Cytogenetic and molecular abnormalities in AML. Dr Elizabeth Tegg Director of haematology Pathology West

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

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

Test Name Results Units Bio. Ref. Interval. Positive

Fluorescence in-situ Hybridization (FISH) ETO(RUNX1T1)/AML1(RUNX1) or t(8;21)(q21.3;q22)

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

WHO Classification of Myeloid Neoplasms with Defined Molecular Abnormalities

Role of FISH in Hematological Cancers

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

Krishna Reddy CH and Ashwin Dalal. Diagnostics Division, Centre for DNA Fingerprinting and Diagnostics, Hyderabad

Test Name Results Units Bio. Ref. Interval. Positive

HEMATOLOGIC MALIGNANCIES BIOLOGY

Acute Myeloid Leukemia with Recurrent Cytogenetic Abnormalities

Significance of Chromosome Changes in Hematological Disorders and Solid Tumors

Significance of Chromosome Changes in Hematological Disorders and Solid Tumors

Acute myeloid leukemia. M. Kaźmierczak 2016

GENETICS OF HEMATOLOGICAL MALIGNANCIES

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

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

Oncology Genetics: Cytogenetics and FISH 17/09/2014

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

Combinations of morphology codes of haematological malignancies (HM) referring to the same tumour or to a potential transformation

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

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

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

CYTOGENETICS Dr. Mary Ann Perle

Test Name Results Units Bio. Ref. Interval. Positive

Addressing the challenges of genomic characterization of hematologic malignancies using microarrays

Acute leukemia and myelodysplastic syndromes

Examining Genetics and Genomics of Acute Myeloid Leukemia in 2017

Chromosomal Aberrations

Corporate Medical Policy. Policy Effective February 23, 2018

Meeting VAKB 8 februari 2011 Nancy Boeckx, MD, PhD

Integration of microarray analysis into the clinical diagnosis of hematological malignancies: How much can we improve cytogenetic testing?

Outline. Chromosomal analysis FISH. Chromosomal abnormalities in cancer. Clinical application of cytogenetics. Procedure Nomenclature

Volume 7, Issue 1 January 2012

Introduction to Cytogenetics

HEMATOPATHOLOGY SUMMARY REPORT RL;MMR;

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

Mixed Phenotype Acute Leukemias

Haematology Probes for Multiple Myeloma

An International System for Human Cytogenetic Nomenclature (2013)

Initial Diagnostic Workup of Acute Leukemia

Acute Lymphoblastic and Myeloid Leukemia

Case 1. Sa A.Wang, MD UT MD Anderson Cancer Center Houston, TX

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

Done By : WESSEN ADNAN BUTHAINAH AL-MASAEED

Acute Myeloid Leukemia with RUNX1 and Several Co-mutations

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

NUP214-ABL1 Fusion: A Novel Discovery in Acute Myelomonocytic Leukemia

Canadian College of Medical Geneticists (CCMG) Cytogenetics Examination. May 4, 2010

WHO Classification 7/2/2009

Therapy-related acute myeloid leukemia with germline TP53 mutation (Li-Fraumeni syndrome) SH Chelsey Deel MD Teresa Scordino MD

Chromosome Abnormalities

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

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

Hematology Unit Lab 2 Review Material

ADVANCES IN CHILDHOOD ACUTE LEUKEMIAS : GENERAL OVERVIEW

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

Classification of Hematologic Malignancies. Patricia Aoun MD MPH

Molecular Hematopathology Leukemias I. January 14, 2005

Session 7 Summary. Magdalena Czader, MD, PhD David Czuchlewski, MD MOLECULAR GENETICS OF HEMATOPOIETIC NEOPLASMS

Acute Promyelocytic Leukemia with i(17)(q10)

Leukemia (2007) 21, Cytoplasmic mutated nucleophosmin (NPM) defines the molecular status of a significant fraction of myeloid sarcomas

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

An Overview of Cytogenetics. Bridget Herschap, M.D. 9/23/2013

Hematopathology Case Study

Template for Reporting Results of Monitoring Tests for Patients With Chronic Myelogenous Leukemia (BCR-ABL1+)

Dr. Anjali Kelkar (DNB Path, IFCAP)

Myeloproliferative Disorders - D Savage - 9 Jan 2002

Nature Medicine: doi: /nm.4439

Application of Whole Genome Microarrays in Cancer: You should be doing this test!!

Supplementary Appendix

The mutations that drive cancer. Paul Edwards. Department of Pathology and Cancer Research UK Cambridge Institute, University of Cambridge

«Adverse Prognosis» Acute Myeloid Leukemia

2010 Hematopoietic and Lymphoid ICD-O Codes - Alphabetical List THIS TABLE REPLACES ALL ICD-O-3 Codes

2012 Hematopoietic and Lymphoid ICD-O Codes - Numerical List THIS TABLE REPLACES ALL ICD-O-3 Codes

Heme 9 Myeloid neoplasms

Acute myeloid leukemia: a comprehensive review and 2016 update

Cytogenetics Update. Lynda J Campbell

Fluorescent in situ hybridization studies in multiple myeloma

Supplementary Appendix

Review. Genetic Tests To Evaluate Prognosis and Predict Therapeutic Response in Acute Myeloid Leukemia

GENETIC MARKERS IN LYMPHOMA a practical overview. P. Heimann Dpt of Medical Genetics Erasme Hospital - Bordet Institute

CGC myeloid malignancy working group updates. Xinjie Xu & Rashmi Kanagal-Shamanna

Molecular Advances in Hematopathology

Do acgh analysis have a place in routine cytogenetic workup in leukemia/cancer? - A single institution experience. Cambridge, April 9 th 2013

Structural Variation and Medical Genomics

The Revised 2016 WHO Classification of Acute Leukemias

Myeloid neoplasms. Early arrest in the blast cell or immature cell "we call it acute leukemia" Myoid neoplasm divided in to 3 major categories:

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

Leukaemia Section Review

Please Silence Your Cell Phones. Thank You

JAK2 V617F analysis. Indication: monitoring of therapy

The Power of Observation

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

Nucleic Acid Testing - Oncology. Molecular Diagnosis. Gain/Loss of Nucleic Acid. Objectives. MYCN and Neuroblastoma. Molecular Diagnosis

Transcription:

Cytogenetic and molecular abnormalities in AML Dr Elizabeth Tegg Director of haematology Pathology West

Outline Classification of AML Types of genetic changes Next generation sequencing in HM

Outline Classification of AML Types of genetic changes Next generation sequencing in HM

Genetics: Importance Diagnosis Prognosis Different treatment options

Classification of HM Current WHO classification 2001, 2008, 2016 Morphology Immunophenotype Cytogenetics Molecular genetic Clinical features

Current Classification: WHO of AML 1.AML with recurrent genetic abnormalities AML with t(8;21)(q22;q22) AML with inv(16)(p13q22) or t(16;16)(p13;q22) Acute promyelocytic leukamia AML with t(15;17)(q22;q12) AML with 11q23 (MLL) abnormalities AML with t(6;9)(p23;q34) AML with inv(3)(q21q26.2) AML with t(1;22)(p13;q13) AML with gene mutations FLT3, NPM1 2.AML with myelodysplasia related changes 3.Therapy related myeloid leuakemia Alkylating agent related Topoisomerase II inhibitor-related 4.AML not otherwise specified AML, minimally differentiated AML without maturation AML with maturation Acute myelomonocytic leukaemia Acute monoblastic and monocytic leukaemia Acute erythroid leukaemia Acute megakaryoblastic leukaemia Acute basophilic leukameia Acute panmyelosis with myelofibrosis 5. Myeloid sarcoma 6. Myeloid proliferations related to Downs syndrome 7. Blastic plasmacytoid dendritic cell neoplasm

Outline Classification of AML Types of genetic changes Next generation sequencing in HM

Types of genetic changes Chromosomal DNA Epigenetic Changes in the tumour compared to germline

Cytogenetics

Cytogenetics Classic abnormalities Cryptic abnormalities All the above have been reported as also being cryptic A translocation that is undetectable to the eye FISH

Conventional Cytogenetics Aim: to get the maximal number of the cell of interest dividing and then halt cell division so that chromosomes can be visualised at their clumpest stage of the cell division (Prometaphase/metaphase). Pros: good overview of the whole genome of the disease Cons: Can be difficult to get cells dividing, low resolution

chromosomes for Conventional cytogenetics Culture: LIVE CELLS Mitogen Spindle inhibitor: HALTS DIVISION Hypotonic solution Fixative Banding: GTL Analysis

Cell cycle

Mitosis

Clones Defined as a cell population derived from a single progenitor. It is common practice to infer a clonal origin when a number of cells have the same or closely related abnormal chromosome complements. The clone size is given in square brackets [ ] after the karyotype At diagnosis we look at only 20 metaphases Follow up 40 metaphases

Stemline, sideline and clonal evolution Cytogenetically related clones (subclones) are presented as far as possible in order of increasing complexity, irrespective of the size of the clone. Stemline (sl) is the basic clone of a tumour and listed first Sideline (sdl) all additional derived clones 46,XX,t(9;22)(q34;q11.2)[3]/47,XX,+8,t(9;22)(q34;q11.2)[17] ml: 47,XX,+8,t(9;22)(q34;q11.2)[17] sl: 46,XX,t(9;22)(q34;q11.2)[3] sdl: 47,XX,+8,t(9;22)(q34;q11.2)[17] 46,XX,t(9;22)(q34;q11.2)[17]/47,XX,+8,t(9;22)(q34;q11.2)[3] ml: 46,XX,t(9;22)(q34;q11.2)[17] sl: 46,XX,t(9;22)(q34;q11.2)[17] sdl: 47,XX,+8,t(9;22)(q34;q11.2)[3]

Karyotype Count chromosomes: Modal number Near-haploid (23+/-) <34 Hypohaploidy <23 Hyperhaploidy 24-34 Near-diploid (46+/-) 35-57 Hypodiploidy 35-45 Hyperdiploidy 47-57 Near-triploid (69+/-) 58-80 Hypotriploidy 58-68 Hypertriploidy 70-80 Near-tetraploidy (92+/-) 81-103 Hypohaploidy 81-91 Hyperhaploidy 93-103

Order of abnormalities All abnormalities are listed in numerical order X, Y then 1-22 Numerical abnormalities before structural Structural abnormalities second in alphabetical order add, del, inv, t, 47,XX,+1,t(1;3)(p32;q21) 47,XX,t(1;3)(p32;q21),+21 If abnormalities occur to the same individual chromosome (ie is a derivative), abnormalities are listed according to breakpoint (pter to qter) and not separated by a comma 46,XX,der(1)t(1;3)(p32;q21)add(1)(q25)

Chromosomal abnormalities Numerical Monosomy Trisomies Structural Add Translocation Duplication Deletion Inversion Isochromosome Ring Marker Double minutes

What does this mean 46,XX[20] Normal female karyotype 70,XXX,+8[20] Near triploid cell line, with additional chromosome 8 (ie 3 copies of all chromosomes with 4 copies of 8) 46,XY,inv(16)(p13q22)[20] AML with inversion 16

Numerical Changes A very common mechanism Seen in both Myeloma, AML and ALL Hyperdiploidy is typical in ALL modal chromosome number is 54 Hyperdiploidy with ~47 chromosome is seen in AML/MDS Typical patterns of chromosomes involved

Cytogenetics: Numerical Monosomy 7 AML, MDS, MPN Trisomy 8 All haematological malignancies Trisomy 21 All haematological malignancies

Structural Abnormalities in Leukaemia Found in 65% of cases 124 different structural abnormalities have been described But a sub-group of 30 represent the majority of abnormalities seen Are translocations 65%, deletions 30% or inversions 5%.

Translocation: Reciprocal exchange of two chromosomal segments Deletion: Removal of a chromosomal segment Inversion: Inversion of a segment around the centromere

Chromosome Translocations Result in the juxtaposition of previously separate DNA sequences eg the t(9;22)(q34;q11.2) When this was first published (1973) the concept of a translocation of genetic material was new 9 der(9)elizabeth der(22) Tegg 22016

Deletions Deletions remove genes that Stop the cell proliferating out of control Whose normal function is to detoxify chemical agents That repair DNA damaged by several mechanisms

Deletions Most common deletions in leukaemia are Deletions of 5q/7q and 20q in MDS/AML Deletions of 6q, 9p and 12p in ALL

Inversions Are an uncommon mechanism The best known is the inv(16) which is diagnostic of AML M4 with eosinophilia Associated with the most favourable outcome for all classes of AML in adults

Inversion 16(p13q22) Vysis LSI CBFB Dual Color, Break Apart Rearrangement Probe CBFB 5 CBFB 3 CBFB The inv(16) interrupts the coding region of the CBFB and MYH11 genes and leads to a chimeric protein that competes with normal

Fluorescence in situ hybridization Fluorescently labelled probes Denature the probe and target DNA (Heat) Anneal stage: complimentary sequences pair Wash off unbound probe Counter stain Analyse under a fluoresent microscope

FISH probes Locus Specific Identifiers Break Apart Dual Colour/Dual Signal, TriColour/Dual Fusion Dual Colour/Single Fusion

FISH nomenclature All changed recently with the ISCN 2005 and in 2013 ish: metaphase nuc ish: interphase nuc ish(abl1 x3),(bcr x3),(abl1 con BCR x2)[400] nuc ish(abl1,bcr)x3(abl1 con BCR x2)[400] All 400 interphase cells showed two dual fusion signals with no evidence of a deletion of the derivative 9 ABL1 x 3 BCR x 3 ABL1 con BCR x 2

Break-apart probes Break-apart probes are made of 2 probes, the short form does not convey that the normal situation is presence of 2 fusion signals nuc ish(mll x 2)[400] nuc ish(5 MLL,3 MLL)x2(5 MLL con 3 MLL x2)[400] Interpret: 400 interphase cells show a normal signal pattern. No evidence of rearrangement of MLL

Break-apart probe: MLL DFBA red=5 MLL green=3 MLL nuc ish(mllx2)[400] nuc ish(mll x 2)(5 MLLsep3 MLL x 1)[400] nuc ish(5 MLL x 2,3 MLL x1)(5 MLL con 3 MLL x 1)[400]

FISH vs Conventional Pros On both metaphase or interphase cells Target genetic aberrations Cons Inability to provide a genomewide assessment Necessity of clinical information to drive what probes to use Requirement of a high quality fluorescence microscope with multiple filters

Molecular karyotype DNA based technology Array comparative genomic hybridization Bac (Bacterial artificial chromsomes) SNP arrays High probe density in cancer relevant regions of the genome Higher sensitivity Don t need dividing cells Detect CNV and LOH Not good for balanced translocations or inversions

SNP arrays Copy neutral-loss Of Heterozygousity Identification of PAX5 as a key target of genetic inactivation in B-ALL Identification of TET2 as a major tumour suppressor in MDS Need germline DNA for comparison Mechanism where heterozygous mutations become homozygous Heinrichs S, Li C, Look AT. SNP array analysis in hematological malignancies: avoiding false discoveries. Blood. 2010 March 19, 2010.

What have we learnt from cytogenetics A lot of subtypes are defined by simple translocations Many of the numerical gains have patterns to them and it is consistently the same chromosome lost or gained

1. AML with recurrent cytogenetic abnormalities AML with recurrent genetic abnormalities AML with t(8;21)(q22;q22) AML with inv(16)(p13q22) or t(16;16)(p13;q22) Acute promyelocytic leukamia AML with t(15;17)(q22;q12) AML with 11q23 (MLL) abnormalities AML with t(6;9)(p23;q34) AML with inv(3)(q21q26.2) AML with t(1;22)(p13;q13)

AML with t(8;21)(q22;q22) 5-12% of AML, mainly younger patients genes involved RUNX1/ RUNX1T1 Can diagnose AML with <20% blasts with this abnormality Good prognosis

AML with inv(16)(p13q22) or t(16;16)(p13;q22) 10-12% of AML, predominantly younger pt genes involved CBF beta to MYH11 (smooth muscle mycin gene) Can diagnose AML with <20% with this abnormality Good prognosis

AML with inv(16)(p13q22) or t(16;16)(p13;q22) Vysis LSI CBFB Dual Color, Break Apart Rearrangement Probe CBFB 5 CBFB 3 CBFB The inv(16) interrupts the coding region of the CBFB and MYH11 genes and leads to a chimeric protein that competes with normal

Acute promyelocytic leukaemia with t(15;17)(q22;q12) 5-8% of AML, usually adults in mid life genes involved PML/RAR alpha Best prognosis if you survive the first week Variant translocations t(11;17)(q23;q21) t(5;17)(q23;q12) t(11;17)(q13;q21) t(11;17)(q23;q21) is resistant to ATRA, and is morphologically the same Independent prognostic factor is WCC at diagnosis <2 is good Numerous Auer rods

Acute promyelocytic leukaemia AML with t(15;17)(q22;q12)

AML with 11q23 (MLL) abnormalities Usually associated with monocytic features 5-6% of AML, mainly children Two clinical subgroups have a higher frequency 1. Aml in infants 2. Therapy related, usually after DNA topoisomerase II inhibitors gene involved MLL gene ( Drosophila trithorax gene) Is a developmental regulator which is structurally altered

Gene names

AML with 11q23 (MLL) abnormalities Mixed Lineage Leukaemia Over 90 reported translocation MLL translocations predicts early relapse and poor prognosis MLL Consists of at least 36 exons, encoding a 3969 amino-acid nuclear protein with molecular weight of nearly 430kDa It is thought to function as a positive regulator of gene expression in early embryonic development and haematopoiesis MLL translocation breakpoints cluster within an 8.3kb region spanning exons 5-11 The mechanisms by which these rearrangements results in leukaemia remain largely unknown Li et al Leukaemia 2005; 19: 183-190

AML with t(6;9)(p23;q34) Morphology Associated with M2 or M4 and basophilia DEK-CAN DEK-NUP214 Poor prognosis High association with FLT3-ITD

AML with inv(3)(q21q26.2) Maybe de novo or arise from MDS Morphology: increased atypical megakaryocytes

AML with t(1;22)(p13;q13) Acute megakaroblastic leukaemia Rare Most commonly occurs in Down syndrome

AML with gene mutations Fms-related tyrosine kinase 3 (FLT3) Nucleophosmin (NPM1) CEBPA KIT WT1 NRAS KRAS

DNA sequence changes Point mutation: simple change in one base Missence: amino acid change Nonsence: changes to a stop codon Frame-shift mutation: one or more bases is inserted or deleted

Molecular Subgroups of Cytogenetically normal AML Dohner H et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood. 2010 January 21, 2010;115(3):453-74.

AML with mutated NPM1 Mutations usually in exon 12 Aberrant cytoplasmic expression of nucleophosmin is a surrogate marker of this gene mutation Morphology: monocytic CC: normal Mutated in 45-64% of adult normal CC AML Good prognosis

FLT3 Located 13q12 Encodes a tyrosine kinase receptor that is involved in HSC differentiation and proliferation 2 primary types of mutations Internal tandem duplications (FLT3-ITD) (adverse outcome) Mutations affecting codon 835 or 836 of the second tyrosine kinase domain (TKD) (outcome?)

KIT Located 4q11-12 Member of type 3 tyrosine kinase family Generally test for KIT mutation in the core binding factor AML Poor prognosis

Epigenetic changes Disruption of DNA methylation Histone modification Chromatin compartments Esteller M. Epigenetics provides a new generation of oncogenes and tumour-suppressor genes. Br J Cancer. Elizabeth 2006;94(2):179-83. Tegg 2016

Outline Classification of AML Types of genetic changes Next generation sequencing in HM

Next generation sequencing in HM Sequencing of cytogenetically normal AML 50% of AMLs will be cytogenetically normal 12 acquired mutations in coding regions 2 in known genes (NPM1 and NRAS) 10 in genes not previously reported to be mutated in AML Mardis ER, Ding L, Dooling DJ, Larson DE, McLellan MD, Chen K, et al. Recurring Mutations Found by Sequencing an Acute Myeloid Leukemia Genome. New England Journal of Medicine. 2009 September 10, 2009;361(11):1058-66

International Cancer Genome http://www.icgc.org/ Consortium ICGC Goal: To obtain a comprehensive description of genomic, transcriptomic and epigenomic changes in 50 different tumor types and/or subtypes which are of clinical and societal importance across the globe.

Clinical utility WGS Ideally every HM to be sequenced Problems from a haematology point of view Normal DNA What is normal Every time a cell undergoes mitosis DNA changes occur Tumour DNA Heterogeneity with in the tumour cells 300-400 coverage needed We know from cytogenetics that different clones are present in tumours Passenger vs driver mutations

Perspectives for therapeutic targeting of gene mutations in acute myeloid leukaemia with normal cytogenetics British Journal of Haematology Volume 170, Issue 3, pages 305 322, August 2015 The acute myeloid leukaemia (AML) genome contains more than 20 driver recurrent mutations. Here, we review the potential for therapeutic targeting of the most common mutations associated with normal cytogenetics AML, focusing on those affecting the FLT3, NPM1 and epigenetic modifier genes (DNMT3A, IDH1/2, TET2). As compared to early compounds, second generation FLT3 inhibitors are more specific and have better pharmacokinetics. They also show higher anti-leukaemic activity, leading to about 50% of composite complete remissions in refractory/relapsed FLT3-internal tandem duplication-mutated AML. However, rapid relapses invariably occur due to various mechanisms of resistance to FLT3 inhibitors. This issue and the best way for using FLT3 inhibitors in combination with other therapeutic modalities are discussed. Potential approaches for therapeutic targeting of NPM1-mutated AML include: (i) reverting the aberrant nuclear export of NPM1 mutant using exportin-1 inhibitors; (ii) disruption of the nucleolus with drugs blocking the oligomerization of wild-type nucleophosmin or inducing nucleolar stress; and (iii) immunotherapeutic targeting of highly expressed CD33 and IL3RA (CD123) antigens. Finally, we discuss the role of demethylating agents (decitabine and azacitidine) and IDH1/2 inhibitors in the treatment of AML patients carrying mutations of genes (DNMT3A, IDH1/2 and TET2) involved in the epigenetic regulation of transcription.