ TCF4, which is in turn regulated by the BET protein BRD4.
malignancies, including BPDCN.84 It is currently being evaluated Consequently, high-throughput drug screening demonstrated that
in a phase I study for R/R hematologic malignancies including BET inhibition induced BPDCN apoptosis secondary to disruption
BPDCN, and preliminary results did not identify a dose-limiting of a BPDCN-specific transcriptional network governed by TCF4-
toxicity.85 A novel potent bispecific antibody, XmAb14045 (also dependent super-enhancers, denoting conceivable pre-clinical
known as SQZ622), targeting both CD123 and CD3 stimulating rationale for clinical trial evaluation of BETi in BPDCN.93
focused T cell-mediated killing of CD123þ cells, is being evaluated
in a phase I study that includes BPDCN.86 Conclusion
Additionally, Chimeric Antigen Receptor T cell (CAR-T) ther- BPDCN is a rare hematopoietic malignancy with universally
apy has shown broad promise in hematopoietic neoplasms. Pre- poor outcomes. Advances in pathobiological understanding of
clinical studies have demonstrated the potent anti-leukemic activity BPDCN and the advent of novel immunotherapeutic options offer
of CD123-CAR-T cells in an AML xenograft model.87 Subse- promising therapeutic strategies to patients with this aggressive
quently, this concept was demonstrated in a 74-year-old man with a neoplasm. As therapeutic approach to BPDCN becomes standard-
bulky subcutaneous mass treated with CD123þ CAR-T cells. The ized, a prognostic model for optimal therapeutic risk stratification
patient maintained CR at 60 days post-infusion with complete needs to be validated. Elucidating the molecular underpinnings of
resolution of clinical symptoms and no histopathologic evidence of BPDCN will aid in the identification of potential actionable ther-
disease at the time of response evaluation. The patient tolerated the apeutic targets to more effectively manage this rare recalcitrant
treatment and did not experience cytokine release syndrome or neoplasm.

8 - Clinical Lymphoma, Myeloma & Leukemia Month 2019


Sangeetha Venugopal et al
Disclosure plasmacytoid dendritic cell neoplasm reveals a unique pattern and suggests selective
sensitivity to NF-kB pathway inhibition. Leukemia 2014; 28:1606-16.
The authors have stated that they have no conflicts of interest. 27. Griffin GK, Togami K, Morgan EA, Lane AA. Developmental ontogeny of blastic
plasmacytoid dendritic cell neoplasm (BPDCN) revealed by recurrent high burden
clonal hematopoiesis, including in "skin-only" disease. Blood 2018; 132(Suppl 1):2755.
28. Alsidawi S, Westin GFM, Al-Kali A, Go RS. Blastic plasmacytoid dendritic cell
References neoplasm. a population-based analysis from the SEER and NCDB databases. Blood
1. Kameoka J, Ichinohasama R, Tanaka M, et al. A cutaneous agranular CD2- 2016; 128:4789.
CD4þ CD56þ “lymphoma”: report of two cases and review of the literature. Am J 29. Suzuki R, Nakamura S, Suzumiya J, et al, NK-cell Tumor Study Group. Blastic
Clin Pathol 1998; 110:478-88. natural killer cell lymphoma/leukemia (CD56-positive blastic tumor): prognosti-
2. Khoury JD, Medeiros LJ, Manning JT, Sulak LE, Bueso-Ramos C, Jones D. cation and categorization according to anatomic sites of involvement. Cancer 2005;
CD56(þ) TdT(þ) blastic natural killer cell tumor of the skin: a primitive systemic 104:1022-31.
malignancy related to myelomonocytic leukemia. Cancer 2002; 94:2401-8. 30. Jegalian AG, Facchetti F, Jaffe ES. Plasmacytoid dendritic cells: physiologic roles
3. Petrella T, Comeau MR, Maynadie M, et al. ‘Agranular CD4þ CD56þ hema- and pathologic states. Adv Anat Pathol 2009; 16:392-404.
todermic neoplasm’ (blastic NK-cell lymphoma) originates from a population of 31. Cota C, Vale E, Viana I, et al. Cutaneous manifestations of blastic plasmacytoid
CD56þ precursor cells related to plasmacytoid monocytes. Am J Surg Pathol 2002; dendritic cell neoplasm-morphologic and phenotypic variability in a series of 33
26:852-62. patients. Am J Surg Pathol 2010; 34:75-87.
4. Lucio P, Parreira A, Orfao A. CD123hi dendritic cell lymphoma: an unusual case 32. Petrella T. Blastic NK-cell lymphomas (agranular CD4 CD56 hematodermic
of non-Hodgkin lymphoma. Ann Int Med 1999; 131:549-50. neoplasms): a review. Am J Clin Pathol 2005; 123:662-75.
5. Chaperot L, Bendriss N, Manches O, et al. Identification of a leukemic coun- 33. Hwang K, Park CJ, Jang S, et al. Immunohistochemical analysis of CD123, CD56
terpart of the plasmacytoid dendritic cells. Blood 2001; 97:3210-7. and CD4 for the diagnosis of minimal bone marrow involvement by blastic
6. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health plasmacytoid dendritic cell neoplasm. Histopathology 2013; 62:764-70.
Organization (WHO) classification of myeloid neoplasms and acute leukemia: 34. Sullivan JM, Rizzieri DA. Treatment of blastic plasmacytoid dendritic cell
rationale and important changes. Blood 2009; 114:937-51. neoplasm. Hematol Am Soc Hematol Educ Program 2016; 2016:16-23.
7. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health 35. Barros Romao C, Santos Junior CJD, Leite LAC, et al. Blastic plasmacytoid
Organization classification of myeloid neoplasms and acute leukemia. Blood 2016; dendritic cell neoplasm with pulmonary involvement and atypical skin lesion. Am J
127:2391-405. Case Rep 2017; 18:692-5.
8. Guru Murthy GS, Pemmaraju N, Atallah E. Epidemiology and survival of blastic 36. Garcia-Colmenero L, Gallardo F, Salar A, Alvarez-Larran A, Colomo L, Pujol RM.
plasmacytoid dendritic cell neoplasm. Leuk Res 2018; 73:21-3. Blastic plasmacytoid dendritic cell neoplasm associated with dystrophic xantho-
9. Pagano L, Valentini CG, Pulsoni A, et al, GIMEMA-ALWP (Gruppo Italiano matosis after chemotherapy. Acta Derm Venereol 2017; 97:293-4.
Malattie EMatologiche dell’Adulto, Acute Leukemia Working Party). Blastic 37. Amitay-Laish I, Sundram U, Hoppe RT, Hodak E, Medeiros BC, Kim YH.
plasmacytoid dendritic cell neoplasm with leukemic presentation: an Italian Localized skin-limited blastic plasmacytoid dendritic cell neoplasm: a subset with
multicenter study. Haematologica 2013; 98:239-46. possible durable remission without transplantation. JAAD Case Rep 2017; 3:310-5.
10. Julia F, Dalle S, Duru G, et al. Blastic plasmacytoid dendritic cell neoplasms: 38. Feuillard J, Jacob MC, Valensi F, et al. Clinical and biologic features of CD4(þ)
clinico-immunohistochemical correlations in a series of 91 patients. Am J Surg CD56(þ) malignancies. Blood 2002; 99:1556-63.
Pathol 2014; 38:673-80. 39. Swerdlow SHCE, Harris NL, Jaffe ES, Pileri SA, Stein H. WHO Classification of
11. Herling M, Jones D. CD4þ/CD56þ hematodermic tumor: the features of an evolving Tumours of Haematopoietic and Lymphoid Tissues. 4th edition. Lyon, France: IARC
entity and its relationship to dendritic cells. Am J Clin Pathol 2007; 127:687-700. Press; 2008.
12. Jing Y, Shaheen E, Drake RR, Chen N, Gravenstein S, Deng Y. Aging is associated 40. Martin-Martin L, Almeida J, Pomares H, et al. Blastic plasmacytoid dendritic cell
with a numerical and functional decline in plasmacytoid dendritic cells, whereas neoplasm frequently shows occult central nervous system involvement at diagnosis
myeloid dendritic cells are relatively unaltered in human peripheral blood. Hum and benefits from intrathecal therapy. Oncotarget 2016; 7:10174-81.
Immunol 2009; 70:777-84. 41. Kong SH, Han SH, Suh HS, Choi YS. Blastic plasmacytoid dendritic cell
13. Vermi W, Lonardi S, Morassi M, et al. Cutaneous distribution of plasmacytoid neoplasm presenting as erythematous nodules with gallbladder involvement. Ann
dendritic cells in lupus erythematosus. Selective tropism at the site of epithelial Dermatol 2017; 29:501-3.
apoptotic damage. Immunobiology 2009; 214:877-86. 42. Tzankov A, Hebeda K, Kremer M, et al. Plasmacytoid dendritic cell proliferations
14. Nestle FO, Conrad C, Tun-Kyi A, et al. Plasmacytoid predendritic cells initiate and neoplasms involving the bone marrow: summary of the workshop cases sub-
psoriasis through interferon-alpha production. J Exp Med 2005; 202:135-43. mitted to the 18th Meeting of the European Association for Haematopathology
15. Vitte F, Fabiani B, Benet C, et al. Specific skin lesions in chronic myelomonocytic (EAHP) organized by the European Bone Marrow Working Group, Basel 2016.
leukemia: a spectrum of myelomonocytic and dendritic cell proliferations: a study Ann Hematol 2017; 96:765-77.
of 42 cases. Am J Surg Pathol 2012; 36:1302-16. 43. Jacob MC, Chaperot L, Mossuz P, et al. CD4þ CD56þ lineage negative ma-
16. Benet C, Gomez A, Aguilar C, et al. Histologic and immunohistologic charac- lignancies: a new entity developed from malignant early plasmacytoid dendritic
terization of skin localization of myeloid disorders: a study of 173 cases. Am J Clin cells. Haematologica 2003; 88:941-55.
Pathol 2011; 135:278-90. 44. Assaf C, Gellrich S, Whittaker S, et al. CD56-positive haematological neoplasms of
17. Leroux D, Mugneret F, Callanan M, et al. CD4(þ), CD56(þ) DC2 acute leu- the skin: a multicentre study of the Cutaneous Lymphoma Project Group of the
kemia is characterized by recurrent clonal chromosomal changes affecting 6 major European Organisation for Research and Treatment of Cancer. J Clin Pathol 2007;
targets: a study of 21 cases by the Groupe Français de Cytogénétique Héma- 60:981-9.
tologique. Blood 2002; 99:4154-9. 45. Herling M, Teitell MA, Shen RR, Medeiros LJ, Jones D. TCL1 expression in
18. Jardin F, Callanan M, Penther D, et al. Recurrent genomic aberrations combined plasmacytoid dendritic cells (DC2s) and the related CD4(þ) CD56(þ) blastic
with deletions of various tumour suppressor genes may deregulate the G1/S tumors of skin. Blood 2003; 101:5007-9.
transition in CD4þCD56þ haematodermic neoplasms and contribute to the 46. Petrella T, Meijer CJ, Dalac S, et al. TCL1 and CLA expression in agranular CD4/
aggressiveness of the disease. Leukemia 2009; 23:698-707. CD56 hematodermic neoplasms (blastic NK-cell lymphomas) and leukemia cutis.
19. Lucioni M, Novara F, Fiandrino G, et al. Twenty-one cases of blastic plasmacytoid Am J Clin Pathol 2004; 122:307-13.
dendritic cell neoplasm: focus on biallelic locus 9p21.3 deletion. Blood 2011; 118: 47. Pulford K, Banham AH, Lyne L, et al. The BCL11AXL transcription factor: its
4591-4. distribution in normal and malignant tissues and use as a marker for plasmacytoid
20. Emadali A, Hoghoughi N, Duley S, et al. Haploinsufficiency for NR3C1, the gene dendritic cells. Leukemia 2006; 20:1439-41.
encoding the glucocorticoid receptor, in blastic plasmacytoid dendritic cell neo- 48. Marafioti T, Paterson JC, Ballabio E, et al. Novel markers of normal and neoplastic
plasms. Blood 2016; 127:3040-53. human plasmacytoid dendritic cells. Blood 2008; 111:3778-92.
21. Sakamoto K, Katayama R, Asaka R, et al. Recurrent 8q24 rearrangement in blastic 49. Montes-Moreno S, Ramos-Medina R, Martínez-López A, et al. SPIB, a novel
plasmacytoid dendritic cell neoplasm: association with immunoblastoid cytomor- immunohistochemical marker for human blastic plasmacytoid dendritic cell neo-
phology, MYC expression, and drug response. Leukemia 2018; 32:2590-603. plasms: characterization of its expression in major hematolymphoid neoplasms.
22. Chang TC, Yu D, Lee YS, et al. Widespread microRNA repression by Myc Blood 2013; 121:643-7.
contributes to tumorigenesis. Nat Genet 2008; 40:43-50. 50. Boiocchi L, Lonardi S, Vermi W, Fisogni S, Facchetti F. BDCA-2 (CD303): a
23. Stenzinger A, Endris V, Pfarr N, et al. Targeted ultra-deep sequencing reveals highly specific marker for normal and neoplastic plasmacytoid dendritic cells. Blood
recurrent and mutually exclusive mutations of cancer genes in blastic plasmacytoid 2013; 122:296-7.
dendritic cell neoplasm. Oncotarget 2014; 5:6404-13. 51. Jordan CT, Upchurch D, Szilvassy SJ, et al. The interleukin-3 receptor alpha chain
24. Alayed K, Patel KP, Konoplev S, et al. TET2 mutations, myelodysplastic features, is a unique marker for human acute myelogenous leukemia stem cells. Leukemia
and a distinct immunoprofile characterize blastic plasmacytoid dendritic cell 2000; 14:1777-84.
neoplasm in the bone marrow. Am J Hematol 2013; 88:1055-61. 52. Dzionek A, Fuchs A, Schmidt P, et al. BDCA-2, BDCA-3, and BDCA-4: three
25. Menezes J, Acquadro F, Wiseman M, et al. Exome sequencing reveals novel and markers for distinct subsets of dendritic cells in human peripheral blood.
recurrent mutations with clinical impact in blastic plasmacytoid dendritic cell J Immunol 2000; 165:6037-46.
neoplasm. Leukemia 2013; 28:823-9. 53. Rizvi H, Paterson JC, Tedoldi S, et al. Expression of the CD2AP adaptor molecule
26. Sapienza MR, Fuligni F, Agostinelli C, et al. AIRC 5xMille consortium ‘Genetics- in normal, reactive and neoplastic human tissue. Pathologica 2012; 104:56-64.
driven targeted management of lymphoid malignancies and the Italian Registry on 54. Teitell M, Damore MA, Sulur GG, et al. TCL1 oncogene expression in AIDS-related
Blastic Plasmacytoid Dendritic Cell Neoplasm. Molecular profiling of blastic lymphomas and lymphoid tissues. Proc Natl Acad Sci U S A 1999; 96:9809-14.

Clinical Lymphoma, Myeloma & Leukemia Month 2019 -9


BPDCNeCurrent Insights
55. Lee B-S, Lee B-K, Iyer VR, et al. Corrected and republished from: BCL11A is a 75. Khwaja R, Daly A, Wong M, Mahé E, Cerquozzi S, Owen C. Azacitidine in the
critical component of a transcriptional network that activates RAG expression and treatment of blastic plasmacytoid dendritic cell neoplasm: a report of 3 cases. Leuk
V(D)J recombination. Mol Cell Biol 2017; 38. Lymphoma 2016; 57:2720-2.
56. Swerdlow S, Campo E, Harris N, et al. WHO Classification of Tumours of Hae- 76. Leitenberger JJ, Berthelot CN, Polder KD, et al. CD4þ CD56þ hematodermic/
matopoietic and Lymphoid Tissues. Revised 4th ed. Lyon, France: IARC; 2017. plasmacytoid dendritic cell tumor with response to pralatrexate. J Am Acad Der-
57. Vermi W, Facchetti F, Rosati S, et al. Nodal and extranodal tumor-forming matol 2008; 58:480-4.
accumulation of plasmacytoid monocytes/interferon-producing cells associated 77. Arranto C, Tzankov A, Halter J. Blastic plasmacytoid dendritic cell neoplasm with
with myeloid disorders. Am J Surg Pathol 2004; 28:585-95. transient response to pralatrexate. Ann Hematol 2017; 96:681-2.
58. Pileri SA, Ascani S, Cox MC, et al. Myeloid sarcoma: clinico-pathologic, pheno- 78. Sato S, Tanaka E, Tamai Y. Blastic plasmacytoid dendritic cell neoplasm with
typic and cytogenetic analysis of 92 adult patients. Leukemia 2006; 21:340-50. response to pralatrexate. Ann Hematol 2019; 98:801-3.
59. Sangle NA, Schmidt RL, Patel JL, et al. Optimized immunohistochemical panel to 79. Betrian S, Guenounou S, Luquet I, et al. Bendamustine for relapsed blastic plas-
differentiate myeloid sarcoma from blastic plasmacytoid dendritic cell neoplasm. macytoid dendritic cell leukaemia. Hematol Oncol 2017; 35:252-5.
Mod Pathol 2014; 27:1137-43. 80. Ishibashi N, Maebayashi T, Aizawa T, et al. Radiation therapy for cutaneous
60. Johnson RC, Kim J, Natkunam Y, et al. Myeloid cell nuclear differentiation an- blastic plasmacytoid dendritic cell neoplasm: a case report and review of the
tigen (MNDA) expression distinguishes extramedullary presentations of myeloid literature. Int J Clin Exp Med 2015; 8:8204-9.
leukemia from blastic plasmacytoid dendritic cell neoplasm. Am J Surg Pathol 81. Sugimoto KJ, Shimada A, Yamaguchi N, et al. Sustained complete remission of a
2016; 40:502-9. limited-stage blastic plasmacytoid dendritic cell neoplasm followed by a simulta-
61. Garnache-Ottou F, Feuillard J, Saas P. Plasmacytoid dendritic cell leukaemia/ neous combination of low-dose DeVIC therapy and radiation therapy: a case
lymphoma: towards a well defined entity? Br J Haematol 2007; 136:539-48. report and review of the literature. Int J Clin Exp Pathol 2013; 6:2603-8.
62. Frankel AE, Woo JH, Ahn C, et al. Activity of SL-401, a targeted therapy directed 82. Steinberg A, Kansal R, Wong M, et al. Good clinical response in a rare aggressive
to interleukin-3 receptor, in blastic plasmacytoid dendritic cell neoplasm patients. hematopoietic neoplasm: plasmacytoid dendritic cell leukemia with no cutaneous
Blood 2014; 124:385-92. lesions responding to 4 donor lymphocyte infusions following transplant. Case Rep
63. Frankel AE, Rossi P, Kuzel TM, Foss F. Diphtheria fusion protein therapy of Transplant 2011; 2011:651906.
chemoresistant malignancies. Curr Cancer Drug Targets 2002; 2:19-36. 83. Unteregger M, Valentin A, Zinke-Cerwenka W, et al. Unrelated SCT induces
64. Frankel AE, Seilles E, Biichle S, et al. Preclinical studies of SL-401, a targeted long-term remission in patients with blastic plasmacytoid dendritic cell neoplasm.
therapy directed to the interleukin-3 receptor (IL3-R), in blastic plasmacytoid Bone Marrow Transplant 2013; 48:799-802.
dendritic cell neoplasm (BPDCN): potent activity in BPDCN cell lines, primary 84. Kovtun Y, Jones GE, Adams S, et al. A CD123-targeting antibody-drug conjugate,
tumor, and in an in vivo model. Blood 2013; 122:3942. IMGN632, designed to eradicate AML while sparing normal bone marrow cells.
65. Pemmaraju N, Lane AA, Sweet KL, et al. Results of pivotal phase 2 clinical trial of Blood Adv 2018; 2:848-58.
tagraxofusp (SL-401) in patients with blastic plasmacytoid dendritic cell neoplasm 85. Daver NG, Erba HP, Papadantonakis N, et al. A phase I, first-in-human study
(BPDCN). Blood 2018; 132(Suppl 1):765. evaluating the safety and preliminary antileukemia activity of IMGN632, a novel
66. ELZONRIS [prescribing information]. New York, NY: Stemline Therapeutics, Inc; CD123-targeting antibody-drug conjugate, in patients with relapsed/refractory
2018. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/ acute myeloid leukemia and other CD123-positive hematologic malignancies.
761116s000lbl.pdf. Blood 2018; 132(Suppl 1):27.
67. Martin-Martin L, Lopez A, Vidriales B, et al. Classification and clinical behavior of 86. Ravandi F, Bashey A, Foran JM, et al. Complete responses in relapsed/refractory
blastic plasmacytoid dendritic cell neoplasms according to their maturation- acute myeloid leukemia (AML) patients on a weekly dosing schedule of
associated immunophenotypic profile. Oncotarget 2015; 6:19204-16. XmAb14045, a CD123 x CD3 T cell-engaging bispecific antibody: initial results
68. Reimer P, Rudiger T, Kraemer D, et al. What is CD4þCD56þ malignancy and of a phase 1 study. Blood 2018; 132(Suppl 1):763.
how should it be treated? Bone Marrow Transplant 2003; 32:637-46. 87. Mardiros A, Dos Santos C, McDonald T, et al. T cells expressing CD123-specific
69. Aoki T, Suzuki R, Kuwatsuka Y, et al. Long-term survival following autologous chimeric antigen receptors exhibit specific cytolytic effector functions and
and allogeneic stem cell transplantation for blastic plasmacytoid dendritic cell antitumor effects against human acute myeloid leukemia. Blood 2013; 122:3138-48.
neoplasm. Blood 2015; 125:3559-62. 88. Budde L, Song JY, Kim Y, et al. Remissions of acute myeloid leukemia
70. Kharfan-Dabaja MA, Al Malki MM, Deotare U, et al. Haematopoietic cell and blastic plasmacytoid dendritic cell neoplasm following treatment with
transplantation for blastic plasmacytoid dendritic cell neoplasm: a North American CD123-specific CAR T cells: a first-in-human clinical trial. Blood 2017; 130(Suppl
multicentre collaborative study. Br J Haematol 2017; 179:781-9. 1):811.
71. Kharfan-Dabaja MA, Reljic T, Murthy HS, Ayala E, Kumar A. Allogeneic he- 89. Montero J, Stephansky J, Cai T, et al. Blastic plasmacytoid dendritic cell neoplasm
matopoietic cell transplantation is an effective treatment for blastic plasmacytoid is dependent on BCL2 and sensitive to venetoclax. Cancer Discov 2017; 7:156-64.
dendritic cell neoplasm in first complete remission: systematic review and meta- 90. DiNardo CD, Rausch CR, Benton C, et al. Clinical experience with the BCL2-
analysis. Clin Lymphoma Myeloma Leuk 2018; 18:703-9.e1. inhibitor venetoclax in combination therapy for relapsed and refractory acute
72. Shepro D, Miller M. Gemcitabine in blastic plasmacytoid dendritic cell neoplasm myeloid leukemia and related myeloid malignancies. Am J Hematol 2018; 93:401-7.
(CD4þCD56þ hematodermic neoplasm). Blood 2014; 124:5457. 91. More on blastic plasmacytoid dendritic-cell neoplasms. N Engl J Med 2019; 380:695-7.
73. Ulrickson ML, Puri A, Lindstrom S, Cassaday RD, De Padova N, Becker PS. 92. Philippe L, Ceroi A, Bôle-Richard E, et al. Bortezomib as a new therapeutic
Gemcitabine and docetaxel as a novel treatment regimen for blastic plasmacytoid approach for blastic plasmacytoid dendritic cell neoplasm. Haematologica 2017;
dendritic cell neoplasm. Am J Hematol 2017; 92:E75-7. 102:1861-8.
74. Laribi K, Denizon N, Ghnaya H, et al. Blastic plasmacytoid dendritic cell 93. Ceribelli M, Hou ZE, Kelly PN, et al. A druggable TCF4- and BRD4-dependent
neoplasm: the first report of two cases treated by 5-azacytidine. Eur J Haematol transcriptional network sustains malignancy in blastic plasmacytoid dendritic cell
2014; 93:81-5. neoplasm. Cancer Cell 2016; 30:764-78.

10 - Clinical Lymphoma, Myeloma & Leukemia Month 2019

You might also like