Objectives DISCLOSURES. Next-Generation Sequencing (NGS) Testing for Hematologic Malignancies

Similar documents
Illumina Trusight Myeloid Panel validation A R FHAN R A FIQ

Next Generation Sequencing in Haematological Malignancy: A European Perspective. Wolfgang Kern, Munich Leukemia Laboratory

Please Silence Your Cell Phones. Thank You

Laboratory Service Report

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

NeoTYPE Cancer Profiles

NeoTYPE Cancer Profiles

The Center for PERSONALIZED DIAGNOSTICS

Laboratory Service Report

Molecular. Oncology & Pathology. Diagnostic, Prognostic, Therapeutic, and Predisposition Tests in Precision Medicine. Liquid Biopsy.

Out-Patient Billing CPT Codes

Introduction of an NGS gene panel into the Haemato-Oncology MPN service

ADRL Advanced Diagnostics Research Laboratory

DISCLOSURE Luca Malcovati, MD. No financial relationships to disclose

Overview. Methods 9/11/2017. Next Generation Sequencing and Precision Medicine in Hematological Malignancies. Genotyping in hematology

Pathogenesis and management of CMML

The Challenges of Precision Medicine: New Advances in Molecular Diagnostic Testing- Impact for Healthcare

Molecular Advances in Hematopathology

Examining Genetics and Genomics of Acute Myeloid Leukemia in 2017

Acute Myeloid Leukemia with RUNX1 and Several Co-mutations

Clonal Evolution of saml. Johnnie J. Orozco Hematology Fellows Conference May 11, 2012

Management of Myelodysplastic Syndromes

Next generation sequencing analysis - A UK perspective. Nicholas Lea

August 17, Dear Valued Client:

Opportunities for Optimal Testing in the Myeloproliferative Neoplasms. Curtis A. Hanson, MD

Blastic Plasmacytoid Dendritic Cell Neoplasm with DNMT3A and TET2 mutations (SH )

Acute Myeloid Leukemia Progress at last

Myelodysplastic syndromes

Session II: Summary. Robert P Hasserjian, MD Professor of Pathology Massachusetts General Hospital and Harvard Medical School

ACCME/Disclosures. History. Hematopathology Specialty Conference Case #4 4/13/2016

Chronic Myelomonocytic Leukemia with molecular abnormalities SH

Molecular profiling in confirming the diagnosis of early myelodysplastic syndrome

West Midlands Regional Genetics Laboratory

SUPPLEMENTARY INFORMATION

Corporate Medical Policy. Policy Effective February 23, 2018

Molecular Markers. Marcie Riches, MD, MS Associate Professor University of North Carolina Scientific Director, Infection and Immune Reconstitution WC

Update on Myelodysplastic Syndromes and Myeloproliferative Neoplasms. Kaaren Reichard Mayo Clinic Rochester

Acute leukemia and myelodysplastic syndromes

Published Ahead of Print on April 14, 2016, as doi: /haematol Copyright 2016 Ferrata Storti Foundation.

Myelodysplastic syndromes Impact of Biology. Lionel Adès Hopital Saint Louis Groupe Francophone des SMD. Épidémiologie

Enhancing Assessment of Myeloid Disorders in the Era of Precision Medicine

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

Myelodysplastic syndromes and the new WHO 2016 classification

Targeted NGS in oncology and hemato-oncology using in-house designed gene panels. Joni Van der Meulen Molecular Diagnostics UZ Ghent (MDG) 24/03/2017

Session 4: Summary and Conclusions

Juan Ma 1, Jennifer Dunlap 2, Lisong Shen 1, Guang Fan 2 1

Fluxion Biosciences and Swift Biosciences Somatic variant detection from liquid biopsy samples using targeted NGS

Kevin Kelly, MD, Phd Acute Myeloid and Lymphoid Leukemias

SWOG ONCOLOGY RESEARCH PROFESSIONAL (ORP) MANUAL LEUKEMIA FORMS CHAPTER 16A REVISED: DECEMBER 2017

Diagnostic Approach for Eosinophilia and Mastocytosis. Curtis A. Hanson, M.D.

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

Objectives. Morphology and IHC. Flow and Cyto FISH. Testing for Heme Malignancies 3/20/2013

Beyond the CBC Report: Extended Laboratory Testing in the Evaluation for Hematologic Neoplasia Disclosure

Evolving Targeted Management of Acute Myeloid Leukemia

Supplemental Material. The new provisional WHO entity RUNX1 mutated AML shows specific genetics without prognostic influence of dysplasia

SureSelect Cancer All-In-One Custom and Catalog NGS Assays

Patricia Aoun MD, MPH Professor and Vice-Chair for Clinical Affairs Medical Director, Clinical Laboratories Department of Pathology City of Hope

Disclosure: Objectives/Outline. Leukemia: Genealogy of Pathology Practice: Old Diseases New Expectations. Nothing to disclose.

Precision Medicine and Molecular Testing.

SESSION 1 Reactive cytopenia and dysplasia

Diagnostic Molecular Pathology of Myeloid Neoplasms

School of Pathology and Laboratory Medicine: Current and New Research Interests

Cost-Effective Strategies in the Workup of Hematologic Neoplasm. Karl S. Theil, Claudiu V. Cotta Cleveland Clinic

Case #1. 65 yo man with no prior history presented with leukocytosis and circulating blasts: Bone marrow biopsy was performed

The Evolving Role of Transplantation for MPN

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

Mutational Impact on Diagnostic and Prognostic Evaluation of MDS

Disclosures for Ayalew Tefferi

TEST MENU TEST CPT CODES TAT. Chromosome Analysis Bone Marrow x 2, 88264, x 3, Days

Next Generation Sequencing in Clinical Practice: Impact on Therapeutic Decision Making

APPROACH TO MYELODYSPLASTIC SYNDROMES IN THE ERA OF PRECISION MEDICINE

Session 5. Pre-malignant clonal hematopoietic proliferations. Chairs: Frank Kuo and Valentina Nardi

Enhancing Assessment of Myeloid Leukemia in the Era of Precision Medicine

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

Should Mutational Status in Primary Myelofibrosis (PMF) Guide Therapy..YES!!!

Disclosures for Ayalew Tefferi

Deep Learning Analytics for Predicting Prognosis of Acute Myeloid Leukemia with Cytogenetics, Age, and Mutations

Myelodysplastic syndromes: revised WHO classification and distinction from non-neoplastic conditions

Myelodysplastic Syndromes. Post-ASH meeting 2014 Marie-Christiane Vekemans

Genomic Medicine: What every pathologist needs to know

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

SUPPLEMENTAL APPENDIX METZELER ET AL.: SPECTRUM AND PROGNOSTIC RELEVANCE OF DRIVER GENE MUTATIONS IN ACUTE MYELOID LEUKEMIA

Blast transformation in chronic myelomonocytic leukemia: Risk factors, genetic features, survival, and treatment outcome

Myelodysplastic syndromes: 2018 update on diagnosis, risk-stratification and management

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

Enhancing Assessment of Myeloid Leukemia in the Era of Precision Medicine

9/25/2017. Disclosure. I have nothing to disclose. Young S. Kim MD Dept. of Pathology

Disclosures for Ayalew Tefferi

BHS Annual Meeting

Myelodysplastic Syndrome Case 158

Clonal Cytopenia and Myeloid Neoplasms

Accel-Amplicon Panels

Liquid biopsy in lung cancer: The EGFR paradigm

Leukemia and subsequent solid tumors among patients with myeloproliferative neoplasms

Clinical Grade Biomarkers in the Genomic Era Observations & Challenges

Objectives and Financial Disclosure

A pediatric patient with acute leukemia of ambiguous lineage with a NUP98-NSD1 rearrangement SH

CHALLENGING CASES PRESENTATION

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

Transcription:

Next-Generation Sequencing (NGS) Testing for Hematologic Malignancies David Viswanatha, M.D. Division of Hematopathology Mayo Clinic, Rochester, Minnesota 2017 MFMER slide-1 DISCLOSURES Relevant Financial Relationship(s) None 2017 MFMER slide 2 Objectives Understand Basic Next Generation Sequencing (NGS) technology and its advantages, shortcomings and costs Overview of current NGS testing applications in hematologic malignancies, with emphasis on myeloid tumors Practical examples: case presentations 2017 MFMER slide 3 1

Part I: Why Next-Gen? 2017 MFMER slide 4 What Is Precision Medicine? The oncologist The pathologist The regulator The payer The patient 2017 MFMER slide 5 Mutation Burden By Cancer The prevalence of somatic mutations across human cancer types LB Alexandrov et al. Nature 500, 415 421 (22 August 2013) doi:10.1038/nature12477 2017 MFMER slide 6 2

Ley T et al. NEJM2013;368:2059 74 Papaemmanuil E, et al. Blood 2013;122:3616 27 2017 MFMER slide 7 Sanger Sequencing and NGS Sanger Next Gen Single target 1 target/sample Sensitivity ~20% Limited range (500-600 bp) CNV, translocations, large deletions not detected Complex sequences difficult to evaluate Multi-target Multiple samples Sensitivity ~5-10% Extended range (1000 s bp) CNV, translocations, large deletions not well detected Complex sequences possibly easier to evaluate 2017 MFMER slide 8 NGS Universe Targeted panels WES WGS RNA Seq Immune Seq CHIP Seq Methylome Seq 2017 MFMER slide 9 3

Key Considerations for NGS Design: Balancing Competing Constraints Comprehensive Analysis Large panel, WES, WGS Higher expense/sample Lower depth & slower TAT Small (targeted) panel Lower expense/sample Higher depth & faster TAT 1 Sample Throughput 100 2017 MFMER slide 10 Overview of NGS Method Adapted from: Jill M. Johnsen et al. Blood 2013;122:3268-3275 2013 by American Society of Hematology 2017 MFMER slide 11 NGS Workflow Wet Bench Process Dry Bench Informatics Library Prep 1 0 Demultiplexing Base calling Pre analytic Template Prep 2 0 Alignment Variant calling Specialized applications Sequencing 3 0 Variant Annotation Variant significance* 2017 MFMER slide 12 4

NGS: Pre-analytic Variables Sample type (e.g. fresh vs. FFPE), anticoagulant, etc. Tumor percentage requirement Need for microdissection (FFPE) Cell sorting DNA (or RNA) extraction and minimum Use of paired normal sample Adjacent normal, buccal, sorted T-cells, etc. 2017 MFMER slide 13 NGS: Library Preparation Amplicon-based Highly multiplexed PCR Less DNA input required Faster library generation Limited target region coverage CNV assessment more difficult Best for hot spot/regions Cheaper? Capture-based Bait (oligonucleotide) probe pull-down of fragmented high quality DNA More DNA input required Longer library preparation Better and more even target region coverage CNV detection easier Note: Sample throughput and accuracy similar 2017 MFMER slide 14 NGS: Platforms Illumina MiSeq V2 Bridge amplification Paired end reads possible Sequencing-by-synthesis Slower (~24 hrs) Less artifact Higher output* More expensive* Ion Torrent PGM Emulsion droplet PCR Single end reads Longer read lengths Semiconductor chip sequencing Homopolymer artifacts Faster (<24 hrs) Less expensive 2017 MFMER slide 15 5

NGS: Analysis Bioinformatics processing Index demultiplexing, base call and quality, sequencer performance data Secondary pipelines: initial alignment to reference, SNV and indel detection, variant calling Additional analyses: breakpoint analysis, structural variant (SV) detection, specialized applications (RNAseq, immunology, microbial, etc.) Data and computationally intensive Interpretation Final variant annotation and curation Pathogenicity determination (5 level) Potential germline association(s) Detailed report creation 2017 MFMER slide 16 Variant Classification is Multidisciplinary! NGS Analysis Team QC review Visual BAM file review Annotation & preliminary variant classification Initial report generation Consultant HP Final review and editing of mutation effect and pathogenic class Final report Genetic Counselors In depth variant classification and comment edits Evaluate possible germline findings 2017 MFMER slide 17 NGS Sequencing Cost $700 1000/sample! $1k per genome 2017 MFMER slide 18 6

NGS Panel Turn-around Time 24 48 hrs 24 36 hrs thermofisher.com 24 hrs genomics.agilent.com 2 to 4 hrs/case illumina.com 48 96 hrs/case wikipedia.com Typical TAT: 7 10 days 2017 MFMER slide 19 INFORMATION OVERLOAD!!! 2017 MFMER slide 20 Although not detracting from the excitement and opportunity that the current situation affords us, we would introduce a note of caution. Like modern day archeologists confronted with ancient Egyptian hieroglyphs, our current ability to confidently translate the presence of these mutations into clinical gains for individual patients significantly lags behind our ability to detect them. Grimwade D, et al. Blood 2016;127(1):29 41 2017 MFMER slide 21 7

Pros and Cons of NGS Panel Testing Advantages Platform convergence Multiplexing capacity Multiple applications Comprehensive variant detection Information density (comprehensive analysis) Integrated reporting Disadvantages Cost and TAT* Technical complexity Interpretive complexity Customization Limited platform availability Information density (uncertainty) and scale Reimbursement 2017 MFMER slide 22 Part II: Applications of NGS to Hematologic Malignancies 2017 MFMER slide 23 2017 MFMER slide 24 8

Targeted Therapies and Cancer (I) Disease Target Therapies Notes Colon Ca EGFR cetuximab Contraindicated if K/NRAS, RAF mutation PD1 pembrolizumab MSI status NSCLC EGFR cetuximab T790M mutation resistance RET cabozantinib Rare, young age, non smoker ROS1, ALK crizotinib Rare, female, younger Other solid T s PD1 pembrolizumab H&N squamous Ca; GU tumors, breast 2017 MFMER slide 25 Targeted Therapies and Cancer (II) Disease Target Therapies Notes CHL PD1 pembrolizumab ABVD Rx effective NHL BCR signaling (BTK, PIK3CD) CD20, CD22, CD25 Ibrutinib, idelasilib Rituximab, epratuzumab, alemtuzumab, blinatumomab ABC DLBCL, LPL/WM, CLL Humanized antibodies, BiTE agents, etc. BCL2 Venetoclax CD19 CAR T ALL, expanding MM 26S proteasome Bortezomib +2 nd generation drugs CRBN pathway Lenalidomide (IMiD) +2 nd generation drugs 2017 MFMER slide 26 Targeted Therapies and Cancer (III) Myeloid neoplasms: BCR-ABL (CML, Ph+ B-LBL) PML-RARA (APL) Highly efficacious targeted drugs for most other myeloid cancers are not significantly prevalent Why? 2017 MFMER slide 27 9

Signal Transduction: JAK2 KIT FLT3 CSF3R K/NRAS MPL CBL PTPN11 RNA Splicing: SF3B1 SRSF2 U2AF1 ZRSR2 Transcription Factor: GATA1/2 RUNX1 CEBPA ETV6 NOTCH1 BCOR WT1 PHF6 Chromosome stability: TERT Cohesin complex (STAG1, SMC3, RAD21) Tumor Suppressor: TP53 Epigenetic: IDH1/2 TET2 EZH2 ASXL1 DNMT3A Other: SETBP1 CALR NPM1 2017 MFMER slide 28 Blood. 2015 Feb 26;125(9):1367 76 Blood. 2016;128(5):686 698 2017 MFMER slide 29 Patterns of AML Mutations SRSF2 SF3B1 U2AF1 ZRSR2 PTPN11 EZH2 CBL ASXL1 STAG2 BCOR K/NRAS FLT3 TP53 IDH1/2 TET2 DNMT3A GATA2 RUNX1 NPM1 CEBPA CBF r KMT2A r Ref: Blood. 2015;125(9):1367 1376 2017 MFMER slide 30 10

N Engl J Med 2016;374:2209 21 2017 MFMER slide 31 References 1. Blood. 2016;127(20):2391 2405 2. Blood. 2017;129(4):424 447 3. J Natl Cancer Net 2017;15:926 957 4. Arch Pathol Lab Med. doi: 10.5858/arpa.2016 0504 CP 2017 MFMER slide 32 Gene Mutation Profiling: Myeloid Tumors AML: Tier 1 Tier 2 FLT3 ITD, IDH2, TP53, CEBPA dm Spliceosome/epigenetic, TP53, KIT, RUNX1,?DNMT3A Tier 3 Germ line associated: GATA2, DDX41, ANKRD26, CEBPA... MDS: SF3B1, TP53, other (mutation burden) MDS/MPN: ASXL1, TET2, SRSF2, JAK2, SETBP1, ETNK1, RAS pathway MPN (PMF): JAK2, CALR, MPL, ASXL1, TP53, CSF3R 2017 MFMER slide 33 11

Part III: Case Presentations 2017 MFMER slide 34 Case 1 62 year old male: macrocytic anemia and decreased platelets CBC: Hgb 10 g/dl, MCV 108.4 fl, WBC 10.3 X10 9 /L, platelets 141 X10 9 /L WBC: Ne 50%, Ly 38%, Mo 12%, 0% blasts Bone marrow evaluated at Mayo Clinic: Hypercellular (50-60%): myelomonocytic hyperplasia with 15% blasts and promonocytes Cytogenetics: 46,XY[20] Diagnosis: Chronic myelomonocytic leukemia-2 (CMML-2) 2017 MFMER slide 35 Case 1 2017 MFMER slide 36 12

Case 1 2017 MFMER slide 37 Case 1 2017 MFMER slide 38 Case 1 2017 MFMER slide 39 13

Case 1 NM_002520.6 NPM1 NM_022552.4 DNMT3A 2017 MFMER slide 40 Case 1 NGS findings: DNMT3A: c.2645g>a; p.arg882his (40%) NPM1: c.860_863dup; p.trp288cysfs*12 (26%) Conservative Rx (decitabine) with follow-up at another institution 6 months later: 22% PB blasts NGS results can inform or refine difficult morphologic diagnosis and/or suggest alternative biologic consequences 2017 MFMER slide 41 CMML: Molecular Pathogenesis 50 45 40 35 30 25 20 15 10 5 0 Adapted from Itzykson R and Solary E Leukemia 2013;27:1441 1450 RAS CBL JAK2 TET2 SRSF2 ASXL1 RUNX1 2017 MFMER slide 42 14

Case 2 33 year old female with diffuse back and hip pain July 2015 CBC: Hgb 12 g/dl, WBC 7.6 X10 9 /L, platelets 24 X10 9 /L WBC: Ne 13%, Ly 18%, blasts 66% Bone marrow biopsy: 92% myeloblasts Flow cytometry: CD45, CD13, CD15, CD33, CD117, HLA-DR, CD7, CD36, CD64, CD38 Cytogenetics 46,XX[20]; AML FISH negative 2017 MFMER slide 43 Case 2 NGS findings: FLT3: c.2503g>t; p.asp835tyr (18%) NPM1: c.860_863dup; p.trp288cysfs*12 (39%) PTPN11: c.218c>t; p.thr73ile (26%) WT1: c.938c>a; p.ser313* (44%) Rx: HiDAC induction chemotherapy with CR1; MUD allogeneic SCT in 10/2015 Remains in complete remission >12 months NGS results may indicate gene mutation patterns with variable, conflicting or uncertain prognostic information 2017 MFMER slide 44 Study Outcome Summary Virappane 2008: 470 CN-AML (94% de novo) WT1+ 10% Paschka 2008: 196 young (<60) with de novo CN-AML WT1+ 10.7%; Associated with FLT3-ITD and higher WBC Median F/U: 122.4 mo RFS: 5 yr: 22% vs 44% (HR 2.16; p 0.005) OS:5 years: 26% vs 47% HR 1.91 p=0.007 Median F/U : 50.4 mo RFS: 3 yr DFS: 13% vs 50% (p<0.001) OS: 10% vs 56% (p<0.001) Worse OS, RFS independent of NPM1 and FLT3 ITD+; Remained significant for OS, RFS at ASCT Poorer DFS (HR 2.7 (p=0.009) controlling for ERG, FLT3 ITD, CEBPA Poorer OS HR 3.2, p=<0.001) controlling for CEBPA, FLT3 ITD/NPM1, WBC Gaidzik 2009: 617 De novo, s-aml, t-aml WT1+ 12.6%; Associated with younger age, FLT3-ITD+ and CEBPA, higher blasts, inverse NPM1+ Med F/U 44 mo RFS: NO difference WT1+/- but WT+/FLT3-ITD + subgroup P=0.006 OS: NO difference, but WT1+/FLT-ITD + subgroup P<0.001 WT1 mutations had NO significant impact. NPM1, FLT3 ITD, CEBPA, MLL were significant Hou 2010: 470 non-m3 AML WT1+ 6.8% (33% M6) Krauth 2015: 3157 AML (1178 CN-AML) WT1+ 6%; FLT3-ITD+ 8.5% Bi-CEBPA 13.6% Median F/U: 53 mo RFS: 6 mo vs 14 mo (p<0.001) OS: 14 mo vs. 29.5 mo (p=0.021) RFS: 10.8 mo vs 17.9 mo (p=0.008) OS: NO significant impact Poorer OS and RFS in all and young CN-AML pts. (RR 3.752, p=0.024; RR 3.8, p-0.003) NPM1+/FLT3 ITD- favorable Poorer EFS controlling for FLT3 ITD, age HR 1.64 (p=0.002) NPM1+/FLT3 ITD- HR 0.68 (P<0.001) 2017 MFMER slide 45 15

Case 3 75 year old with recent Dx lung adenocarcinoma History of thrombocytopenia, apparently for several years, previous normal bone marrow Hgb 12.4 g/dl, MCV 99.2 fl, WBC 3.5 X10 9 /L, platelets 45 X10 9 /L WBC: Ne 50%, Ly 743%, Mo 4%, Eo 3% Bone marrow 12/15: hypercellular (50%) with 5% ring sideroblasts, slightly decreased megakaryocytes and no increase in blasts Diagnostic morphologic features of MDS not established Cytogenetics: 46,XX,del(20)(q11.2q13.2)[1/46,XY[19] 2017 MFMER slide 46 2017 MFMER slide 47 2017 MFMER slide 48 16

Case 3 NGS findings: NM_001025203.1 U2AF1: c.4708a>c; p.gln157pro (16%) 2017 MFMER slide 49 2017 MFMER slide 50 Jaiswal S. N Engl J Med 2014;371:2488 98 2017 MFMER slide 51 17

CHIP-ICUS-CCUS-Cancer Spectrum CHIP ICUS CCUS Individuals without history or concurrent hematologic cancer Somatic mutations in one or sometimes >1 gene detected at >2% variant allele fraction (VAF) TET2, DNMT3A, ASXL1, SF3B1, TP53 Increasing incidence with age Identified from WES/WGS of large populations Variable risk of progression to myeloid cancer (0.5 1% per year; 10X OR risk) Unexplained cytopenia(s) in absence of cytogenetic and NGS detectable clonal population Uncertain risk of neoplastic progression, likely low CHIP + ICUS Risk of progression to myeloid cancer is higher and becoming more quantifiable 2017 MFMER slide 52 CHIP: Associations CHIP is highly prevalent (>90%) after age 50 with high sensitivity techniques {Nat Commun. 2016;7:12484} As defined, CHIP increases risk for developing a hematologic neoplasm Risk increases if multiple mutations, higher VAF% Risk of all-cause mortality and cardiovascular disease Therapy-related myeloid neoplasms: higher risk for development of T-MN with pre-existing CHIP {Lancet Oncol 2017; 18: 100 11; Lancet Oncol 2017; 18: 112 21} Presence of CHIP in donor HSCT allograft can result in impaired graft function in recipient but not donor-derived leukemia {Blood 2017;130:91-94} Not all CHIP is equal {Blood. 2017;130(6):753-762} TET2 and DNMT3A very common, yet not clearly associated with adverse effects on hematopoietic function, or risk of myeloid neoplasia Clonal size expands with age in some individuals:? Compensation for failing wild type HSCs 2017 MFMER slide 53 CCUS: Risk of Neoplastic Progression Earlier studies: highly suggestive of increased risk for ICUS with clonal hematopoietic mutations to progress to overt myeloid cancers {Blood. 2015;126(21):2362-2365; Blood. 2015;126(21):2355-2361} Recent large studies confirm and delineate the risk more specifically {Blood. 2017;129(25):3371-3378; Am. J. Hematol. 91:1234 1238, 2016} Longitudinal follow-up VAF>10%; >2 mutations and spliceosome mutations with classic CHIP alterations have very high PPV for progression to myeloid neoplasm HR for progression 13.9 Certain mutation patterns may be more strongly predictive and can provide presumptive evidence for early myeloid neoplasia in the absence of definitive morphologic criteria or cytogenetic abnormalities Conversely the absence of mutations (with sufficiently comprehensive screening) has very high NPV for neoplasia, even in the setting of morphologic atypia 2017 MFMER slide 54 18

Suggested Indications for NGS Testing Useful New AML Relapse AML New Dx MDS MDS/MPN (e.g. CMML) MPN-PMF or triple negative Patient with suspicion of GL syndrome Unexplained cytopenias ( ICUS ) Not Indicated/Uncertain Palliative situation Mastocytosis (KIT D816V, KIT seq) MPN-PV, MPN-ET (MPN-R) MPN-CNL (CSF3R) CML (BCR-ABL) CEL/HES and other neoplastic eosinophilias (FISH) 2017 MFMER slide 55 NGS and Myeloma GENETIC CATEGORY RISK STATUS Hyperdiploid/CNA HD (odd chromosome # s) Standard (35 40%) CNA at +1q, del1p, del17p High (5 10% each) Translocations t(11;14) CCND1, t(6;14) CCND3 Standard (15%) t(14;16) and t(14;20) MAF;MAFB High (<5%) t(4;14) FGFR3; NSD2 (MMSET, WHSC1) Intermediate (10 15%) 8q24 MYC High (20%) Notes: Based on Mayo msmart 2.0 High risk GEP signature is also high risk feature 2017 MFMER slide 56 NGS and Myeloma Modify or additionally stratify current FISH-defined risk classification Focus on additional prognostic alterations and therapeutic/resistance genotypes Ideal state is a single test to capture most relevant alterations in SMM, MM (e.g. SNV, SV, CNV) Technically difficult Possible utility for refractory/relapsed disease In early stages of application and clinical utility determination (indications) 2017 MFMER slide 57 19

NGS and Myeloma MAPK N/KRAS BRAF Other FGFR3 CCND1 FAM46C DIS3 IMiD CRBN CUL4B IKZF1 IRF4 NFKB TRAF3 CYLD DNA DR TP53 ATM ATR EGR1 2017 MFMER slide 58 Summary NGS is a comprehensive and convergent technology platform for hematologic cancer genetics NGS is currently relatively expensive and information dense Useful for diagnosis, prognosis and limited theranostics Evolving understanding of co-mutation patterns and clonal hematopoiesis 2017 MFMER slide 59 Questions & Discussion 2017 MFMER slide 60 20