Clearance of ‘Driver-COSMIC’ mutations post-CR1 with persisting RUNX1_L56S is unlikely to contribute towards disease progression L . Rai1, T. Boneva1,

Slides:



Advertisements
Similar presentations
Making Sense of Novel Prognostics: NOTCH1, SF3B1 Jennifer R Brown, MD PhD Director, CLL Center Dana-Farber Cancer Institute October 24, 2014.
Advertisements

Supervisor: VS 高志平 Reporter: R4 張妙而.  Mutations in nucleophosmin 1 ( NPM1 ) gene, one of the most common gene mutations (25%-30%) in AML  NPM1 mut co-occurs.
IN ACUTE MYELOID LEUKEMIA, THE USE IN INDUCTION OF STANDARD DOSE ARA-C IS ASSOCIATED WITH A BETTER QUALITY OF RESPONSE AS COMPARED TO AN INDUCTION REGIMEN.
Long Term Follow-Up After Imatinib Cessation for Patients in Deep Molecular Response: The Update Results of the STIM1 Study1 Preliminary Report of the.
ACUTE MYELOID LEUKEMIA Irit Avivi
Acute Myeloid Leukemia
A 14-gene prognosis signature predicts metastasis risk in node-negative, estrogen receptor-positive, Tamoxifen-treated breast cancer in different ethnogeographic.
DR. YETUNDE T. ISRAEL-AINA PAEDIATRICIAN, UNIVERSITY OF BENIN TEACHING HOSPITAL, BENIN CITY BENIN BLOOD AND MARROW TRANSPLANT WORKSHOP, UNIVERSITY OF BENIN.
5-Azacitidine For Myelodysplasia Before Allogeneic Hematopoietic Cell Transplantation Field T et al. Bone Marrow Transplant 2009:[Epub ahead of print].
Current Status of Acute Myeloid Leukemia in China Jianxiang Wang Institute of Hematology Hospital of Blood Disease Chinese Academy of Medical Sciences.
M. Sales1, N. Foster1, S. Tauro2, J. Cunningham1, N. Pratt1
Single-Agent Lenalidomide Induces Complete Remission of Acute Myeloid Leukemia in Patients with Isolated Trisomy 13 Fehniger TA et al. Blood 2009;113(5):
A Phase II Study of Lenalidomide for Previously Untreated Deletion (del) 5q Acute Myeloid Leukemia (AML) Patients Age 60 or Older Who Are Not Candidates.
Low Dose Decitabine Versus Best Supportive Care in Elderly Patients with Intermediate or High Risk MDS Not Eligible for Intensive Chemotherapy: Final Results.
Case report Sudden blastic transformation in patient with chronic myeloid leukemia treated with imatinib mesylate Mehrdad Payandeh,MD Hematology, Medical.
R2 김재민 / Prof. 윤휘중 Journal conference 1.
Case 251: Clinical Information Raymond E Felgar, MD, PhD University of Pittsburgh, Pittsburgh, PA 45-year-old man with recent history of shingles, night.
SH/EAHP Workshop 2013 Case 93 Winnie Wu, M.D. Sheeja Pullarkat, M.D.
P Ferguson, R Hills, A Grech, L Kjeldsen, M Dennis, P Vyas, R Clark, N Russell, C Craddock, On behalf of the NCRI AML Working Group. An operational definition.
Preliminary Results of a Multicenter Phase II Trial of 5-Day Decitabine as Front-Line Therapy for Elderly Patients with Acute Myeloid Leukemia (AML) Cashen.
May 29 - June 2, 2015 Leukemia Stem Cell Phenotypes Correlate With Cytogenetic Risk Factors and Outcomes CCO Independent Conference Highlights of the 2015.
Identification of mutations prognostic of relapse after allogeneic transplantation and novel clone emergence at disease recurrence: implications for strategies.
LOCALIZED FOLLICULAR LYMPHOMA: SPREAD OF Bcl-2/IgH+ CELLS IN THE BLOOD AND BONE MARROW FROM THE PRIMARY SITE OF DISEASE AND POSSIBILITY OF CLEARANCE AFTER.
Margaret L. Gulley, Thomas C. Shea, Yuri Fedoriw 
Immunotherapy with CD19 CAR redirected T-cells for high risk, relapsed paediatric CD19+ acute lymphoblastic leukaemia (ALL) and other haematological malignancies.
39th ESMO Congress Madrid, Spain – 30 September Poster 979P
Figure S1. A B C Survival probability (%) P = 0.21 P = 0.37 P = 0.12
19-28z CAR T-Cell Efficacy and Toxicity in Adults With R/R B-Cell ALL
Final Results from a Phase 2 Study of Pracinostat in Combination with Azacitidine in Elderly Patients with Acute Myeloid Leukemia (AML)1   CC-486 (Oral.
1 Stone RM et al. Proc ASH 2015;Abstract 6.
  PROGNOSTIC SIGNIFICANCE OF GENE MUTATIONS IN MDS DEPENDS ON THE LOCI OF GENE VARIANCES PROGNOSTIC SIGNIFICANCE OF GENE MUTATIONS IN MDS DEPENDS ON THE.
at First Pavlov State Medical University of Saint-Petersburg, Russia
New Findings in Hematology: Independent Conference Coverage
Verstovsek S et al. Proc ASH 2011;Abstract 793.
Haploidentical Transplantation with Post-transplant Cyclophosphamide and Melphalan-based Conditioning– A retrospective Analysis of the First 100 Patients.
FLT3 Internal Tandem Duplication(ITD) mutation in Acute Myeloid Leukemia and its clincopathological correlation – Study from a tertiary care centre in.
Clinically useful prognostic factors in acute myeloid leukemia
Goede V et al. Proc ASH 2014;Abstract 3327.
Image 1 Detection of minimal residual disease (MRD) in consecutive bone marrow (BM) samples from a patient with relapse (A) and a patient still in remission.
New Molecular Abnormalities Recognized in AML
HS 4160 Critical Scientific Analysis
Department of Hematopathology
by David Grimwade, Adam Ivey, and Brian J. P. Huntly
Erba HP et al. Blood 2008;112: Abstract 558
Kantarjian H et al. Cancer 2009;[Epub ahead of print].
Margaret L. Gulley, Thomas C. Shea, Yuri Fedoriw 
Fenaux P et al. Lancet Oncol 2009;10(3):
Multicolor Flow Cytometry and Multigene Next-Generation Sequencing Are Complementary and Highly Predictive for Relapse in Acute Myeloid Leukemia after.
Figure S1. AML550 and AML719 arrays
Anthracycline Dose Intensification in Acute Myeloid Leukemia
European Focus on MPN and MDS 2014
Clinical Implications of Clonal Hematopoiesis
Assessment of Allogeneic HCT in Older Patients with AML and MDS: A CIBMTR Analysis McClune B et al. ASCO/ASH Symposium 2009;The Best of ASH Special & Plenary.
ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION for MULTIPLE MYELOMA
by R. Coleman Lindsley, and Benjamin L. Ebert
Qualitative and quantitative PCR monitoring of minimal residual disease (MRD) in relapsed poor-risk chronic lymphocytic leukemia (CLL): early assessment.
Grövdal M et al. Blood 2008;112:Abstract 223.
Quantifying the Survival Benefit for Allogeneic Hematopoietic Stem Cell Transplantation in Relapsed Acute Myelogenous Leukemia  Paul M. Armistead, Marcos.
The Genetic Basis for Cancer Treatment Decisions
NPM1 mutated AML can relapse with wild-type NPM1: persistent clonal hematopoiesis can drive relapse by Alexander Höllein, Manja Meggendorfer, Frank Dicker,
Pollyea DA et al. Proc ASCO 2011;Abstract 6505.
Zaja F et al. Proc ASH 2010;Abstract 966.
Emerging Importance of Mutational Analysis in Myelodysplastic Syndrome and Acute Myelogenous Leukemia  Aaron T. Gerds, Matthew J. Walter, Bart L. Scott 
On the Origin of Leukemic Species
Neoplastic disorder.
Wiestner A et al. Proc ICML 2013;Abstract 008.
NPM1 mutations define a specific subgroup of MDS and MDS/MPN patients with favorable outcomes with intensive chemotherapy by Guillermo Montalban-Bravo,
Chronic Myeloid Leukemia: MD-2025 Chisinau, Republic of Moldova
Kantarjian HM et al. Blood 2008;112:Abstract 635.
Timing for HCT Consultation
Presentation transcript:

Clearance of ‘Driver-COSMIC’ mutations post-CR1 with persisting RUNX1_L56S is unlikely to contribute towards disease progression L . Rai1, T. Boneva1, D. Brazma1, R. Dunn1, C. Grace1, E. Nacheva1,2 1 Department of OncoGenomics, HSL Analytics LLP, London UK 2 Cancer Institute, University College London, London UK Introduction Clinical significance of gene variants in AML is well established (Papaemmanuil E et al, NEJM 2016) and is increasingly being implemented into routine diagnostic algorithms. Although 80% of patients achieve morphological remission after induction chemotherapy, long-term relapse free survival is a meagre 50% (Walter RB et al, JCO 2010). Monitoring of disease kinetics, is therefore, very critical. Results summARY A median of 4 mutations per patient was observed in each clinical subgroup. Driver mutation was identified in 38 patients. Patient characteristics are described in Table 1 and examples of gene variant kinetics in the three clinical subgroups is illustrated in Figure 2. D-C mutations persisted in 85.7% of relapse patients compared to only 11% of patients in remission [P-value: 0.001, figure 3(i)]. Additionally, in intermediate risk cytogenetics group D-C mutations were retained in all 13 relapse patients while complete clearance was observed in all 6 patients with sustained remission [P-value: 0.001, Figure 3(ii)]. Eight of 9 patients who remained in morphological remission post-induction had a persistent D-C/S mutation, while all D-C disappeared. This suggests that driver mutations with both COSMIC and SNP reference may not always contribute towards disease progression. Further investigation of persisting exonic D-C/S mutations in the remission cohort revealed that 4 patients had persistent DNMT3A-p.R693H, 1 had DNMT3A-p.R693C and two had RUNX1-p.L56S 1 (Fig 4). These mutations were seen in both myeloid and lymphoid compartments of remission bone marrow samples in all 7 patients (Fig.4iii) As DNMT3A mutations are considered to occur in pre-leukemic stem cells contributing to clonal hematopoiesis (Shlush et al, Nature 2014) we studied the distribution of RUNX1 gene variants in 119 additional AML diagnostic samples. 34 patients (21%) harboured RUNX1 mutations, of which 5 had RUNX1_L56S. Finally, we evaluated kinetics of D-C in 40 MDS cases (Fig 5) of which 34 had chronic MDS and 6 had secondary AML. No significant difference was observed in the no. of patients with persistent D-C mutation in the two subgroups (P-value: 1.000). AML Cohort (Total no. of patients: 45) Clinical Outcome   Morphological remission AML relapse Refractory disease No. of patients in each clinical outcome category 19 21 5 Mean Age (Range) (in years) 55.8 (32-71) 57.3 (30-77) 63.8 (43-82) No. of patients ≤60 years 10 of 19 10 of 21 2 of 5 Time to clinical end-point(Range) (in months) 5.2 (3-15) 17.8 (3-66) 4 (3.7-22.7) Cytogenetics Good 2 1 Intermediate 16 17 Poor 3 Table 1: AML patient characteristics (i) (ii) (iii) dg AML Remission Relapse Objectives To study the kinetics of gene variants post-clinical remission (post-CR1) in patients with Acute Myeloid Leukaemia. To ascertain the prognostic relevance of gene variants, especially in intermediate cytogenetic risk group. dg AML Refractory disease Figure 2: Graph illustrating typical gene variant kinetics post CR1 in AML patients in; (i) morphological remission, (ii) relapse, (iii) refractory disease P-value: 0.001 P-value: 0.001 (ii) (i) Methods 130 follow-up samples from 45 de novo AML patients were screened for gene variants using TruSight Myeloid panel (Illumina, CA, USA) covering 54 genes with relevance in myeloid diseases. Bone marrow (BM) or peripheral blood (PB) was obtained at presentation (BM:44; PB:1) and follow-up (BM-130). All AML patients had standard induction chemotherapy followed by personalized treatment options. Gene variants kinetics was compared between patients in morphological remission (MR), relapse (R ) and refractory disease (RD). Gene variants in 95 samples from 40 MDS patients were also evaluated for progression to secondary AML. Gene variants at Variant allele frequency (VAF) of ≥10% at diagnosis and VAF of ≥1.5% during follow-up; both with target coverage of ≥300 reads were considered. Public databases-GRCh37/hg19, Catalogue of Somatic Mutations In Cancer (COSMIC), dbSNP and 1000 genome (>2%) were used to classify somatic gene variants as either [i] driver with COSMIC reference (D-C) [ii] driver with both COSMIC and SNP reference (D-C/S) P-value was generated with 2-tailed Fisher Exact (GraphPad Software, Inc, USA). Conclusions Regardless of patient age at presentation and post-induction treatment regimen followed, with the exception of allogeneic SCT; persistence of ‘D-C’ mutations post-CR1 is associated with high risk of relapse. This is especially obvious in AML with intermediate risk cytogenetics. DNMT3A-p.R693H, DNMT3A-p.R693C and RUNX1L56S are not associated with disease progression. Targeted NGS offers a cost effective method of monitoring the kinetics of somatic variants in AML with an ability to capture disease progression. Figure 3: Graph (i) compares no. of AML patients with persistent D-C mutations between clinical sub-groups. Graph (ii) compares no. of AML intermediate risk cytogenetics patients with persistent D-C mutations between clinical sub-groups (i) (ii) (iii) Gene Variant Case No. Present in Granulocytes (Y/N) Present in MNC (Y/N) DNMT3A-p.R693H 1682 Y 16125 161025 DNMT3A-p.R693C 16667 RUNX1-p.L56S 161070 161398 17140 CBL-119149011-p.? N DNMT3A-p.R693H DNMT3A-p.R693C RUNX1-p.L56S CBL-119149011-p.? (intronic, splice-region variant) References Papaemmanuil E et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N Engl J Med 2016; Jun 9;374(23):2209-21 Walter RB et al. Effect of complete remission and responses less than complete remission on survival in acute myeloid leukemia: a combined Eastern Cooperative Oncology Group, Southwest Oncology Group, and M. D. Anderson Cancer Center Study. JCO 2010; Apr 1;28(10):1766-71 Shlush LI et al. Identification of pre-leukaemic haematopoietic stem cells in acute leukaemia. Nature. 2014 Feb 20;506(7488):328-33. Figure 4: Graph (i) compares no. of AML patients with persistent D-C/S mutations between clinical sub-groups; figure (ii) lists the D-C/S mutations in the remission sub-group that persist after CR1; Table (iii) demonstrates presence of a gene variant in the granulocyte and/or mononuclear compartment of bone marrow samples from AML patients in remission P-value: 1.0000 CONTACT INFORMATION lena.rai@nhs.net; e.nacheva@ucl.ac.uk Figure 5: Graph compares no. of patients with persistent D-C mutations between those with chronic MDS and those with secondary AML Figure1: Graph illustrating no. of diagnostic and follow-up samples from de novo AML and MDS patients included in this study