eha 2019 results from ongoing phase 1/2 clinical trial of...

1
Results from Ongoing Phase 1/2 Clinical Trial of Tagraxofusp (SL - 401) in Patients with Relapsed/Refractory Chronic Myelomonocytic Leukemia (CMML) 1 Mayo Clinic, Rochester, 2 City of Hope, Duarte, 3 Kansas University Cancer Center, Westwood, 4 Princess Margaret Cancer Centre, Toronto, Canada, 5 Weill Cornell Medical Center, New York, 6 Roswell Park Comprehensive Cancer Center, Buffalo, 7 Ronald Reagan UCLA Medical Center, Los Angeles, 8 Stemline Therapeutics, New York, 9 University of Alberta Hospital, Edmonton, AB, Canada, 10 University of Michigan Health System, Ann Arbor, 11 The University of Texas MD Anderson Cancer Center, Houston EHA 2019 PF672 References Trial Design Baseline Demographics and Characteristics Safety and Tolerability Clinical Activity Overview: CMML Mrinal M. Patnaik 1 , Haris Ali 2 , Abdulraheem Yacoub 3 , Vikas Gupta 4 , Sangmin Lee 5 , Eunice Wang 6 , Gary Schiller 7 , Megan Sardone 8 , Halyna Wysowskyj 8 , Shay Shemesh 8 , Janice Chen 8 , Chris Brooks 8 , Enrique Poradosu 8 , Peter McDonald 8 , Nicole Rupprecht 8 , Animesh Pardanani 1 , Ayalew Tefferi 1 , Minakshi Taparia 9 , Moshe Talpaz 10 , Srdan Verstovsek 11 , Joseph Khoury 11 , Naveen Pemmaraju 11 Summary of Tagraxofusp Trial Results Introduction and Highlights CMML (all doses); Stages 1 and 2 (n=23) Duration of Treatment Background: CMML 0 2 4 6 8 10 12 14 16 Bone Marrow and Spleen Responses Individual patients Months Predictable and manageable safety profile No apparent cumulative AEs, including in the bone marrow, over multiple cycles In this Phase 1/2 trial, tagraxofusp was clinically active, with a predictable and manageable safety profile in patients with relapsed/refractory CMML – in particular, in patients with baseline splenomegaly (historically associated with advanced disease, morbidity, and poor prognosis) - 3 bone marrow CRs - 1 patient bridged to stem cell transplant (SCT) - 100% (12/12) of evaluable patients had a reduction in baseline splenomegaly - 67% (8/12) had reduction by ≥50% - 50% (4/8) with baseline spleen size ≥5cm had reduction by ≥50% Most common TRAEs include hypoalbuminemia (35%), thrombocytopenia (30%), nausea (26%) and vomiting (26%). Most common TRAEs, grade 3+, include thrombocytopenia (30%) and nausea (4%) Next steps include a pivotal program in patients with CMML Most Common Adverse Events (≥ 15% of treatment related adverse effects, TRAEs) Preferred Term All Grades n (%) TRAEs n (%) TRAEs All AEs G1 & 2 G3 G4 G5 Hypoalbuminaemia 8 (35) 10 (43) 8 (35) -- -- -- Thrombocytopenia 7 (30) 7 (30) -- 3 (13) 4 (17) -- Nausea 6 (26) 7 (30) 5 (22) 1 (4) -- -- Vomiting 6 (26) 9 (39) 6 (26) -- -- -- Anaemia 5 (22) 8 (35) 1 (4) 4 (17) -- -- Fatigue 4 (17) 8 (35) 4 (17) -- -- -- Oedema peripheral 4 (17) 10 (43) 4 (17) -- -- -- Weight increased 4 (17) 7 (30) 4 (17) -- -- -- 3 bone marrow complete responses (BMCRs) 1 patient bridged to stem cell transplant in remission on tagraxofusp 100% (12/12) spleen responses 67% (8/12) had reduction of ≥50% 50% (4/8) with baseline ≥5 cm had reduction ≥50% Patient CMML type CMML-1 CMML-1 CMML-2 CMML-1 CMML-1 CMML-1 CMML-1 CMML-1 CMML-1 CMML-2 CMML-2 CMML-2 CMML-2 CMML-1 CMML-2 CMML-1 CMML-1 CMML-1 CMML-1 CMML-2 Discontinued, progression Discontinued, AE Discontinued, physician decision Discontinued, withdrawal by patient u a u b u c u d Ongoing 7 μg/kg/day dose cohort 9 μg/kg/day dose cohort 12 μg/kg/day dose cohort Spleen Response Stem Cell Transplant (SCT) SR Tagraxofusp Novel targeted therapy directed to CD123 FDA-approved for the treatment of adult and pediatric patients, 2 years and older, with blastic plasmacytoid dendritic cell neoplasm (BPDCN) - Breakthrough Therapy Designation (BTD) designation Marketing Authorization Application (MAA) for BPDCN granted accelerated assessment, and under review, by the EMA CD123 target Expressed by multiple malignancies, including certain myeloproliferative neoplasms (MPN) such as chronic myelomonocytic leukemia (CMML) and myelofibrosis (MF), certain acute myeloid leukemia (AML) patient subsets, BPDCN and others Tagraxofusp and CMML Tagraxofusp has demonstrated clinical activity, with a predictable and manageable safety profile, in this Phase 1/2 trial (NCT02268253) of patients with relapsed/refractory CMML Patient enrollment is ongoing Ø Given the encouraging data from this trial and the unmet medical need in patients with CMML, a pivotal program is being constructed Aggressive myeloid malignancy, characterized by monocytosis Median age: 72-76 years Poor prognosis Presents with myelodysplastic (MDS) or myeloproliferative (MPN) features - Originally classified as an myelodysplastic syndrome (MDS) - Has since been re-classified as an MDS/MPN o MD-CMML (myelodysplastic CMML): WBC <13 x 10 9 /L o MP-CMML (myeloproliferative CMML): WBC 13 x 10 9 /L; characterized by advanced disease, splenomegaly, RAS pathway mutations, poor prognosis Historically: Hypomethylating agents (HMAs) were approved for myelodysplastic syndrome (MDS) at a time when CMML was considered an MDS - First-line CMML, historically: o In MDS pivotal trials, ORR 11%-27% o Subsequent to approvals, additional clinical trials demonstrated ORR centered at ~30%-40% (range ~25%-75%) upon initial exposure to HMAs, but responses generally not sustained, with CR rates ~15% o Median overall survival (OS): 24-36 months - Relapsed/refractory CMML, historically: o Outcomes have been described as dismal, irrespective of management o Median OS: 6-7 months Currently: ~50% of CMML now considered a myeloproliferative neoplasm (MPN) - International consortium recommended revising response criteria (historically MDS- focused) to capture MPN elements (Savona, 2015) a 12 μg/kg/day was highest tested dose (MTD not reached) and selected for Stage 2. CMML = chronic myelomonocytic leukemia; MF = myelofibrosis, SM = systemic mastocytosis; PED = primary eosinophilic disorders; IV = intravenous; MTD = maximum tolerated dose Stage 1 Lead-in (Complete) MPN: CMML or MF without evidence of transformation Tagraxofusp (12 μg/kg) a via IV infusion, days 1-3 of a 21-day cycle (cycles 1-4), a 28-day cycle (cycles 5-7); a 42-day cycle thereafter Key objectives: To further define safety and efficacy Stage 2 Expansion (Enrolling) MPN: CMML, MF, SM, and PED Tagraxofusp (7, 9, or 12 ug/kg) via IV infusion, days 1-3 of a 21-day cycle (cycles 1-4), a 28-day cycle (cycles 5-7); a 42-day cycle thereafter Key objectives: To determine optimal dose and regimen for Stage 2 1 BM involvement defined as blast count ≥5% 2 Patient received prior therapy and SCT 3 One patient did not have these data at the time of cut-off Prior Therapy for CMML, n (%) Hypomethylating agent (HMA) 11 (48) Prior Systemic Therapy (PST) 9 (39) Stem cell transplant (SCT) 2 3 (13) No prior systemic therapy for CMML 2 (9) Cytogenetic Risk Category 3 High risk 8 (35) Intermediate risk 8 (35) Low risk 4 (17) Other mutations 2 (9) Age, years n = 23 Median [range] 69 [42-80] Gender Male 19 (83) CMML Type CMML-1 15 (65) CMML-2 8 (35) ECOG Median [range] 1 [0-2] Median Blast Count, % Median [range] 6.0 [0-18] Baseline Sites of Disease, n (%) Bone marrow (BM) 1 17 (74) Spleen 12 (52) Liver 4 (17) ECOG = Eastern Cooperative Oncology Group * There were 3 cases of capillary leak syndrome, all grade 2 Tagraxofusp, Mechanism of Action, and Rationale in CMML Truncated diphtheria toxin payload IL-3 IL-3R/CD123 Cancer cell Tagraxofusp Tagraxofusp is a targeted therapy directed to CD123 Novel targeted therapy directed to CD123 FDA-approved for the treatment of adult and pediatric patients, 2 years and older, with blastic plasmacytoid dendritic cell neoplasm (BPDCN) - Breakthrough Therapy Designation (BTD) Marketing Authorization Application (MAA) granted accelerated assessment, and under review, by the EMA CMML and BPDCN overlap CMML and BPDCN can share genetic alterations and have a common clonal origin CMML can transform into BPDCN CD123 expression in CMML CMML blasts CMML monocytes Neoplastic pDCs 1 in CMML microenvironment Riaz. Cancer Control. 2014; Patnaik. Blood Cancer J. 2018; Brunetti. Leukemia. 2017; Krishnan. ASH 2018.; Ji. Blood. 2014 Spleen CD123 expression CD123+ Bone Marrow 1 Plasmacytoid dendritic cell (pDC) is the cell of origin of BPDCN CD123+ = Patient bridged to SCT in remission on tagraxofusp 1 Measured by physical exam (cm below costal margin [BCM]) HMA = hypomethylating agent; PST = prior systemic therapy; SCT = stem cell transplant; Clo = clofarabine; N/E = not evaluable; N/L = not listed; N/A = not available; SD=stable disease; PD=progressive disease CMML-2 CMML-1 CMML-2 6 1 3 15 17 5 10 4 23 11 19 2 22 12 13 9 18 20 7 16 8 14 21 Patient bridged to SCT u a u a u a u b u a u a u a u a u a u c u a u a u a u b u b u b u a u a u a u a u d SR (33%) SR (80%) and BMCR SR (100%) and BMCR SR (100%) and BMCR SR (100%) SR (100%) SR (21%) SR (100%) SR (100%) SR (50%) SR (36%) SR (26%) Pt # Line Bone Marrow CR Spleen Responses 1 Pre → Post Pre → Post % Response 11 3 10 cm → 0 cm 100% 5 2 6% → 1% 5 cm → 0 cm 100% 12 2 10% → 1% 4 cm → 0 cm 100% 3 2 2 cm → 0 cm 100% 9 2 N/E 2 cm → 0 cm 100% 20 1 N/E 2 cm → 0 cm 100% 17 2 15% → 2% (bridged to stem cell transplant) 10 cm → 2 cm 80% 1 2 20 cm → 10 cm 50% 8 2 22 cm → 14 cm 36% 23 2 N/E 6 cm → 4 cm 33% 21 2 N/E 27 cm → 20 cm 26% 6 2 14 cm → 11 cm 21% 1 Patients with palpable spleen at baseline. BCM = below costal margin (by physical exam); N/E = not evaluable Next Steps for Tagraxofusp in Patients with CMML Given the encouraging data from this trial and the unmet medical need in patients with CMML, a pivotal program is being constructed The protocol is currently being designed to incorporate these elements - Eligibility § Patients with CMML who failed first-line cytoreductive therapy - Endpoints and criteria § ORR (CR + PR), supported by duration, transfusion independence, safety § Additional endpoints and criteria to be assessed for potential clinical benefit include o BM response with partial hematopoietic recovery; correlation to transfusion- independence and decreased risk of infections o Spleen size o CD123 expression level - Trial design § Single-arm, non-randomized § Open new cohort (Stage 3) to current study 0314 o Stage 3a: assess potential clinical benefit of additional efficacy endpoints and criteria o Stage 3b: ORR +/- additional elements assessed in Stage 3a § An additional arm of patients with first-line CMML unlikely to benefit from available therapies is also under consideration ELZONRIS TM [prescribing information]. New York, NY: Stemline Therapeutics Inc; 2018 Frankel et al. Blood 2014; 124:385-92 Jordan et al. Leukemia 2000; 14:1177-84 Pardanani et al. Leukemia 2015; 29:1605-8 Chauhan et al. Cancer Cell 2009; 16:309-23 Frovola et al. Br J Haematol. 2014; 166:862-74 Coustan-Smith et al. Blood 2011; 117:6267-6276 Munoz et al. Haematologica 2001; 86:1261-9 Aldinucci et al. Leuk Lymphoma 2005; 46:303-11 Tehranchi et al. NEJM 2010; 363:1025-37 Loghavi et al. Blood Adv. 2018;2:1807 Moyo et al. Cur. Hematol. Malig. Reports 2017; 12: 468 Orazi et al. Leukemia 2008; 22:1308 VIDAZA® [prescribing info.]. Summit, NJ: Celgene; 2018 Padron et al. Cl. Adv. In Hematol. & Oncol. 2014; 12:172 Pophali et al. Am. J. Hematol. 2018; 93:1347 Patnaik et al. Am. J. Hematol. 2016; 91:631 Christie et al. ASH 2015; Abstract #3797 Black et al. Leukemia 2003; 17:155-9 Diefenbach et al. Blood 2011; 118:3737 Brooks et al. Blood 2013; 122:4104 Pemmaraju et al. N Engl J Med; 2019; 380(17):1628-1637 Chauhan et al. Leukemia 2017; 135 Solary et al. Blood 2017; 130:126-136 Alfonso et al. Amer J Hematol 2017; 92:599-606 Savona et al. Blood 2015; 125:1857 Alfonso et al. Am. J. Hematol. 2017; 92:599 Itzykson et al. Intl. J. Hematol. 2017; 150:711 DACOGEN® [prescribing info.]. Rockville, MD: Otsuka; 2018 Patient Dose (μg/kg/d) Line Prior Therapy CMML Type WBC (10 9 /L) BONE MARROW SPLEEN 1 Baseline (BM blast %) Best Response (BM blast %) BMCR Baseline (cm) Best Response (cm) Spleen Size Reduction 11 12 3 HMA CMML-1 5.3 5 PD 10 0 100% 5 9 2 HMA CMML-1 44.7 6 1 6% → 1% 5 0 100% 12 12 2 PST CMML-2 9.3 10 1 10% → 1% 4 0 100% 3 12 2 HMA CMML-1 99.2 7.6 SD 2 0 100% 9 12 2 HMA CMML-2 66.1 18 N/A N/E 2 0 100% 20 12 1 -- CMML-1 16.3 4 SD N/E 2 0 100% 17 12 2 PST CMML-2 8.1 15 2 15% → 2% 10 2 80% 1 7 2 PST; SCT CMML-2 33.8 15 SD 20 10 50% 8 12 2 HMA CMML-1 64.2 6 PD 22 14 36% 23 12 2 PST CMML-1 31.8 2 Pending N/E 6 4 33% 21 12 2 PST CMML-1 25.6 2 N/A N/E 27 20 26% 6 12 2 PST CMML-1 27.2 8 N/A 14 11 21% 10 12 2 HMA CMML-1 2.7 11 7 No splenomegaly N/E 7 12 2 HMA CMML-1 15.0 9 7 No splenomegaly N/E 13 12 2 PST CMML-1 26.1 6 4.9 No splenomegaly N/E 2 9 2 PST CMML-2 21.9 5 PD No splenomegaly N/E 14 12 2 PST CMML-2 33.8 14 SD No splenomegaly N/E 4 12 3 HMA; Clo CMML-1 18.5 0 3 N/E No splenomegaly N/E 15 12 1 -- CMML-1 12.3 6 SD No splenomegaly N/E 16 12 2 HMA; SCT CMML-1 3.3 3 N/A N/E No splenomegaly N/E 18 12 N/L N/L CMML-2 2.3 14 PD No splenomegaly N/E 19 12 2 HMA CMML-1 8.8 3 PD N/E No splenomegaly N/E 22 12 3 HMA; SCT CMML-2 4.3 6 N/A N/E No Splenomegaly N/E Blastic plasmacytoid dendritic cell neoplasm and chronic myelomonocytic leukemia: a shared clonal origin Leukemia (2017) 31, 12381240; doi:10.1038/leu.2017.38 Biallelic inactivation of the retinoblastoma gene results in transformation of chronic myelomonocytic leukemia to a blastic plasmacytoid dendritic cell neoplasm: shared clonal origins of two aggressive neoplasms Patnaik et al. Blood Cancer Journal (2018)8:82 DOI 10.1038/s41408-018-0120-5 Disclosures: Sardone: Stemline - employment, equity ownership; Wysowskyj: Stemline - employment, equity ownership; Shemesh: Stemline - employment, equity ownership; Chen: Stemline - employment, equity ownership; Brooks: Stemline - employment, equity ownership; Poradosu: Stemline - employment, equity ownership; McDonald: Stemline - employment, equity ownership; Rupprecht: Stemline - employment, equity ownership; Khoury: Stemline - research funding; Pemmaraju: Stemline - research funding; Schiller: Stemline - research funding; Patnaik: Stemline - research funding

Upload: others

Post on 30-Sep-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: EHA 2019 Results from Ongoing Phase 1/2 Clinical Trial of ...stemline.com/wp-content/uploads/2019/06/EHA-CMML-poster-060519-Print.pdfEHA 2019 PF672 References Trial Design Baseline

Results from Ongoing Phase 1/2 Clinical Trial of Tagraxofusp (SL-401) in Patients with Relapsed/Refractory Chronic Myelomonocytic Leukemia (CMML)

1Mayo Clinic, Rochester, 2City of Hope, Duarte, 3Kansas University Cancer Center, Westwood, 4Princess Margaret Cancer Centre, Toronto, Canada, 5Weill Cornell Medical Center, New York, 6Roswell Park Comprehensive Cancer Center, Buffalo, 7Ronald Reagan UCLA Medical Center, Los Angeles, 8Stemline Therapeutics, New York, 9University of Alberta Hospital, Edmonton, AB, Canada,10University of Michigan Health System, Ann Arbor, 11The University of Texas MD Anderson Cancer Center, Houston

EHA 2019 PF672

References

Trial Design

Baseline Demographics and Characteristics

Safety and Tolerability

Clinical Activity Overview: CMML

Mrinal M. Patnaik1, Haris Ali2, Abdulraheem Yacoub3, Vikas Gupta4 , Sangmin Lee5, Eunice Wang6, Gary Schiller7, Megan Sardone8, Halyna Wysowskyj8, Shay Shemesh8, Janice Chen8, Chris Brooks8, Enrique Poradosu8, Peter McDonald8, Nicole Rupprecht8, Animesh Pardanani1, Ayalew Tefferi1, Minakshi Taparia9, Moshe Talpaz10 ,Srdan Verstovsek11, Joseph Khoury11, Naveen Pemmaraju11

Summary of Tagraxofusp Trial Results

Introduction and Highlights

CMML (all doses); Stages 1 and 2 (n=23)

Duration of Treatment

Background: CMML

0 2 4 6 8 10 12 14 16

Bone Marrow and Spleen Responses

Indi

vidu

al p

atie

nts

Months

• Predictable and manageable safety profile• No apparent cumulative AEs, including in the bone marrow, over multiple cycles

• In this Phase 1/2 trial, tagraxofusp was clinically active, with a predictable and manageable safety profile in patients with relapsed/refractory CMML – in particular, in patients with baseline splenomegaly (historically associated with advanced disease, morbidity, and poor prognosis)

- 3 bone marrow CRs

- 1 patient bridged to stem cell transplant (SCT)

- 100% (12/12) of evaluable patients had a reduction in baseline splenomegaly

- 67% (8/12) had reduction by ≥50%

- 50% (4/8) with baseline spleen size ≥5cm had reduction by ≥50%

• Most common TRAEs include hypoalbuminemia (35%), thrombocytopenia (30%), nausea (26%) and vomiting (26%). Most common TRAEs, grade 3+, include thrombocytopenia (30%) and nausea (4%)

• Next steps include a pivotal program in patients with CMML

Most Common Adverse Events (≥ 15% of treatment related adverse effects, TRAEs)

Preferred TermAll Grades n (%) TRAEs n (%)

TRAEs All AEs G1 & 2 G3 G4 G5Hypoalbuminaemia 8 (35) 10 (43) 8 (35) -- -- --

Thrombocytopenia 7 (30) 7 (30) -- 3 (13) 4 (17) --

Nausea 6 (26) 7 (30) 5 (22) 1 (4) -- --

Vomiting 6 (26) 9 (39) 6 (26) -- -- --

Anaemia 5 (22) 8 (35) 1 (4) 4 (17) -- --

Fatigue 4 (17) 8 (35) 4 (17) -- -- --

Oedema peripheral 4 (17) 10 (43) 4 (17) -- -- --

Weight increased 4 (17) 7 (30) 4 (17) -- -- --

• 3 bone marrow complete responses (BMCRs)• 1 patient bridged to stem cell transplant in remission on tagraxofusp• 100% (12/12) spleen responses

– 67% (8/12) had reduction of ≥50%– 50% (4/8) with baseline ≥5 cm had reduction ≥50%

PatientCMML type

CMML-1

CMML-1

CMML-2

CMML-1

CMML-1

CMML-1

CMML-1

CMML-1

CMML-1

CMML-2

CMML-2

CMML-2

CMML-2

CMML-1

CMML-2

CMML-1

CMML-1

CMML-1

CMML-1

CMML-2 Discontinued, progressionDiscontinued, AEDiscontinued, physician decisionDiscontinued, withdrawal by patient

ua

ub

uc

ud

Ongoing

7 µg/kg/day dose cohort9 µg/kg/day dose cohort12 µg/kg/day dose cohortSpleen Response

Stem Cell Transplant (SCT)

SR

Tagraxofusp • Novel targeted therapy directed to CD123• FDA-approved for the treatment of adult and pediatric patients, 2 years and older, with

blastic plasmacytoid dendritic cell neoplasm (BPDCN) - Breakthrough Therapy Designation (BTD) designation

• Marketing Authorization Application (MAA) for BPDCN granted accelerated assessment, and under review, by the EMA

CD123 target• Expressed by multiple malignancies, including certain myeloproliferative neoplasms

(MPN) such as chronic myelomonocytic leukemia (CMML) and myelofibrosis (MF), certain acute myeloid leukemia (AML) patient subsets, BPDCN and others

Tagraxofusp and CMML• Tagraxofusp has demonstrated clinical activity, with a predictable and manageable

safety profile, in this Phase 1/2 trial (NCT02268253) of patients with relapsed/refractory CMML

• Patient enrollment is ongoingØ Given the encouraging data from this trial and the unmet medical need in

patients with CMML, a pivotal program is being constructed

• Aggressive myeloid malignancy, characterized by monocytosis• Median age: 72-76 years• Poor prognosis• Presents with myelodysplastic (MDS) or myeloproliferative (MPN) features

- Originally classified as an myelodysplastic syndrome (MDS)- Has since been re-classified as an MDS/MPN

o MD-CMML (myelodysplastic CMML): WBC <13 x 109/Lo MP-CMML (myeloproliferative CMML): WBC ≥13 x 109/L; characterized by

advanced disease, splenomegaly, RAS pathway mutations, poor prognosis • Historically: Hypomethylating agents (HMAs) were approved for myelodysplastic

syndrome (MDS) at a time when CMML was considered an MDS- First-line CMML, historically:

o In MDS pivotal trials, ORR 11%-27%o Subsequent to approvals, additional clinical trials demonstrated ORR centered

at ~30%-40% (range ~25%-75%) upon initial exposure to HMAs, but responses generally not sustained, with CR rates ~15%

o Median overall survival (OS): 24-36 months- Relapsed/refractory CMML, historically:

o Outcomes have been described as dismal, irrespective of managemento Median OS: 6-7 months

• Currently: ~50% of CMML now considered a myeloproliferative neoplasm (MPN)- International consortium recommended revising response criteria (historically MDS-

focused) to capture MPN elements (Savona, 2015)

a12 μg/kg/day was highest tested dose (MTD not reached) and selected for Stage 2.CMML = chronic myelomonocytic leukemia; MF = myelofibrosis, SM = systemic mastocytosis; PED = primary eosinophilic disorders; IV = intravenous; MTD = maximum tolerated dose

Stage 1 Lead-in (Complete)• MPN: CMML or MF without evidence of

transformation• Tagraxofusp (12 μg/kg)a via IV infusion, days 1-3 of a

21-day cycle (cycles 1-4), a 28-day cycle (cycles 5-7);a 42-day cycle thereafter

• Key objectives: To further define safety and efficacy

Stage 2 Expansion (Enrolling)

• MPN: CMML, MF, SM, and PED• Tagraxofusp (7, 9, or 12 ug/kg) via IV infusion, days

1-3 of a 21-day cycle (cycles 1-4), a 28-day cycle (cycles 5-7); a 42-day cycle thereafter

• Key objectives: To determine optimal dose and regimen for Stage 2

1 BM involvement defined as blast count ≥5%2 Patient received prior therapy and SCT3 One patient did not have these data at the time of cut-off

Prior Therapy for CMML, n (%)Hypomethylating agent (HMA) 11 (48)Prior Systemic Therapy (PST) 9 (39)Stem cell transplant (SCT)2 3 (13)No prior systemic therapy for CMML 2 (9)

Cytogenetic Risk Category3

High risk 8 (35)Intermediate risk 8 (35)Low risk 4 (17)Other mutations 2 (9)

Age, years n = 23Median [range] 69 [42-80]

GenderMale 19 (83)

CMML TypeCMML-1 15 (65)CMML-2 8 (35)

ECOGMedian [range] 1 [0-2]

Median Blast Count, %Median [range] 6.0 [0-18]

Baseline Sites of Disease, n (%)Bone marrow (BM)1 17 (74)Spleen 12 (52)Liver 4 (17)

ECOG = Eastern Cooperative Oncology Group

* There were 3 cases of capillary leak syndrome, all grade 2

Tagraxofusp, Mechanism of Action, and Rationale in CMML

Truncated diphtheria

toxin payload

IL-3

SL-401

IL-3R/CD123 Cancer cell

Tagraxofusp

Tagraxofusp is a targeted therapy directed to CD123• Novel targeted therapy directed to CD123

• FDA-approved for the treatment of adult and pediatric patients, 2 years and older, with blastic plasmacytoid dendritic cell neoplasm (BPDCN)

- Breakthrough Therapy Designation (BTD)

• Marketing Authorization Application (MAA) granted accelerated assessment, and under review, by the EMA

CMML and BPDCN overlap• CMML and BPDCN can share genetic alterations and

have a common clonal origin• CMML can transform into BPDCN

CD123 expression in CMML• CMML blasts• CMML monocytes• Neoplastic pDCs1 in CMML microenvironment

Riaz. Cancer Control. 2014; Patnaik. Blood Cancer J. 2018; Brunetti. Leukemia. 2017; Krishnan. ASH 2018.; Ji. Blood. 2014

Spleen

CD123 expression

CD123+

Bone Marrow

1Plasmacytoid dendritic cell (pDC) is the cell of origin of BPDCN

CD123+

= Patient bridged to SCT in remission on tagraxofusp1Measured by physical exam (cm below costal margin [BCM])HMA = hypomethylating agent; PST = prior systemic therapy; SCT = stem cell transplant; Clo = clofarabine; N/E = not evaluable; N/L = not listed; N/A = notavailable; SD=stable disease; PD=progressive disease

CMML-2

CMML-1

CMML-2

6

1

3

15

17

5

10

4

23

11

19

2

22

12

13

9

18

20

7

16

8

14

21

Patient bridged to SCT

ua

ua

ua

ub

ua

ua

ua

ua

ua

uc

ua

ua

ua

ub

ub

ub

ua

ua

ua

ua

ud

SR (33%)

SR (80%) and BMCR

SR (100%) and BMCR

SR (100%) and BMCR

SR (100%)

SR (100%)

SR (21%)

SR (100%)

SR (100%)

SR (50%)

SR (36%)

SR (26%)

Pt # Line Bone Marrow CR Spleen Responses1

Pre → Post Pre → Post % Response11 3 10 cm → 0 cm 100%5 2 6% → 1% 5 cm → 0 cm 100%12 2 10% → 1% 4 cm → 0 cm 100%3 2 2 cm → 0 cm 100%9 2 N/E 2 cm → 0 cm 100%20 1 N/E 2 cm → 0 cm 100%

17 2 15% → 2% (bridged to stem cell transplant) 10 cm → 2 cm 80%

1 2 20 cm → 10 cm 50%8 2 22 cm → 14 cm 36%23 2 N/E 6 cm → 4 cm 33%21 2 N/E 27 cm → 20 cm 26%6 2 14 cm → 11 cm 21%

1Patients with palpable spleen at baseline.

BCM = below costal margin (by physical exam); N/E = not evaluable

Next Steps for Tagraxofusp in Patients with CMML

• Given the encouraging data from this trial and the unmet medical need in patients with CMML, a pivotal program is being constructed

• The protocol is currently being designed to incorporate these elements- Eligibility

§ Patients with CMML who failed first-line cytoreductive therapy - Endpoints and criteria

§ ORR (CR + PR), supported by duration, transfusion independence, safety§ Additional endpoints and criteria to be assessed for potential clinical benefit include

o BM response with partial hematopoietic recovery; correlation to transfusion-independence and decreased risk of infections

o Spleen sizeo CD123 expression level

- Trial design § Single-arm, non-randomized§ Open new cohort (Stage 3) to current study 0314

o Stage 3a: assess potential clinical benefit of additional efficacy endpoints and criteriao Stage 3b: ORR +/- additional elements assessed in Stage 3a

§ An additional arm of patients with first-line CMML unlikely to benefit from available therapies is also under consideration

• ELZONRISTM [prescribing information]. New York, NY: Stemline Therapeutics Inc; 2018

• Frankel et al. Blood 2014; 124:385-92• Jordan et al. Leukemia 2000; 14:1177-84• Pardanani et al. Leukemia 2015; 29:1605-8• Chauhan et al. Cancer Cell 2009; 16:309-23• Frovola et al. Br J Haematol. 2014; 166:862-74• Coustan-Smith et al. Blood 2011; 117:6267-6276• Munoz et al. Haematologica 2001; 86:1261-9• Aldinucci et al. Leuk Lymphoma 2005; 46:303-11• Tehranchi et al. NEJM 2010; 363:1025-37• Loghavi et al. Blood Adv. 2018;2:1807• Moyo et al. Cur. Hematol. Malig. Reports 2017; 12: 468 • Orazi et al. Leukemia 2008; 22:1308• VIDAZA® [prescribing info.]. Summit, NJ: Celgene; 2018

• Padron et al. Cl. Adv. In Hematol. & Oncol. 2014; 12:172• Pophali et al. Am. J. Hematol. 2018; 93:1347 • Patnaik et al. Am. J. Hematol. 2016; 91:631• Christie et al. ASH 2015; Abstract #3797• Black et al. Leukemia 2003; 17:155-9• Diefenbach et al. Blood 2011; 118:3737• Brooks et al. Blood 2013; 122:4104• Pemmaraju et al. N Engl J Med; 2019; 380(17):1628-1637• Chauhan et al. Leukemia 2017; 135• Solary et al. Blood 2017; 130:126-136• Alfonso et al. Amer J Hematol 2017; 92:599-606 • Savona et al. Blood 2015; 125:1857• Alfonso et al. Am. J. Hematol. 2017; 92:599• Itzykson et al. Intl. J. Hematol. 2017; 150:711• DACOGEN® [prescribing info.]. Rockville, MD: Otsuka; 2018

Patient Dose (μg/kg/d) Line Prior

TherapyCMML Type

WBC (109/L)

BONE MARROW SPLEEN1

Baseline(BM blast %)

Best Response

(BM blast %)

BMCR Baseline (cm)

Best Response (cm)

Spleen Size

Reduction

11 12 3 HMA CMML-1 5.3 5 PD 10 0 100%5 9 2 HMA CMML-1 44.7 6 1 6% → 1% 5 0 100%12 12 2 PST CMML-2 9.3 10 1 10% → 1% 4 0 100%3 12 2 HMA CMML-1 99.2 7.6 SD 2 0 100%9 12 2 HMA CMML-2 66.1 18 N/A N/E 2 0 100%20 12 1 -- CMML-1 16.3 4 SD N/E 2 0 100%

17 12 2 PST CMML-2 8.1 15 2 15% → 2% 10 2 80%

1 7 2 PST; SCT CMML-2 33.8 15 SD 20 10 50%

8 12 2 HMA CMML-1 64.2 6 PD 22 14 36%23 12 2 PST CMML-1 31.8 2 Pending N/E 6 4 33%21 12 2 PST CMML-1 25.6 2 N/A N/E 27 20 26%6 12 2 PST CMML-1 27.2 8 N/A 14 11 21%10 12 2 HMA CMML-1 2.7 11 7 No splenomegaly N/E7 12 2 HMA CMML-1 15.0 9 7 No splenomegaly N/E13 12 2 PST CMML-1 26.1 6 4.9 No splenomegaly N/E2 9 2 PST CMML-2 21.9 5 PD No splenomegaly N/E14 12 2 PST CMML-2 33.8 14 SD No splenomegaly N/E4 12 3 HMA; Clo CMML-1 18.5 0 3 N/E No splenomegaly N/E15 12 1 -- CMML-1 12.3 6 SD No splenomegaly N/E

16 12 2 HMA;SCT CMML-1 3.3 3 N/A N/E No splenomegaly N/E

18 12 N/L N/L CMML-2 2.3 14 PD No splenomegaly N/E19 12 2 HMA CMML-1 8.8 3 PD N/E No splenomegaly N/E

22 12 3 HMA; SCT CMML-2 4.3 6 N/A N/E No Splenomegaly N/E

Blastic plasmacytoid dendritic cell neoplasm and chronicmyelomonocytic leukemia: a shared clonal origin

Leukemia (2017) 31, 1238–1240; doi:10.1038/leu.2017.38

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare,distinct entity of the WHO 2008 classification of hematopoietictumors1 that is thought to arise from plasmacytoid dendritic cellprecursors. BPDCN is characterized by the expansion of mono-morphic immature tumor cells expressing CD4, CD56, CD123 andTLC-1, usually in the bone marrow and the skin.1 The disease affectsboth young and old adults and the outcome of patients treated withstandard-dose therapeutic regimens is usually poor.1 Tumor cellsfrequently carry recurrent chromosomal abnormalities1 and, morerecently, several recurrently mutated genes were identified inBPDCN, including TET2 (36%), ASXL1 (32%) and NRAS (20%).2

About 10–20% of BPDCN patients are also affected by othermyeloid neoplasms, including myelodysplastic syndromes, chronicmyelomonocytic leukemia (CMML) and acute myeloid leukemia.CMML is a myelodysplastic/myeloproliferative neoplasm charac-terized by hypercellular marrow, persistent peripheral monocy-tosis, anemia and splenomegaly.1 Interestingly, CMML may carrymutations of the TET2, ASXL1 and RAS genes that are also involvedin BPDCN.2,3 Although the co-occurrence of such myeloidneoplasms and BPDCN has been widely reported in the literature,it remains uncertain whether these diseases are clonally related.Here we report the case of an 82-year-old patient with

asymptomatic, clinically stable CMML who, 2 years after CMMLdiagnosis, developed BPDCN with exclusive cutaneous involve-ment (Figure 1a). Blood counts showed only a mild monocytosisand thrombocytopenia at both CMML (WBC 4.2× 109/l, monocytes1.1× 109/l, Hb 14 g/dl, platelets 97× 109/l) and BPDCN diagnosis(WBC 3.4× 109/l, monocytes 1.1× 109/l, Hb 13.8 g/dl, platelets76× 109/l). BPDCN was confirmed by histological examination andimmunohistochemistry, demonstrating dermal infiltration byimmature cells expressing CD4, CD56, CD123 (Figure 1b), TLC-1and BDCA-2 antigens. The morphological picture as well asnegativity for myeloperoxidase (Figure 1b) and PGM1/CD68 (notshown) excluded the possibility of a concomitant CMMLcutaneous localization. Bone marrow examination was consistentwith the previous diagnosis of CMML but showed no BPDCNinvolvement, as documented by morphology, immunohistochem-istry (Figure 1c) and flow cytometry (o0.05% of bone marrowcells were CD4+, CD45+, CD123+). Bone marrow karyotyping andFISH were unremarkable. The patient underwent six cycles ofCHOP-like chemotherapy leading to complete resolution of theskin lesions with tolerable toxicities. Shortly after the treatmentcompletion, the patient relapsed with multiple neoplastic skinlesions and eventually died 2 months later due to bone marrowBPDCN progression.We performed targeted sequencing of 54 genes recurrently

mutated in myeloid malignancies (Supplementary Table 1) fromboth whole bone marrow (CMML) and skin (BPDCN) samplescollected at the time of BPDCN diagnosis. Biopsies were also takenfrom non-affected regions of the skin for germline control. Writteninformed consent was obtained before sampling of both skin andbone marrow. DNA was extracted from fresh-frozen samples andDNA libraries were generated with the TruSight Myeloid Sequen-cing Panel kit (Illumina, San Diego, CA, USA). Sequencing wasperformed with the MiSeq sequencer (Illumina). Average amplicon

coverages for BPDCN (skin), CMML (bone marrow) and controlnon-affected skin samples were 2375, 3781 and 2650, respectively.A custom filtering pipeline that included the removal of knownartifacts and normal polymorphisms, as well as a variant allelefrequency (VAF) of X 5% in tumor samples and a minimum readdepth of 300 in tumor and normal samples was used.Strikingly, the same mutations of TET2 (Y1244fsX1266; Q810X)

and SRSF2 (P95H) were detected in both bone marrow (CMML)and skin (BPDCN) samples (Figure 1d). In addition, a JAK2 V617Fmutation was present, but only in the bone marrow (Figure 1d).TET2 and SRSF2 mutations were likely clonal in both BPDCN (VAFsranging between 34 and 38%) and CMML samples (VAFs rangingbetween 46 and 50%), while the JAK2 mutation was present onlyin a CMML bone marrow subclone (VAF 15%; Figure 1d). Theslightly lower VAFs of TET2 and SRSF2 mutated alleles observed inthe BPDCN sample might be attributable to a lower purity ofneoplastic cells in the skin (Figure 1b). Sequencing of the clinicallyunaffected skin sample revealed the JAK2 mutation to be absentand the other three mutations to be present at low VAFs (o5%),possibly due to sub-clinical involvement by BPDCN.TET2 encodes for a methylcytosine dioxygenase that catalyzes

the conversion of 5-methylcytosine to 5-hydroxymethylcytosine,promoting active DNA demethylation.4 This gene is recurrentlymutated in a large variety of hematologic malignancies includingCMML3 and BPDCN.2,5 SRSF2 is a component of the spliceosomemachinery and it is commonly mutated at codon 95 in severalmyeloid malignancies, including 40% of CMML.3 While one studyalready reported SRSF2 mutations at codon 122 in two cases ofBPDCN,2 this is the first report of the P95H mutation in thisdisease. In keeping with our data, JAK2 V617F has been widelydescribed in CMML,3 but not in BPDCN.These findings clearly demonstrate that in our patient the CMML

and the BPDCN shared three early genetic events, two TET2mutations (Y1244fsX1266; Q810X) and one SRSF2 mutation (P95H),strongly suggesting the same clonal origin. Although it is notpossible to infer the exact order of acquisition of each mutation inour patient, it is clear that TET2 and SRSF2 mutations were earlyevents, being present in the vast majority of CMML and BPDCN cells.In contrast, the JAK2 V617F mutation was acquired later and only ina subclone of the CMML. The co-occurence of TET2 and SRSF2mutations has been reported as highly specific for CMML amongmyeloid neoplasms.6 As a consequence, one possible scenario isthat BPDCN arose from TET2/SRSF2-mutated CMML that only lateracquired a JAK2mutation in a subclone. An alternative model wouldinvolve a pre-leukemic clone carrying mutated TET2 and/or SRSF2,which later gave rise to CMML and BPDCN independently from oneanother. The latter hypothesis may be supported by the finding thatolder individuals with no previous history of hematopoieticneoplasm may carry clonal hematopoiesis of indeterminatepotential (CHIP), which is sustained by mutations of genes such asDNMT3A, TET2 and SRSF2.7–11 A similar phenomenon has beenobserved in pre-leukemic hematopoietic stem cells of patients withacute myeloid leukemia.12,13 Not surprisingly, CHIP is associated withan increased risk of developing hematologic cancers7 and interest-ingly the spectra of mutations of CHIP, CMML and BPDCN aresignificantly overlapping.2,5,7,14,15

Recently, several sequencing studies attempted to uncover thegenetic events that drive BPDCN.2,5,14 Although several myeloidgenes (for example, TET2, NRAS) were consistently reported as

Accepted article preview online 23 January 2017; advance online publication, 10 February 2017

Letters to the Editor

1238

Leukemia (2017) 1217 – 1250

Blastic plasmacytoid dendritic cell neoplasm and chronicmyelomonocytic leukemia: a shared clonal origin

Leukemia (2017) 31, 1238–1240; doi:10.1038/leu.2017.38

Blastic plasmacytoid dendritic cell neoplasm (BPDCN) is a rare,distinct entity of the WHO 2008 classification of hematopoietictumors1 that is thought to arise from plasmacytoid dendritic cellprecursors. BPDCN is characterized by the expansion of mono-morphic immature tumor cells expressing CD4, CD56, CD123 andTLC-1, usually in the bone marrow and the skin.1 The disease affectsboth young and old adults and the outcome of patients treated withstandard-dose therapeutic regimens is usually poor.1 Tumor cellsfrequently carry recurrent chromosomal abnormalities1 and, morerecently, several recurrently mutated genes were identified inBPDCN, including TET2 (36%), ASXL1 (32%) and NRAS (20%).2

About 10–20% of BPDCN patients are also affected by othermyeloid neoplasms, including myelodysplastic syndromes, chronicmyelomonocytic leukemia (CMML) and acute myeloid leukemia.CMML is a myelodysplastic/myeloproliferative neoplasm charac-terized by hypercellular marrow, persistent peripheral monocy-tosis, anemia and splenomegaly.1 Interestingly, CMML may carrymutations of the TET2, ASXL1 and RAS genes that are also involvedin BPDCN.2,3 Although the co-occurrence of such myeloidneoplasms and BPDCN has been widely reported in the literature,it remains uncertain whether these diseases are clonally related.Here we report the case of an 82-year-old patient with

asymptomatic, clinically stable CMML who, 2 years after CMMLdiagnosis, developed BPDCN with exclusive cutaneous involve-ment (Figure 1a). Blood counts showed only a mild monocytosisand thrombocytopenia at both CMML (WBC 4.2× 109/l, monocytes1.1× 109/l, Hb 14 g/dl, platelets 97× 109/l) and BPDCN diagnosis(WBC 3.4× 109/l, monocytes 1.1× 109/l, Hb 13.8 g/dl, platelets76× 109/l). BPDCN was confirmed by histological examination andimmunohistochemistry, demonstrating dermal infiltration byimmature cells expressing CD4, CD56, CD123 (Figure 1b), TLC-1and BDCA-2 antigens. The morphological picture as well asnegativity for myeloperoxidase (Figure 1b) and PGM1/CD68 (notshown) excluded the possibility of a concomitant CMMLcutaneous localization. Bone marrow examination was consistentwith the previous diagnosis of CMML but showed no BPDCNinvolvement, as documented by morphology, immunohistochem-istry (Figure 1c) and flow cytometry (o0.05% of bone marrowcells were CD4+, CD45+, CD123+). Bone marrow karyotyping andFISH were unremarkable. The patient underwent six cycles ofCHOP-like chemotherapy leading to complete resolution of theskin lesions with tolerable toxicities. Shortly after the treatmentcompletion, the patient relapsed with multiple neoplastic skinlesions and eventually died 2 months later due to bone marrowBPDCN progression.We performed targeted sequencing of 54 genes recurrently

mutated in myeloid malignancies (Supplementary Table 1) fromboth whole bone marrow (CMML) and skin (BPDCN) samplescollected at the time of BPDCN diagnosis. Biopsies were also takenfrom non-affected regions of the skin for germline control. Writteninformed consent was obtained before sampling of both skin andbone marrow. DNA was extracted from fresh-frozen samples andDNA libraries were generated with the TruSight Myeloid Sequen-cing Panel kit (Illumina, San Diego, CA, USA). Sequencing wasperformed with the MiSeq sequencer (Illumina). Average amplicon

coverages for BPDCN (skin), CMML (bone marrow) and controlnon-affected skin samples were 2375, 3781 and 2650, respectively.A custom filtering pipeline that included the removal of knownartifacts and normal polymorphisms, as well as a variant allelefrequency (VAF) of X 5% in tumor samples and a minimum readdepth of 300 in tumor and normal samples was used.Strikingly, the same mutations of TET2 (Y1244fsX1266; Q810X)

and SRSF2 (P95H) were detected in both bone marrow (CMML)and skin (BPDCN) samples (Figure 1d). In addition, a JAK2 V617Fmutation was present, but only in the bone marrow (Figure 1d).TET2 and SRSF2 mutations were likely clonal in both BPDCN (VAFsranging between 34 and 38%) and CMML samples (VAFs rangingbetween 46 and 50%), while the JAK2 mutation was present onlyin a CMML bone marrow subclone (VAF 15%; Figure 1d). Theslightly lower VAFs of TET2 and SRSF2 mutated alleles observed inthe BPDCN sample might be attributable to a lower purity ofneoplastic cells in the skin (Figure 1b). Sequencing of the clinicallyunaffected skin sample revealed the JAK2 mutation to be absentand the other three mutations to be present at low VAFs (o5%),possibly due to sub-clinical involvement by BPDCN.TET2 encodes for a methylcytosine dioxygenase that catalyzes

the conversion of 5-methylcytosine to 5-hydroxymethylcytosine,promoting active DNA demethylation.4 This gene is recurrentlymutated in a large variety of hematologic malignancies includingCMML3 and BPDCN.2,5 SRSF2 is a component of the spliceosomemachinery and it is commonly mutated at codon 95 in severalmyeloid malignancies, including 40% of CMML.3 While one studyalready reported SRSF2 mutations at codon 122 in two cases ofBPDCN,2 this is the first report of the P95H mutation in thisdisease. In keeping with our data, JAK2 V617F has been widelydescribed in CMML,3 but not in BPDCN.These findings clearly demonstrate that in our patient the CMML

and the BPDCN shared three early genetic events, two TET2mutations (Y1244fsX1266; Q810X) and one SRSF2 mutation (P95H),strongly suggesting the same clonal origin. Although it is notpossible to infer the exact order of acquisition of each mutation inour patient, it is clear that TET2 and SRSF2 mutations were earlyevents, being present in the vast majority of CMML and BPDCN cells.In contrast, the JAK2 V617F mutation was acquired later and only ina subclone of the CMML. The co-occurence of TET2 and SRSF2mutations has been reported as highly specific for CMML amongmyeloid neoplasms.6 As a consequence, one possible scenario isthat BPDCN arose from TET2/SRSF2-mutated CMML that only lateracquired a JAK2mutation in a subclone. An alternative model wouldinvolve a pre-leukemic clone carrying mutated TET2 and/or SRSF2,which later gave rise to CMML and BPDCN independently from oneanother. The latter hypothesis may be supported by the finding thatolder individuals with no previous history of hematopoieticneoplasm may carry clonal hematopoiesis of indeterminatepotential (CHIP), which is sustained by mutations of genes such asDNMT3A, TET2 and SRSF2.7–11 A similar phenomenon has beenobserved in pre-leukemic hematopoietic stem cells of patients withacute myeloid leukemia.12,13 Not surprisingly, CHIP is associated withan increased risk of developing hematologic cancers7 and interest-ingly the spectra of mutations of CHIP, CMML and BPDCN aresignificantly overlapping.2,5,7,14,15

Recently, several sequencing studies attempted to uncover thegenetic events that drive BPDCN.2,5,14 Although several myeloidgenes (for example, TET2, NRAS) were consistently reported as

Accepted article preview online 23 January 2017; advance online publication, 10 February 2017

Letters to the Editor

1238

Leukemia (2017) 1217 – 1250

Patnaik et al. Blood Cancer Journal �(2018)�8:82�DOI 10.1038/s41408-018-0120-5 Blood Cancer Journal

CORRESPONDENCE Open Ac ce s s

Biallelic inactivation of the retinoblastomagene results in transformation of chronicmyelomonocytic leukemia to a blasticplasmacytoid dendritic cell neoplasm:shared clonal origins of two aggressiveneoplasmsMrinal M. Patnaik 1, Terra Lasho1, Matthew Howard1, Christy Finke1, Rhett L. Ketterling2, Aref Al-Kali 1,Animesh Pardanani1, Nathalie Droin 3, Naseema Gangat1, Ayalew Tefferi1 and Eric Solary3

To the Editor,Chronic myelomonocytic leukemia (CMML) is a clonal

hematopoietic stem cell disorder characterized by sus-tained peripheral blood (PB) monocytosis, bone marrow(BM) dysplasia, and an inherent risk for transformation toacute myeloid leukemia (AML); ~20–30% over 3–5years1,2. Patients with CMML have ~10–15 gene muta-tions in coding DNA regions, with common mutationsinvolving TET2 (~60%), ASXL1 (~40%), SRSF2 (~40%),and the oncogenic RAS pathway (~30%)3. Blastic plas-macytoid dendritic cell neoplasm (BPDCN) is a rarehematodermic malignancy of dendritic cell origin that isknown to infiltrate the blood, BM, skin, and lymph nodes;a diagnosis of which is based on a blast immunopheno-type characterized by the expression of CD4, CD43,CD56, CD123, BDCA-2/CD303, TCL1, and CTLA1.Cytogenetic studies, array-based comparative genomichybridization, and gene expression profiling havedemonstrated recurrent inactivation/losses/decreasedexpression of RB1 (retinoblastoma 1; 13q13-q21), LATS2,CDKN1B, CDKN2A, and TP53 genes,4 while next-generation sequencing (NGS) studies have demonstrated

point mutations involving TET2 (~36%), ASXL1 (~32%),NRAS (~30%), ATM (21%), along with deletions involvingRB1 and APC (~6%)5,6. In addition, recurrent MYB generearrangements have been documented in both childrenand adults, with fusion oncogenes including MYB-ZFAT,MYB-PLEKHO1, and MYB-DCPS7. Common clonal ori-gins for CMML and BPDCN have been suggested basedon similar genetic and epigenetic deregulations identified,along with sporadic case reports of CMML transforma-tion to BPDCN8,9. We provide the first in-depth whole-exome sequencing (WES) results on a patient withCMML that transformed to BPDCN and provide infor-mation with regards to possible mechanisms oftransformation.A 61-year-old male was referred to the hematology

clinic in 2014 for sustained PB monocytosis. A BM biopsywas suggestive of CMML-0 (World Health Organization2016 criteria) with normal cytogenetics (Fig. 1). He didhave plasmacytoid dendritic cell nodules comprising~20% of the BM cellularity. NGS at diagnosis identifiedmutations involving TET2 (variant allele frequency (VAF)40%) and SRSF2 (43%). He was conservatively managedwith routine blood count checks. In 2017, he presentedwith worsening constitutional symptoms, a generalizedskin rash, and progressive cytopenias. A BM biopsy andskin biopsy were suggestive of BPDCN with normal BMmetaphase cytogenetics (Fig. 1). He was treated withAML-like induction chemotherapy using idarubicin and

© The Author(s) 2018OpenAccessThis article is licensedunder aCreativeCommonsAttribution 4.0 International License,whichpermits use, sharing, adaptation, distribution and reproductionin any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if

changesweremade. The images or other third partymaterial in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to thematerial. Ifmaterial is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtainpermission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Correspondence: Mrinal M. Patnaik ([email protected])1Department of Internal Medicine, Division of Hematology, Mayo Clinic,Rochester, MN, USA2Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester,MN, USAFull list of author information is available at the end of the article.

Blood Cancer Journal

1234

5678

90():,;

1234567890():,;

1234

5678

90():,;

1234567890():,;

Patnaik et al. Blood Cancer Journal �(2018)�8:82�DOI 10.1038/s41408-018-0120-5 Blood Cancer Journal

CORRESPONDENCE Open Ac ce s s

Biallelic inactivation of the retinoblastomagene results in transformation of chronicmyelomonocytic leukemia to a blasticplasmacytoid dendritic cell neoplasm:shared clonal origins of two aggressiveneoplasmsMrinal M. Patnaik 1, Terra Lasho1, Matthew Howard1, Christy Finke1, Rhett L. Ketterling2, Aref Al-Kali 1,Animesh Pardanani1, Nathalie Droin 3, Naseema Gangat1, Ayalew Tefferi1 and Eric Solary3

To the Editor,Chronic myelomonocytic leukemia (CMML) is a clonal

hematopoietic stem cell disorder characterized by sus-tained peripheral blood (PB) monocytosis, bone marrow(BM) dysplasia, and an inherent risk for transformation toacute myeloid leukemia (AML); ~20–30% over 3–5years1,2. Patients with CMML have ~10–15 gene muta-tions in coding DNA regions, with common mutationsinvolving TET2 (~60%), ASXL1 (~40%), SRSF2 (~40%),and the oncogenic RAS pathway (~30%)3. Blastic plas-macytoid dendritic cell neoplasm (BPDCN) is a rarehematodermic malignancy of dendritic cell origin that isknown to infiltrate the blood, BM, skin, and lymph nodes;a diagnosis of which is based on a blast immunopheno-type characterized by the expression of CD4, CD43,CD56, CD123, BDCA-2/CD303, TCL1, and CTLA1.Cytogenetic studies, array-based comparative genomichybridization, and gene expression profiling havedemonstrated recurrent inactivation/losses/decreasedexpression of RB1 (retinoblastoma 1; 13q13-q21), LATS2,CDKN1B, CDKN2A, and TP53 genes,4 while next-generation sequencing (NGS) studies have demonstrated

point mutations involving TET2 (~36%), ASXL1 (~32%),NRAS (~30%), ATM (21%), along with deletions involvingRB1 and APC (~6%)5,6. In addition, recurrent MYB generearrangements have been documented in both childrenand adults, with fusion oncogenes including MYB-ZFAT,MYB-PLEKHO1, and MYB-DCPS7. Common clonal ori-gins for CMML and BPDCN have been suggested basedon similar genetic and epigenetic deregulations identified,along with sporadic case reports of CMML transforma-tion to BPDCN8,9. We provide the first in-depth whole-exome sequencing (WES) results on a patient withCMML that transformed to BPDCN and provide infor-mation with regards to possible mechanisms oftransformation.A 61-year-old male was referred to the hematology

clinic in 2014 for sustained PB monocytosis. A BM biopsywas suggestive of CMML-0 (World Health Organization2016 criteria) with normal cytogenetics (Fig. 1). He didhave plasmacytoid dendritic cell nodules comprising~20% of the BM cellularity. NGS at diagnosis identifiedmutations involving TET2 (variant allele frequency (VAF)40%) and SRSF2 (43%). He was conservatively managedwith routine blood count checks. In 2017, he presentedwith worsening constitutional symptoms, a generalizedskin rash, and progressive cytopenias. A BM biopsy andskin biopsy were suggestive of BPDCN with normal BMmetaphase cytogenetics (Fig. 1). He was treated withAML-like induction chemotherapy using idarubicin and

© The Author(s) 2018OpenAccessThis article is licensedunder aCreativeCommonsAttribution 4.0 International License,whichpermits use, sharing, adaptation, distribution and reproductionin any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if

changesweremade. The images or other third partymaterial in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to thematerial. Ifmaterial is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtainpermission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

Correspondence: Mrinal M. Patnaik ([email protected])1Department of Internal Medicine, Division of Hematology, Mayo Clinic,Rochester, MN, USA2Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester,MN, USAFull list of author information is available at the end of the article.

Blood Cancer Journal

1234

5678

90():,;

1234

5678

90():,;

1234

5678

90():,;

1234

5678

90():,;

Disclosures: Sardone: Stemline - employment, equity ownership; Wysowskyj: Stemline - employment, equity ownership; Shemesh: Stemline -employment, equity ownership; Chen: Stemline - employment, equity ownership; Brooks: Stemline - employment, equity ownership; Poradosu: Stemline - employment, equity ownership; McDonald: Stemline - employment, equity ownership; Rupprecht: Stemline - employment, equity ownership; Khoury: Stemline - research funding; Pemmaraju: Stemline - research funding; Schiller: Stemline - research funding; Patnaik: Stemline - research funding