Clinical Overview on Measurable Residual Disease (MRD) in Acute Myeloid Leukemia(AML)

Similar documents
Molecularly Targeted Therapies - Strategies of the AMLSG

Impact of Biomarkers in the Management of Patients with Acute Myeloid Leukemia

Measurable/Minimal Residual Disease in Patients with Acute Myeloid Leukemia Undergoing Transplantation

Meeting VAKB 8 februari 2011 Nancy Boeckx, MD, PhD

Minimal residual disease (MRD) in AML; coming of age. Dr. Mehmet Yılmaz Gaziantep University Medical School Sahinbey Education and Research hospital

Minimal Residual Disease as a Surrogate Endpoint in Acute Myeloid Leukemia Clinical Trials

Tools for MRD in AML: flow cytometry

All patients with FLT3 mutant AML should receive midostaurin-based induction therapy. Not so fast!

Risk-adapted therapy of AML in younger adults. Sergio Amadori Tor Vergata University Hospital Rome

Leukemia. Andre C. Schuh. Princess Margaret Cancer Centre Toronto

ESTABLISHED AND EMERGING THERAPIES FOR ACUTE MYELOID LEUKAEMIA. Dr Rob Sellar UCL Cancer Institute, London, UK

Which is the best treatment for relapsed APL?

MRD detection in AML. Adriano Venditti Hematology Fondazione Policlinico Tor Vergata Rome

Acute Myeloid Leukemia

Acute Myeloid Leukemia Progress at last

Corporate Medical Policy. Policy Effective February 23, 2018

Molecular Markers in Acute Leukemia. Dr Muhd Zanapiah Zakaria Hospital Ampang

Supplementary Appendix

AML: WHO classification, biology and prognosis. Dimitri Breems, MD, PhD Internist-Hematoloog Ziekenhuis Netwerk Antwerpen

Treatments and Current Research in Leukemia. Richard A. Larson, MD University of Chicago

5/21/2018. Disclosures. Objectives. Normal blood cells production. Bone marrow failure syndromes. Story of DNA

Review Minimal Residual Disease in Acute Myeloid Leukemia: Still a Work in Progress?

Evolving Targeted Management of Acute Myeloid Leukemia

CARE at ASH 2014 Leukemia. Julie Bergeron, MD Maisonneuve-Rosemont Hospital

Neue zielgerichtete Behandlungsoptionen der neu diagnostizierten FLT3-positiven Akuten Myeloischen Leukämie (AML)

Acute myeloid leukemia: prognosis and treatment. Dimitri A. Breems, MD, PhD Internist-Hematoloog Ziekenhuis Netwerk Antwerpen Campus Stuivenberg

Remission induction in acute myeloid leukemia

The Past, Present, and Future of Acute Myeloid Leukemia

Acute Myeloid and Lymphoid Leukemias

FLT-3 inhibitors in AML

How the Treatment of Acute Myeloid Leukemia is Changing in 2019

Examining Genetics and Genomics of Acute Myeloid Leukemia in 2017

Personalized Therapy for Acute Myeloid Leukemia. Patrick Stiff MD Loyola University Medical Center

Acute Myeloid Leukemia

Summary of Key AML Abstracts Presented at the American Society of Hematology (ASH) December 2-6, San Diego CA

New drugs in Acute Leukemia. Cristina Papayannidis, MD, PhD University of Bologna

MRD in AML: does it already guide therapy decision-making?

7th International Symposium on Acute Promyelocytic Leukemia, Rome, September 24 27, Eva Lengfelder

Objectives. I do not have anything to disclose.

Jordi Esteve Hospital Clínic (Barcelona) Acute Leukemia Working Party. The European Group for Blood and Marrow Transplantation

Evaluating measurable residual disease in acute myeloid leukemia Ravandi, Farhad; Walter, Roland B; Freeman, Sylvie D

In t(8;21)(q22;q22) acute myeloid leukemia, the prognostic value of

Acute leukemia and myelodysplastic syndromes

Kevin Kelly, MD, Phd Acute Myeloid and Lymphoid Leukemias

Concomitant WT1 mutations predicted poor prognosis in CEBPA double-mutated acute myeloid leukemia

Future Targets for Acute Myeloid Leukemia

New concepts in the management of elderly patients with AML

Update on the WHO Classification of Acute Myeloid Leukemia. Kaaren K. Reichard, MD Mayo Clinic Rochester

Acute Leukemia. Sebastian Giebel. Geneva 03/04/

Current Techniques in Molecular Biology Friedel Nollet, Ph.D.

AML Emerging Treatment Strategies

Acute promyelocytic leukemia

Acute myeloid leukemia. M. Kaźmierczak 2016

Frontline Induc.on Therapy in 2017 Alan K Burne+

Cautionary Note Regarding Forward-Looking Statements

GENETIC TESTING FOR FLT3, NPM1 AND CEBPA VARIANTS IN CYTOGENETICALLY NORMAL ACUTE MYELOID LEUKEMIA

Trapianto allogenico convenzionale

Acute Myeloid Leukemia with Recurrent Cytogenetic Abnormalities

Anthracycline dose intensification improves molecular response and outcome of patients treated for core binding factor acute myeloid leukemia

N Engl J Med Volume 373(12): September 17, 2015

ACUTE LEUKEMIA. Zwi N. Berneman University of Antwerp & Antwerp University Hospital. 16th Post-ASH Meeting Zaventem, 10 January 2014

ARTICLES. Introduction

Initial Diagnostic Workup of Acute Leukemia

Acute Myeloid Leukemia: State of the Art in 2018

Regulatory Aspects - AML & CLL

Actinium Pharmaceuticals, Inc.

Meet-the-Expert: AML Treating older patients with AML

National Institute for Health and Care Excellence. Single Technology Appraisal (STA)

Role of consolidation therapy in Multiple Myeloma. Pieter Sonneveld. Erasmus MC Cancer Institute Rotterdam The Netherlands

Chronic Myeloid Leukemia A Disease of Young at Heart but Not of Body

Changing AML Outcomes via Personalized Medicine: Transforming Cancer Management with Genetic Insight

MOLECULAR AND CLINICAL ONCOLOGY 3: , 2015

PDF of Trial CTRI Website URL -

Belgium recommendations for the management of acute promyelocytic leukaemia

Stem cell transplantation for patients with AML in Republic of Macedonia: - 15 years of experience -

Characteristics and Outcome of Therapy-Related Acute Promyelocytic Leukemia After Different Front-line Therapies

Acute Myeloid Leukemia with RUNX1 and Several Co-mutations

Treatment of APL. M a tth e w M e i, M.D. A s s is ta n t P ro fe s s o r C ity o f H o p e C o m p re h e n s iv e C a n c e r C e n te r

Acute myeloid leukemia: a comprehensive review and 2016 update

Diagnostic Molecular Pathology of Myeloid Neoplasms

Dana-Farber Cancer Institute, Boston, MA; 2 City of Hope National Medical Center, Duarte, CA; 3 Roswell Park Cancer Institute, Buffalo, NY; 4

Cytogenetic heterogeneity negatively impacts outcomes in patients with acute myeloid leukemia

Single Technology Appraisal (STA) Midostaurin for untreated acute myeloid leukaemia

Consolidation and maintenance therapy for transplant eligible myeloma patients

Progress in the treatment of acute promyelocytic leukemia. Lionel Adès, MD PhD Hopital Saint Louis, Paris Diderot University

2/10/2017. Updates in Acute Leukemia Therapy Blood Cancer Incidence in the United States, Leukemia Incidence in the Unites States, 2016

Standard risk ALL (and its exceptions

SUPPLEMENTARY FIG. S3. Kaplan Meier survival analysis followed with log-rank test of de novo acute myeloid leukemia patients selected by age <60, IA

Update on Optimal Management of Acute Myeloid Leukemia

Improvements with Risk-adapted PETHEMA Protocols in New Diagnosed Acute Promyelocytic Leukemia

ANCO Hematological Malignancies Update: The year in review. Midostaurin Vyxeos Gemtuzumab ozogamicin Enasidenib. The year in preview

Therapy-related MDS/AML with KMT2A (MLL) Rearrangement Following Therapy for APL Case 0328

ANCO 2015: Treatment advances in acute leukemia

AIH, Marseille 30/09/06

Post-remission therapy for acute myeloid leukemia

Reporting cytogenetics Can it make sense? Daniel Weisdorf MD University of Minnesota

Myelodysplasia/Myeloproliferative Neoplasms (MDS/MPN) Post-HCT Data

Subset Specific Therapy in High Risk Myeloid Malignancies. Are We Making Progress? Olatoyosi Odenike, MD. The University of Chicago

Medical Policy. MP Hematopoietic Cell Transplantation for Acute Myeloid Leukemia

Transcription:

Clinical Overview on Measurable Residual Disease (MRD) in Acute Myeloid Leukemia(AML) Konstanze Döhner Department of Internal Medicine III, University Hospital of Ulm, Ulm Germany

DISCLOSURES OF COMMERCIAL SUPPORT Konstanze Döhner Name of Company Research support Employee Honoraria Stockholder Speaker s Bureau Advisory Board Novartis Janssen Celgene Daiichi Sankyo CTI BioPharma Roche

Measurable residual disease in AML Achievement of complete remission (CR) is the most important prerequisite for cure and long-term survival of patients with acute myeloid leukemia (AML) The increasing number of new molecular markers and the development of novel technologies [real-time quantitative polymerase chain reaction (RQ- PCR), multi-color flow cytometry, digital polymerase chain reaction (dpcr), next-generation sequencing (NGS)] allow to determine measurable residual disease (MRD) with high sensitivity MRD allows to refine our current definition of morphological CR New response category proposed by the 2017 ELN recommendations: Complete remission without MRD (CR MRD- )

Measurable residual disease in AML MRD monitoring: clinical implications Impact on prognosis Early detection of relapse Guiding pre-emptive therapy Treatment decision making, in particular within the context of postremission therapy [e.g.allogeneic stem cell transplantation (allosct] Monitoring of treatment effects (novel drugs) MRD as a surrogate endpoint in clinical trials > rapid approval of novel drugs

Methods for MRD monitoring Leukemia associated immunophenotype (LAIP) CD7 Multicolor flow cytometry (MCF) CD117 CD38 CD33 CD45 s CD14 CD56 CD34 PCR-based techniques Quantitative RT-PCR (RQ-PCR) Digital PCR (dpcr), droplet digital PCR (ddpcr) NGS-based techniques Mainly targeted approaches Quantification / identification of multiple gene mutations Sensitivity 10-3 to 10-4 Sensitivity 10-5 to 10-6 Sensitivity 10-4

Molecular markers used for RQ-PCR based MRD monitoring in AML So far, MRD monitoring in AML has been restricted to distinct AML subtypes mainly characterized by gene fusions resulting from translocations/inversions or by hot spot mutations PML/RARA RUNX1/RUNXT1 CBFB/MYH11 BCR/ABL (KMT2A/MLLT3) NPM1 ~ 50% of all AML RUNX1/RUNXT1 (~5%) CBFB/MYH11 (~5-8%) PML/RARA (5-8%) NPM1 mutations (~30%) BCR/ABL <1% KMT2A /MLLT3 (2%) t(11q23) (2%)

MRD monitoring in clinical AML trials - important issues - Most, if not all studies published so far are retrospective and MRD was not included as a primary or secondary endpoint Studies were performed on heterogeneous patient populations with respect to age, treatment, cohort size, or type of material MRD monitoring has not been standardized yet; existence of different MRD assays with distinct sensitivities and definitions for MRD negativity Studies are not comparable with regard to cut-off values / values for transcript levels / copy numbers However, in most studies, achievement of MRD-negativity / RQ-PCR-negativity after two cycles of therapy and/or at the end of treatment was significantly associated with outcome

Prognostic impact of MRD in APL Acute Promyelocytic Leukemia Grimwade D et al. J Clin Oncol 2009; 27(22): 3650-3658 Prospective study on 406 newly diagnosed adult APL pts (MRC AML15 trial) Study design for MRD monitoring Material: paired BM and PB samples used for RQ-PCR analysis Time points: - after each treatment course - paired PB and BM samples obtained every 3 months until 36 months of post-consolidation Quality control: sensitivity of at least 1 in 10 4 Transport: samples were sent by courier / overnight delivery Report: Clinicians were informed of PCR results

Prognostic impact of MRD in APL Acute Promyelocytic Leukemia Grimwade D et al. J Clin Oncol 2009; 27(22): 3650-3658 6.727 serial BM/PB samples (2.276 paired samples) were analyzed by RQ-PCR At the end of treatment achievement of RQ- PCR-negativity was highly predictive for clinical relapse and relapse-free survival (RFS) Persistent PCR positivity and molecular relapse were significantly associated with clinical relapse and RFS Pre-emptive therapy with arsenic trioxide in pts with persistent PCR positivity or molecular relapse prevented progression to overt relapse in the majority of the pts CIR in patients treated with pre-emptive therapy (blue)

Prognostic impact of MRD in Core-binding Factor (CBF) Leukemia t(8;21)(q22;q22.1); inv(16)(p13.1q22) MRD-negativity at the end of treatment in PB impacts clinical outcome French Intergroup CBF-2006 trial. Willekens et al., Haematologica 2016, [t(8;21), n=94] MRD-negativity at end of treatment in BM impacts clinical outcome AML Study Group Agrawal et al., ASH meeting 2016, abstract #1207 [t(8;21), n=120]) Transcript level reduction (3-log) before consolidation II influences relapse risk French Intergroup. Jourdan et al., Blood 2013, [t(8;21), n=96; inv(16), n=102] Distinct absolute transcript levels and log reduction after induction I and during follow-up correlate with clinically relevant endpoints UK MRC15. Yin et al., Blood 2012, [t(8;21), n=163; inv(16), n=115] Minimal residual disease monitoring and mutational landscape in AML with RUNX1- RUNX1T1: a study on 134 patients. Höllein et al., Leukemia 2018

Prognostic impact of RUNX1/RUNX1T1 MRD in AML Hong-Hu Zhu et al. Blood 2013; 121: 4056-4062 Prospective study on 116 newly diagnosed pts with t(8;21)-positive AML achieving CR after 2 induction cyles MRD directed risk stratification treatment in pts in 1.CR Study design for MRD monitoring Material: BM samples were used for RQ-PCR analysis Time points: - at diagnosis - after induction therapy - after each consolidation cycle - 3-monthly for 1 year Major molecular remission (MMR): > 3 log reduction (<0.4%) in RUNX1/RUNX1T1 transcripts compared to pretreatment sample Loss of MMR: RUNX1/RUNX1T1 transcript levels > 0.4% in MMR pts

Prognostic impact of RUNX1/RUNX1T1 MRD in AML MRD directed risk stratification treatment in t(8;21)-positive AML in 1.CR: The AML05 multicenter trial Study design n=18 n=47 n=29 CT/autoSCT n=69 n=40 n=29 Hong-Hu Zhu et al. Blood 2013; 121: 4056-4062

Prognostic impact of RUNX1/RUNX1T1 MRD status in AML MRD directed risk stratification treatment in t(8;21)-positive AML in 1.CR: The AML05 multicenter trial Hong-Hu Zhu et al. Blood 2013; 121: 4056-4062

Phase III study of chemotherapy with or without Dasatinib in CBF AML: AMLSG 21-13 Study Induction Consolidation x3 Maintenance CBF mutation screening within 48 hours R Daunorubicin Cytarabine n=204 Daunorubicin Cytarabine +Dasatinib High-Dose Cytarabine* MRD assessment by RQ-PCR High-Dose Cytarabine* +Dasatinib 1-monthly PB for 6 months; 3 monthly PB and BM 1-yr Dasatinib All adult patients eligible for intensive therapy, no upper age limit * Cytarabine: 18-60yrs: 3g/m 2, q12hr, d1-3; >60yrs: 1g/m 2, q12hr, d1-3 ClinicalTrials.gov NCT02013648 Salvage / transplantation if MRD persists or recurs

log(10)-reduction -8-6 -4-2 0 Kinetics of RUNX1/RUNXT1 transcript levels: AMLSG 11-08 cohort versus historical control - t(8;21) p=0.26 p=0.55 p=0.66 p=0.02 p=0.24 p=0.02 p=0.28 p=0.52 p=0.27 historical control AMLSG 11-08 Maintenance / Follow-up Cycle I Cycle II Cycle III Cycle IV Cycle V 3 M 6 M 9 M 12 M N 34 28 34 25 35 24 33 20 26 17 27 15 23 11 16 11 16 10 Median 1861 680 62 26 28 12 32 0 8 0 21 0 17 0 0 0 0 0 Negative, n 1 1 3 7 9 9 5 12 12 11 9 8 11 9 9 11 10 10 Negative % 3.0 3.6 8.8 28.0 25.7 37.5 15.2 60.0 46.2 35.3 66.7 46.7 47.8 81.2 56.3 100 62.5 100 Paschka P et al., Leukemia 2018

Prognostic impact of MRD in NPM1 mutated AML MRD levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML. Schnittger et al., Blood 2009;114:2220-31; [n=252] MRD monitoring in NPM1 mutated AML: a study from the German-Austrian Acute Myeloid Leukemia Study Group. Krönke et al., JCO 2011;19:2709-2716; [n=245] The level of residual disease based on mutant NPM1 is an independent prognostic factor for relapse and survival in AML. Shayegi et al., Blood 2013;122:83-92; [n=155] MRD assessed by WT1 and NPM1 transcript levels identifies distinct outcomes in AML patients and is influenced by gemtuzumab ozogamicin. Lambert et al., Oncotarget 2014; 5:6280-8; [n=77] Assessment of minimal residual disease in standard-risk AML. Ivey et al., N Engl J Med 2016; 374(5):422-33; [n=437]

Prognostic impact of MRD in NPM1 mutated AML Retrospective study on 437 NPM1 mutated AML pts (pediatric and adults, NCRI AML17 trial) 2569 BM/PB (902/1667) samples were analyzed by RQ-PCR after each treatment cycle and during followup; sensitivity 10-5 MRD positivity in PB after 2 cycles of therapy was significantly associated with inferior OS (24% vs 73%) and higher risk of relapse (82% vs 30%) after 3 years In multivariate analysis MRD positivity in PB was significantly associated with death (HR 4.38) and relapse (HR 5.09) Ivey A et al. N Engl J Med 2016

Prognostic impact of MRD in NPM1 mutated AML Impact of concurrent FLT3-ITD mutation Relapse in pts without FLT3-ITD Relapse in pts with FLT3-ITD Ivey A et al. N Engl J Med 2016

Prognostic impact of MRD in NPM1 mutated AML Impact of concurrent DNMT3A mut Relapse in pts without DNMT3A mut Relapse in pts with DNMT3A mut Ivey A et al. N Engl J Med 2016

Prognostic impact of MRD in NPM1 mutated AML - A Study of the German-Austrian AML Study Group (AMLSG) - Retrospective/since 2008 prospective study on 611 NPM1 mut adult AML pts enrolled in one of 4 AMLSG treatment trials; median follow-up for all patients/trials: 3.2 years 6339 BM/PB (3527/2812) samples were analyzed by RQ-PCR after each treatment cycle and during follow-up; sensitivity 10-5 to 10-6 Achievement of RQ-PCR negativity in the BM after 2 treatment cycles was significantly associated with superior OS (at 4 yrs: 82% vs 63%) and lower CIR (at 4 yrs: 15% vs 40%) compared to RQ-PCR positive pts Multivariate analysis: NPM1 mut transcript levels (continuous variable) in BM were significantly associated with relapse (HR 1.87) and OS (HR 1.44) Krönke et al., JCO 2011;19:2709-2716; Kapp-Schwoerer S et al., ASH meeting 2017, #183 Cumulative incidence Overall Survival 0.0 0.2 0.4 0.6 0.8 1.0 1 0.8 0.6 0.4 0.2 0 RQ-PCR negativity, n=63 RQ-PCR positivity, n=332 0 1 2 3 4 5 6 7 8 9 10 11 0 >0 Time (years) P = 0.01 RQ-PCR positivity, n=328 RQ-PCR negativity, n=59 0 2 4 6 8 10 Time (years) P < 0.0001

Impact of concurrent FLT3-ITD/DNMT3A mutations on kinetics of NPM1 mut transcript levels DNMT3A neg + FLT3 neg DNMT3A pos + FLT3 pos log(10)-npm1mut transcript level -2 0 2 4 6 8 p=0.81 p=0.20 p<0.0001 p=0.0005 p<0.0001 p=0.0002 Diagnosis Cycle I Cycle II Cycle III Cycle IV Cycle V Cycle VI Samples, n 140 111 95 70 114 71 98 43 92 19 83 16 1 0 Kapp-Schwoerer S et al., ASH meeting 2017, #183 Median 66054 7 67083 2 102 2 133 0 17 108 5 55 1 54 0 36 11 --- Negative, n 0 0 7 1 30 6 39 10 42 3 43 3 0 --- Negative % 0.0 0.0 7.4 1.4 26.3 8.5 39.8 23.3 45.7 15.8 51.8 18.8 0-0 ---

Achievement of RQ-PCR negativity in NPM1 mut patients according to FLT3-ITD/DNMT3A mutation status in BM After 2 cycles of therapy NPM1 mut NPM1 mut NPM1 mut NPM1 mut Genotype FLT3-ITD WT FLT3-ITD mut FLT3-ITD WT FLT3-ITD mut DNMT3A WT DNMT3A WT DNMT3A mut DNMT3 mut RQ-PCR negative (n) 30 (26%) 14 (25%) 10 (8%) 6 (8%) RQ-PCR positive (n) 84 (74%) 41 (75%) 110 (92%) 65 (92%) % negative 26% 25% 8% 8% P=0.0002 Kapp-Schwoerer S et al., unpublished data

Impact of MRD-negativity in BM after two cycles of intensive treatment on type of post-remission therapy Overall survival Cumulative incidence of relapse Overall Survival 1 0.8 0.6 0.4 0.2 0 HIDAC, n=43 AlloSCT, n=19 P<0.00001 Cumulative incidence 0.0 0.2 0.4 0.6 0.8 1.0 ALLO CHEMO HIDAC, n=41 AlloSCT, n=17 P=0.18 0 1 2 3 4 5 6 7 8 9 10 11 Time (years) 0 2 4 6 8 10 Time (years) Kapp-Schwoerer S et al., EHA 2018, PS973

Phase III Study of Chemotherapy in Combination with ATRA with or without Gemtuzumab Ozogamicin (GO) in NPM1 mut AML patients [AMLSG 09-09 Trial] Induction x2 Consolidation 1 Consolidation 2+3 NPM1 Mutation Screening Within 48 Hours R n = 588 ATRA-ICE ATRA-ICE +Gemtuzumab Ozogamicin (GO) ATRA Cytarabine* ATRA Cytarabine* +GO ATRA Cytarabine* NPM1 MRD monitoring by RQ-PCR ATRA Cytarabine* *Salvage / transplantation if MRD persists or recurs All adult patients eligible for intensive therapy, no upper age limit * Cytarabine: 18-60yrs: 3g/m 2, q12hr, d1-3; >60yrs: 1g/m 2, q12hr, d1-3 PI: H. Döhner; supported by Else Kröner-Fresenius-Foundation & Pfizer

Impact of GO on kinetics of NPM1 mut transcript levels log(10)-npm1 mut TL -2 0 2 4 6 8 p=0.75 p=0.001 p=0.008 p<0.001 p=0.006 p=0.009 Arm A: Standard Arm B: Investigational Diagnosis Induction I Induction II Consolidation I Consolidation II Consolidation III Treatment Cycle Kapp-Schwoerer S et al., oral presentation ASH 2018, #991

Impact of GO on MRD at the end of treatment BM samples (n=288); all pts in CR CIR 0.0 0.2 0.4 0.6 0.8 1.0 Arm A, n= 154 p= 0.04 Arm B, n= 134 Achievement of RQ-PCR negativity in BM RQ-PCR neg; n (%) Arm A Arm B p 63/154 (41%) 74/134 (55%) 0.02 0 1 2 3 4 5 6 7 Time (yrs) Kapp-Schwoerer S et al., oral presentation ASH 2018, #991

How to implement MRD detection in clinical AML trials Lambert J et al. Oncotarget 2014; 5(15):6280-8 Evaluation of the prognostic significance of MRD levels in adult AML pts treated in the randomized gemtuzumab ozogamicin (GO) ALFA-0701 trial 79 pts with monitorable NPM1 mut (GO-arm n=42, control arm n=37) BM/PB samples at diagnosis, after induction therapy and after each consolidation course 61 pts achieved CR and were subsequently tested for MRD (GO-arm n=33, control arm n=28) cdna based RQ-PCR assay MRD positivity: > 0.1% in BM sample

How to implement MRD detection in clinical AML trials Lambert J et al. Oncotarget 2014; 5(15):6280-8 CIR after double induction CIR at the end of treatment Effect of GO on MRD p=0.028 p=0.006

MRD monitoring by next generation sequencing (NGS) 482 AML pts (18 to 65 years) treated with 1 to 2 cycles of standard induction chemotherapy followed by consolidation in HOVON-SAKK clinical trials Mutation detection at diagnosis and in CR NGS panel of 54 genes (Illumina) at diagnosis and in BM in morphological CR after completion of induction therapy 430/482 (89.2%) pts had somatic driver mutations at diagnosis (2.9 mutations per case) 51.4% of pts had persisting mutations in BM in morphological CR at highly variable variant allele frequencies (VAF 0.02-47%), predominantly in DNMT3A (78.7%), TET2 (54.2%) and ASXL1 (51.6%) >> DTA mutations Illumina TruSight Myeloid Panel; mean coverage ~3500x Mojca Jongen-Lavrencic et al., N Engl J Med; 2018

MRD monitoring by next generation sequencing (NGS) DTA mutations were not associated with the incidence of relapse at any VAF cut-off >> stage of clonal hematopoiesis rather than impending relapse After exclusion of persistent DTA mutations, NGS MRD was significantly associated with higher relapse rate (55.4% vs 31.9%), lower RFS (36,6% vs 58.1%) and inferior OS (41.9% vs 66.1%) than no detection Persistence on non-dta mutations revealed as an independent prognostic variable in multivariate analysis for relapse (HR 1.89), RFS (HR 1.64) and OS (HR 1.64) non-dta mutations present in CR non-dta mutations absent in CR non-dta mutations absent in CR non-dta mutations present in CR Mojca Jongen-Lavrencic et al., N Engl J Med; 2018

MRD monitoring by next generation sequencing (NGS) Relapse incidence of residual leukemia: NGS MRD and multiparameter flow MRD both were significantly associated with relapse in AML flow MRD - + NGS MRD - + 205 64 41 30 Multivariate analysis: the combined use of the two assays conferred independent prognostic value with respect to RFS and OS

MRD monitoring by next generation sequencing (NGS) 131 AML pts (39 to 55 years) with intensive induction therapy and achievement of morphologic CR at day 30 Detection of mutations in paired BM samples (Dx/CR) by targeted capture deep sequencing (coverage at DX 257 x/ in CR 575 x) Definition of three levels of mutation clearance (MC) of residual mutations in CR on the basis of VAF: MC2.5: VAF < 2.5% MC1.0: VAF < 1% CMC: complete mutation clearance Correlation of mutation clearance with clinical endpoints (EFS, OS, CIR) Morita K et al., J Clin Oncol; 2018

MRD monitoring by next generation sequencing (NGS) MC2.5 MC1.0 CMC MC2.5 MC1.0 CMC Morita K et al., J Clin Oncol; 2018

MRD monitoring by next generation sequencing (NGS) Prognostic impact of allogeneic stem cell transplantation in 1.CR Pts without CMC Pts with CMC Morita K et al., J Clin Oncol; 2018

ELN recommendations for MRD assessment Suitable markers for MRD: NPM1, RUNX1-RUNXT1, CBFB-MYH11, PML-RARA; DNMT3A, ASXL1 or TET2 are NOT usable Relevant MRD time points: after 2 treatment cycles, at the end of treatment (EOT); 3 monthly for 24 months after EOT Definitions for molecular remission, molecular progression and molecular relapse Molecular and/or MFC MRD should be integrated into all clinical trials at all times of evaluation of response, using the technical ELN guidelines Schuurhuis GJ et al., Blood 2018;131:1275-1291 Schuurhuis GJ et al., Blood 2018;131:1275-1291

The U.S. Food and Drug Administration (FDA) recently issued a draft guidance titled Hematologic Malignancies: Regulatory Considerations for Use of Minimal Residual Disease (MRD) in Development of Drug an Biologic Products for Treatment

Midostaurin vs Gilteritinib plus chemotherapy for AML with FLT3 mutation HOVON 156 / AMLSG 28-18 Induction I Induction II Consolidation 1-yr Maintenance 18 yrs AML with FLT3 mutation R NGS gene panel MFC (LAIP) Dauno Cytarabine Midostaurin n=768 Dauno Cytarabine Gilteritinib Dauno IDAC Midostaurin Dauno IDAC Gilteritinib MRD assessment IDAC Midostaurin ME / auto HCT Midostaurin IDAC Gilteritinib ME / auto HCT Gilteritinib IDAC Midostaurin IDAC Gilteritinib IDAC Midostaurin IDAC Gilteritinib Midostaurin Gilteritinib Assessment of measurable residual disease (MRD) by RT-qPCR (NPM1, CBF), next-generation sequencing (NGS), and multiparameter flow cytometry (MFC) of leukemia-associated immunophenotypes (LAIP) Centralized diagnostic assessment (Ulm, Rotterdam, Amsterdam) harmonized assays CR MRD- as secondary endpoint HCT, hematopoietic cell transplantation; IDAC, intermediate-dose cytarabine; ME, mitoxantrone, etoposide

Summary and Conclusions I Most of the studies were performed retrospectively >>> patient selection according to the presence of a molecular marker, the availability of a BM/PB sample at defined time points, and the CR status (1. CR) Achievement of MRD/RQ-PCR-negativity was evaluated after 2 cycles of therapy or at the end of treatment in patients in 1.CR Here, achievement of MRD-negativity or significant reduction of transcript levels /mutations by RQ-PCR/NGS was associated with reduced relapse risk and improved survival However, a significant proportion of patients do not achieve MRD-negativity in BM at this early time point in particular when highly sensitive assays are used

Summary and Conclusions II MRD kinetics allow monitoring of treatment effects NGS-based MRD monitoring has been shown to be useful in ~ 90% of AML patients; further development of the techniques is ongoing; sensitivities are still low and data analysis is challenging Standardization/harmonization guidelines for MRD MRD monitoring (molecular / MCF) should be included in all clinical trials

S. Cocciardi A. Corbacioglu J. Herzig S. Kapp-Schwoerer J. Krönke N. Jahn E. Panina L. Schmalbrock D. Späth F. Theis F. Stegelmann V. Teleanu V. Gaidzik P. Paschka F. Rücker H. Döhner Ulm University L. Bullinger A. Dolnik T. Blätte Charité, Universitätsmedizin Berlin SFB 1074 Experimental Models and Clinical Translation in Leukemia M. Heuser G. Göhring F. Thol B. Schlegelberger A. Ganser MHH, Hannover