Cytogenetic heterogeneity negatively impacts outcomes in patients with acute myeloid leukemia

Similar documents
Cytogenetic heterogeneity negatively impacts outcomes in patients with acute myeloid leukemia

Background CPX-351. Lancet J, et al. J Clin Oncol. 2017;35(suppl): Abstract 7035.

CME/SAM. Acute Myeloid Leukemia With Monosomal Karyotype. Morphologic, Immunophenotypic, and Molecular Findings

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

Corporate Medical Policy. Policy Effective February 23, 2018

Long-Term Outcome of Autologous Hematopoietic Stem Cell Transplantation (AHSCT) for Acute Myeloid Leukemia (AML)- Single Center Retrospective Analysis

Monosomal Karyotype Provides Better Prognostic Prediction after Allogeneic Stem Cell Transplantation in Patients with Acute Myelogenous Leukemia

Citation for final published version:

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

Acute myeloid leukemia (AML) is a

Evolving Targeted Management of Acute Myeloid Leukemia

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

Impact of additional genetic alterations on the outcome of patients with NPM1-mutated cytogenetically normal acute myeloid leukemia

CLINICAL STUDY REPORT SYNOPSIS

Published Ahead of Print on December 14, 2017, as doi: /haematol Copyright 2017 Ferrata Storti Foundation.

Where a licence is displayed above, please note the terms and conditions of the licence govern your use of this document.

Reference: NHS England 1602

Long-term risk of cancer development in adult patients with idiopathic aplastic anemia after treatment with anti-thymocyte globulin

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

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

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

New treatment strategies in myelodysplastic syndromes and acute myeloid leukemia van der Helm, Lidia Henrieke

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

KEY WORDS: CRp, Platelet recovery, AML, MDS, Transplant

Introduction. of some recurrent aberrations, for example, 8, del(9q), or CLINICAL OBSERVATIONS, INTERVENTIONS, AND THERAPEUTIC TRIALS

Acute Myeloid Leukemia

Outcome of acute leukemia patients with central nervous system (CNS) involvement treated with total body or CNS irradiation before transplantation

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

Early Clearance of Peripheral Blood Blasts Predicts Response to Induction Chemotherapy in Acute Myeloid Leukemia

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

Acute myeloid leukemia. M. Kaźmierczak 2016

Myelodyspastic Syndromes

New treatment strategies in myelodysplastic syndromes and acute myeloid leukemia van der Helm, Lidia Henrieke

REVIEW. Prognostic Value of RUNX1 Mutations in AML: A Meta-Analysis

Remission induction in acute myeloid leukemia

HHS Public Access Author manuscript Leukemia. Author manuscript; available in PMC 2015 August 01.

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

Association of Acute Myeloid Leukemia s Most Immature Phenotype with Risk Groups and Outcomes

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

Introduction CLINICAL OBSERVATIONS, INTERVENTIONS, AND THERAPEUTIC TRIALS

Daunorubicin, Cytarabine, and Midostaurin in Treating Patients With Newly Diagnosed Acute Myeloid Leukemia

Impact of Day 14 Bone Marrow Biopsy on Re-Induction Decisions and Prediction of a Complete Response in Acute Myeloid Leukemia Cases

Molecularly Targeted Therapies - Strategies of the AMLSG

Impact of ibrutinib dose intensity on patient outcomes in previously treated Waldenström macroglobulinemia

HEMATOLOGIC MALIGNANCIES BIOLOGY

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

Transfusion independence and survival in patients with acute myeloid leukemia treated with 5-azacytidine

Aalborg Universitet. DOI (link to publication from Publisher): /bloodadvances Creative Commons License Unspecified

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

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

Myeloperoxidase Expression in Acute Myeloid Leukemia Helps Identifying Patients to Benefit from Transplant

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

Supplementary Appendix

Cytogenetics and FLT3 ITD mutation predict clinical outcomes in non transplant patients with acute myeloid leukemia

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

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

Hull and East Yorkshire and North Lincolnshire NHS Trusts Haematology Multidisciplinary Team Guideline and Pathway. Acute Myeloid Leukaemia

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

Keywords: Acute Myeloid Leukemia, FLT3-ITD Mutation, FAB Subgroups, Cytogenetic Risk Groups

Better Prognosis for Patients With Del(7q) Than for Patients With Monosomy 7 in Myelodysplastic Syndrome

Time from diagnosis to treatment initiation predicts survival in younger, but not older, acute myeloid leukemia patients

BHS training course. Laboratory Hematology Cytogenetics. Lucienne Michaux. Centrum voor Menselijke Erfelijkheid, UZLeuven

RESEARCH ARTICLE. Introduction. Methods Wiley Periodicals, Inc.

Supplementary Appendix

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

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

Jeanne Palmer February 26, 2017 Mayo Clinic, Phoenix, AZ

AML IN OLDER PATIENTS Whenever possible, intensive induction therapy should be considered

Acute leukemia and myelodysplastic syndromes

Johann Hitzler, MD, FRCPC, FAAP Jacqueline Halton, MD, FRCPC Jason D. Pole, PhD

Protocol. Hematopoietic Stem-Cell Transplantation for Acute Myeloid Leukemia

Test Name Results Units Bio. Ref. Interval. Positive

Introduction CLINICAL OBSERVATIONS, INTERVENTIONS, AND THERAPEUTIC TRIALS

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

Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, Texas; 2 Sunesis Pharmaceuticals, Inc, South San Francisco

Cytarabine Dose for Acute Myeloid Leukemia

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

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

Disclosure. Study was sponsored by Karyopharm Therapeutics No financial relationships to disclose Other disclosures:

Case Report Blasts-more than meets the eye: evaluation of post-induction day 21 bone marrow in CBFB rearranged acute leukemia

Indication for unrelated allo-sct in 1st CR AML

Acute Myeloid Leukemia Progress at last

Mayer et al. BMC Cancer 2014, 14:69

Published Ahead of Print on January 12, 2017, as doi: /haematol Copyright 2017 Ferrata Storti Foundation.

PDF of Trial CTRI Website URL -

Page: 1 of 12. Genetic Testing for FLT3, NPM1, and CEBPA Mutations in Acute Myeloid Leukemia

midostaurin should be extended to patients who are deemed fit to receive intensive induction and consolidation, regardless of age.

Mixed Phenotype Acute Leukemias

Update: Chronic Lymphocytic Leukemia

The Past, Present, and Future of Acute Myeloid Leukemia

Acute Leukemia. Sebastian Giebel. Geneva 03/04/

Enasidenib Monotherapy is Effective and Well-Tolerated in Patients with Previously Untreated Mutant-IDH2 Acute Myeloid Leukemia

Acute Lymphoblastic and Myeloid Leukemia

Elisabeth Koller 3rd Medical Dept., Center for Hematology and Oncology, Hanusch Hospital, Vienna, Austria

Supplementary Appendix to manuscript submitted by Trappe, R.U. et al:

Arecent source data meta-analysis of randomized trials in adults

RESEARCH ARTICLE. Outcomes of 1st Remission Induction Chemotherapy in Acute Myeloid Leukemia Cytogenetic Risk Groups

Kevin Kelly, MD, Phd Acute Myeloid and Lymphoid Leukemias

Standard risk ALL (and its exceptions

VYXEOS : CHEMOTHERAPY LIPOSOME INJECTION FOR ACUTE MYELOID LEUKEMIA

Transcription:

Published Ahead of Print on December 19, 2014, as doi:10.3324/haematol.2014.117267. Copyright 2014 Ferrata Storti Foundation. Cytogenetic heterogeneity negatively impacts outcomes in patients with acute myeloid leukemia by Bruno C. Medeiros, Megan Othus, Min Fang, Frederick R. Appelbaum, and Harry P. Erba Haematologica 2014 [Epub ahead of print] Citation: Medeiros BC, Othus M, Fang M, Appelbaum FR, and Erba HP. Cytogenetic heterogeneity negatively impacts outcomes in patients with acute myeloid leukemia. Haematologica. 2014; 99:xxx doi:10.3324/haematol.2014.117267 Publisher's Disclaimer.0 E-publishing ahead of print is increasingly important for the rapid dissemination of science. Haematologica is, therefore, E-publishing PDF files of an early version of manuscripts that have completed a regular peer review and have been accepted for publication. E-publishing of this PDF file has been approved by the authors. After having E-published Ahead of Print, manuscripts will then undergo technical and English editing, typesetting, proof correction and be presented for the authors' final approval; the final version of the manuscript will then appear in print on a regular issue of the journal. All legal disclaimers that apply to the journal also pertain to this production process.

Cytogenetic heterogeneity negatively impacts outcomes in patients with acute myeloid leukemia Running Head: Clonal heterogeneity as an adverse feature in AML Bruno C. Medeiros, 1 Megan Othus, 2,3 Min Fang, 3,4 Frederick R. Appelbaum 3,4 and Harry P. Erba 5 1 Stanford University School of Medicine, Stanford, CA; 2 Southwest Oncology Group Statistical Center, Seattle, WA; 3 Fred Hutchinson Cancer Research Center, Seattle, WA; 4 University of Washington, Seattle, WA; 5 The University of Alabama at Birmingham, USA Conflict of Interest Disclosures: The authors indicated no potential conflicts of interest. Acknowledgments Clinical trials registration numbers: ClinicalTrials.gov Identifier: NCT014343329; NCT01338974; NCT00899171; NCT1059734; NCT01059734; NCT00899743; NCT0143329, NCT00023777; NCT00085709; NCT01360125; NCT00004217. This work was supported in part by the following Public Health Service Cooperative Agreement grant numbers awarded by the National Cancer Institute, National Institutes of Health, Department of Health and Human Services: CA32102 and CA38926. Correspondence Bruno C. Medeiros, MD Department of Medicine, Stanford University School of Medicine, 875 Blake Wilbur Drive, room 2329, Stanford, CA 94305, USA. Phone: 650.498.6000 - Fax: 650.724.5203. E-mail: brunom@stanford.edu

ABSTRACT Clonal heterogeneity is a hallmark of malignant transformation. In acute myeloid leukemia, acquired cytogenetic abnormalities are important independent predictors of initial response to therapy, remission duration, and overall survival. It remains poorly defined, however, whether the presence of multiple cytogenetically characterized clones affects outcomes in acute myeloid leukemia. The aim of this study was to assess the prognostic impact of cytogenetic clonal heterogeneity in acute myeloid leukemia. This analysis included 1403 newly diagnosed acute myeloid leukemia patients fit for intensive chemotherapy between the ages of 15 and 88 years enrolled on SWOG protocols. The presence of multiple cytogenetic clones was found in 164 (24%) patients with abnormal karyotype. The proportion of patients with clonal heterogeneity increased with age, being present in 20% of patients younger than 40 years, but in 30% of those over age 70 (p=0.03). Clonal heterogeneity was significantly more common in association with unfavorable karyotype. Clonal heterogeneity was associated with decreased response rates and inferior event-free, relapse-free and overall survival and was confirmed as an independent predictor of poor prognosis in multivariable analysis. Subgroup analysis showed that clonal heterogeneity adds prognostic information particularly in the unfavorable karyotype group. Our results confirm the negative prognostic impact of clonal heterogeneity in acute myeloid leukemia patients with abnormal karyotype. (ClinicalTrials.gov Identifiers: NCT014343329; NCT01338974; NCT00899171; NCT1059734; NCT01059734; NCT00899743; NCT0143329, NCT00023777; NCT00085709; NCT01360125; NCT00004217)

INTRODUCTION Acquired cytogenetic abnormalities can be detected in approximately 50% of patients and are important independent predictors of initial response to therapy, remission duration, and overall survival in acute myeloid leukemia (AML),. 1,2 The commonly used cytogenetic risk classifications are in general agreement with each other, and were developed following large cooperative clinical trials. Newly diagnosed AML patients can be grouped into favorable, intermediate and poor risk prognostic categories based on the diagnostic cytogenetic abnormalities. These cytogenetic classifications did not account for the presence or absence of clonal heterogeneity. Cancer evolution is a complex adaptive process that involves genetic diversification coupled with clonal selection and subclonal expansion. 3 It has been recognized for decades that leukemias are composed of genetically heterogeneous clones, where some genomic alterations are shared by the entire tumor, but not all cancer cells demonstrate an identical genomic and cytogenetic profile. 4,5 Next generation sequencing studies in AML revealed that more than 50% of AML patients have, at least, one subclone at the time of diagnosis (average: 1.5 clones/genome). 6,7 These clonal subpopulations in an individual tumor can be morphologically and functionally distinct with differential sensitivity and/or resistance to therapeutic agents. In fact, karyotypic clonal evolution is frequently encountered at the time of AML relapse. 8 In AML, the impact of cytogenetic clonal heterogeneity, defined as the presence of two or more distinct cytogenetic clones, either related or unrelated, has not been extensively studied. Previous studies have demonstrated the positive effect of residual normal metaphases in patients with monosomal karyotype AML and the deleterious impact of residual normal metaphases in AML with translocations affecting the genes for the core binding factors. 9,10 More recently, a retrospective analysis on behalf of the Study Alliance Leukemia revealed that cytogenetic heterogeneity exists in approximately one-third of AML patients with abnormal karyotypes and that clonal heterogeneity is associated with adverse prognosis. 11 Given that the outcomes of these patients depend not only on the clinical, genetic, and molecular characteristics of AML, but also on the chemotherapeutic regimens used for treatment, we investigated the prognostic impact of clonal heterogeneity in AML patients treated in North America under SWOG clinical trials.

METHODS Study Population Classic metaphase karyotyping was performed according to routine SWOG protocols. A total of 1403 (49.8% of 2816 patients registered) previously untreated adult AML patients with evaluable cytogenetic information at diagnosis and enrolled in one of 10 successive prospective SWOG studies 1 were included in this analysis. Initial treatment consisted of standard induction therapy with cytarabine and anthracycline in most patients. For patients achieving a complete remission, consolidation therapy varied based on protocol design. In brief, patients received 1-2 cycles of standard dose cytarabine plus anthracycline after enrollment in studies activated for the first 10 to 15 years of the study period. For more recent studies, the preferred post-remission therapy was 4 cycles of high dose cytarabine. Autologous and/or allogeneic transplants were included as post-remission therapies in only a minority of patients included in these analyses. Only patients with complete and centrally reviewed cytogenetic studies were included in this analysis. Patients with the t(15;17)(q22;q12) were excluded. All participating patients gave informed consent prior to enrollment into the studies. The studies were approved by ethics committees of all participating institutions and conducted in accordance with the Declaration of Helsinki. Cytogenetic studies At diagnosis, samples from bone marrow aspirates were analyzed in SWOG-approved laboratories for cytogenetic abnormalities using standard culturing and banding techniques. Karyotype designation was in accordance to the most current version of the International System for Human Cytogenetic Nomenclature. 12 Abnormalities were considered clonal when at least 2 metaphases had the same aberration in the case of either a structural abnormality or an additional chromosome. If there was loss of a chromosome, it had to be present in at least 3 metaphase cells to be considered a clonal monosomy. Cytogenetic abnormalities were grouped according to published criteria adopted by SWOG as favorable, intermediate, unfavorable, and unknown. 13 The karyotype analysis was based on 20 or more metaphase cells for more than 90% of patients included in this analysis. Clonal heterogeneity was defined as presence of 2 or more cytogenetically abnormal clonal populations as previously described. 11 In brief; a defined ancestral clonal evolution followed either mother-daughter and/or branched patterns. In the mother-daughter pattern, a subclone displays all cytogenetic aberrations of the original clone plus additional aberration(s), which define a distinct

subclone. In a branched pattern, all subclones harbor common cytogenetic aberrations suggestive of a common ancestry, but also acquire unique additional aberrations that define them as separate subclones. Both patterns of ancestral clonal evolution were often present in the same patient. Finally, composite karyotypes are those where common ancestry cannot be unequivocally determined for the multiple clones sharing some common cytogenetic characteristics. In addition, all karyotypes were individually reviewed by the authors and only cases with complete karyotype information available were included. Outcome definitions and statistical methods Available as an online supplement

RESULTS Characteristics of AML patients with subclonal populations Metaphase analysis at diagnosis identified 683 (48.6%) patients with cytogenetic abnormalities. A single cytogenetically abnormal clone was detected in most of these patients (519/683 76%), while multiple clones (range 2 12) were seen in the remaining 24% of patients. A defined ancestral pattern of subclonal evolution was detected in over 90% of patients (mother-daughter 61%; branched 15%; and both patterns 15%) while the remaining 10% of patients demonstrated composite karyotypes. Baseline characteristics of patients with abnormal karyotypes are summarized in Table 1. The incidence of clonal heterogeneity in AML patients with abnormal karyotype increased with advancing age (p=0.03). When compared to AML patients with a single cytogenetically abnormal clone, patients with subclone formation were older (p= 0.025), were more likely to have an unfavorable karyotype (p<0.001), and more frequently had secondary AML (p=0.048). Clinical outcomes associated with cytogenetic subclonal evolution in AML Initially, we determined whether pattern of ancestral clonal heterogeneity (mother-daughter versus branched) impacted outcomes in AML patients. Univariate models failed to demonstrate an association between pattern of heterogeneity and complete remission rate (OR 1.44 (95% CI 0.56, 3.68) (p= 0.45)) or overall survival (HR 0.75 (95% CI 0.47, 1.2) (p= 0.23)). However, motherdaughter pattern was associated with trend for improvement in EFS (HR 0.66 (95% CI 0.42, 1.04) (p= 0.074)) and RFS (HR 0.38 (95% CI 0.17, 0.84) (p= 0.017)) compared to branched pattern of clonal heterogeneity. Multivariate analyses failed to demonstrate an independent association of pattern of ancestral heterogeneity and response to therapy (OR 0.96 (95% CI 0.33, 2.8) (p= 0.95)) or survival outcomes (OS- (HR 0.91 (95% CI 0.54, 1.54) (p= 0.73)), EFS- (HR 0.85 (95% CI 0.5, 1.42) (p= 0.53)), RFS- (HR 0.62 (95% CI 0.22, 1.69) (p= 0.35)). For the remaining of the analyses, both patterns of ancestral clonal heterogeneity were combined as a single cohort. Effect of clonal heterogeneity on response to therapy We then evaluated the effect of clonal heterogeneity on initial response to therapy. In univariate analysis, presence of multiple cytogenetic clones, measured quantitatively, was associated with inferior complete remission (CR) rates (Odds ratio (OR) 0.86 (95% confidence interval (CI) 0.74, 1) (p=0.043). However, in multivariable analysis adjusting for multiple prognostic cofounders the

number of cytogenetic clones was not independently associated with CR rates (OR 0.96 95% CI 0.82, 1.13) (p=0.66). Prognostic variables independently associated with lower CR rates included advanced age, ECOG PS (2+) and unfavorable karyotype. (Table 2) Similarly, in univariate analysis increasing number of cytogenetic clones in AML was associated with inferior EFS (hazard ratio (HR) 1.12 (95% CI 1.05, 1.2) (p<0.001)) and RFS (HR 1.24 (95% CI 1.07, 1.44) (p=0.0038)). In multivariable analysis, presence of two or more clones was not independently associated with EFS (HR 1.05 (95% CI 0.98, 1.14) (p=0.17)), but it demonstrated a trend towards significance for RFS (HR 1.18 (95% CI 1, 1.39) (p=0.054)). Clonal Heterogeneity and survival outcomes Next, we evaluated the impact of the number of cytogenetic clones on OS. In all patients with abnormal karyotype, the presence of, at least, one additional cytogenetic clone was associated with inferior OS (HR 1.49 (95% CI 1.23, 1.81) (p<0.001)) (Figure 1A). As previously shown, presence of clonal heterogeneity in patients with CBF-AML did not impact CR rate, EFS, RFS or OS (data not shown). However, when the analysis was limited only to patients with unfavorable risk cytogenetics, presence of one of more additional clone was also associated with worse OS (HR 1.41 (95% CI 1.12, 1.76) (p=0.003)) (Figure 1B). In univariate analysis, the presence of more than one cytogenetic clone was associated with inferior OS (HR 1.16 (95% CI 1.08, 1.25) (p<0.001)). A multivariable analysis confirmed that number of abnormal clones was independently associated with worse outcomes in AML (HR 1.09 (95% CI 1, 1.18) (p=0.039)). Other prognostic variables independently associated with survival outcomes are summarized in Table 2. Finally, we used interaction models to assess whether the association between presence of clonal heterogeneity and outcome varied by cytogenetic risk group. Clonal heterogeneity continued to be independently associated with worse survival outcomes (OS: p=0.0016, RFS: p=0.0037 and EFS: p<0.001), but not associated with CR rates (p=0.27). We did not observe any evidence of an interaction between cytogenetic risk group and clinical outcomes. (Supplemental Table 1) Impact of cytarabine dose on clinical outcomes in patients with clonal heterogeneity An exploratory analysis was conducted to determine the differential effect of cytarabine dose on response to therapy and survival. In patients with clonal heterogeneity, the use of high doses of cytarabine during induction was not independently associated with higher complete remission rates

(OR 1.54 (95% CI 0.51, 4.64) (p= 0.44)) or improved OS (HR 1.36 (95% CI 0.75, 2.46) (p= 0.31)), EFS (HR 1.06 (95% CI 0.59, 1.88) (p= 0.85)), or RFS (HR 1.8 (95% CI 0.79, 4.11) (p= 0.16)). DISCUSSION The present study was undertaken to better define the effect of cytogenetic clonal heterogeneity in AML patients with abnormal karyotype. Several previous reports have described novel cytogenetic and molecular abnormalities with prognostic importance in AML. Monosomal karyotype, initially described by the HOVON/SAKK cooperative groups, identified AML patients with particular poor outcomes. 14 Incorporation of mutational abnormalities into a cytogenetic-based classification was proposed on behalf of the European LeukemiaNet and outlined 4 separate prognostic cohorts of AML patients. 15,16,17 Activating mutations in tyrosine kinases, such as c-kit and FLT3-ITD, collaborate with the fusion proteins in core-binding factor (CBF) acute myeloid leukemia and defines cohorts of patients with increased incidence of relapse and worse outcomes. 18 Bochtler et al. on behalf of the Study Alliance Leukemia has recently demonstrated that presence of cytogenetically defined subclones identifies a cohort of AML patients with resistance to induction therapy and shortened overall survival. 11 Our results confirm these previous observations where the presence of cytogenetic clonal heterogeneity is a frequent and important prognostic factor in AML and it is independently associated with decreased response to therapy and worse outcomes, especially among patients with poor-risk cytogenetics, regardless of the chemotherapy regimens used. Also, we demonstrated an association between cytogenetic clonal heterogeneity and poor prognostic features, such as advancing age and secondary AML, further explaining the worse outcomes observed in these patients. Clonal evolution is not a new concept in cancer and leukemia. In AML, clonal heterogeneity may be a reflection of the genome-wide chromosomal instability that occurs during leukomogenesis. For example, TP53 alterations have been described in nearly 80% of AML patients with monosomal karyotype and are associated with a higher degree of genomic complexity. 19 Furthermore, inactivation of TP53 in AML has been linked to the phenomenon known as chromothripsis, which indicates acquisition of numerous rearrangements through a single catastrophic DNA event. 20 The impact of clonal heterogeneity on the outcomes in AML patients may have functional consequences. Selection of chemotherapy resistant subclonal populations following exposure to genotoxic therapies, according to Darwinian principles, represents a plausible explanation for the reduced

response rates and decreased survival associated with clonal heterogeneity. In fact, whole genome sequencing studies have demonstrated that AML relapses are commonly driven by subclonal frequency and diversity. 21 Originally described by Nowell in 1976, the classic model of clonal evolution in cancer follows a sequential acquisition of genomic alterations. 4 Our study confirms prior observations that a significant proportion (24% in this study and 32.8% in Bochtler et al.) of AML patients with abnormal karyotype have multiple clones. In our study, 90% of these cases demonstrated a clear ancestral pattern between clones (mother-daughter or branched). This proportion is somewhat higher than previous reports where approximately 60% of cases had such associations. 11 Reasons for this discrepancy could be either due to the more complex nature of cases with composite karyotypes or due to laboratory preference in describing karyotype as distinct clones or as a composite clone for simplicity. For example, SWOG discourages the use of composite karyotype whenever possible, consistent with ISCN guideline that every effort should be made to describe the subclones so that clonal evolution is made evident. Finally, although the significance of clonal heterogeneity was shown to be more profound in cases where the clones were obviously related than those that were so-called composite in the Bochter study, the small proportion (10%) of patients with composite karyotypes did not allow us to define the prognostic significance of this subtype of clonal evolution in our cohort. Also, we were unable to define the effect of different post-remission therapies on the outcomes of patients with clonal heterogeneity. Two prior studies have shown that induction regimens containing high doses of cytarabine may improve the outcomes of patients with monosomal karyotype AML, another recently described cytogenetic cohort associated with particular poor outcomes. 22,23 Our exploratory analysis failed to demonstrate any therapeutic benefit of escalation of cytarabine doses during induction in patients with clonal heterogeneity. Post-remission therapies (median 2 (range 1-4) cycles) also did not affect the overall findings of the study. It has been previously suggested that the immunologic effects of allogeneic stem cell transplantation (allohsct) may be more successful than chemotherapy in eradicating all leukemic clones in patients with clonal heterogeneity. Although, we were unable to investigate the impact of allohsct on the outcomes of our cohort of patients with multiple cytogenetic clones, early transplant evaluation remains indicated for these patients.

In summary, our results demonstrate that cytogenetically defined clonal heterogeneity is an adverse prognostic feature in AML. Patients with non-cbf abnormal karyotype AML and presence of multiple cytogenetically abnormal clones should be considered at high risk for treatment failure. High dose cytarabine containing regimens cannot improve the poor outcomes associated with cytogenetic clonal heterogeneity. Authorship Contributions: BCM and MO designed the research study and performed the research, MF validated the cytogenetic classifications, FRA and HPE lead clinical investigations. All authors analyzed the data. BCM and MO wrote the paper. All authors revised and approved the manuscript.

REFERENCES 1 Medeiros BC, Othus M, Fang M, Roulston D, Appelbaum FR. Prognostic impact of monosomal karyotype in young adult and elderly acute myeloid leukemia: the Southwest Oncology Group (SWOG) experience. Blood. 2010;116(13):2224-2228. 2 Grimwade D, Hills RK, Moorman AV et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood. 2010;116(3):354-365. 3 Greaves M, Maley CC. Clonal Evolution in Cancer. Nature. 2012; 481(7381): 306 313. 4 Nowell PC. The clonal evolution of tumor cell populations. Science. 1976;194(4260):23-28. 5 Ding L, Ley TJ, Larson DE et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by wholegenome sequencing. Nature. 2012;481(7382):506-510. 6 Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059-2074. 7 Welch JS, Ley TJ, Link DC et al. The origin and evolution of mutations in acute myeloid leukemia. Cell. 2012;150(2):264-278. 8 Garson OM, Hagemeijer A, Sakurai M et al. Cytogenetic studies of 103 patients with acute myelogenous leukemia in relapse. Cancer Genet Cytogenet. 1989;40(2):187-202. 9 Xie B, Othus M, Medeiros BC, Fang M, Appelbaum FR, Estey EH. Influence of residual normal metaphases in acute myeloid leukemia patients with monosomal karyotype. Haematologica. 2011;96(4):631-632. 10 Medeiros BC, Othus M, Fang M, Appelbaum FR, Estey EH. Impact of residual normal metaphases in core binding factor acute myeloid leukemia. Cancer. 2012;118(9):2420-2423. 11 Bochtler T, Stölzel F, Heilig CE et al. Clonal heterogeneity as detected by metaphase karyotyping is an indicator of poor prognosis in acute myeloid leukemia. J Clin Oncol. 2013;31(31):3898-3905.

12 ISCN; Shaffer LG, Slovak ML, Campbell LJ, editors. An international system for human cytogenetic nomenclature. Basel, Switzerland: S. Krager; 2009. 13 Slovak ML, Kopecky KJ, Cassileth PA et al. Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group study. Blood 2000;96(13):4075-4083. 14 Breems DA, Van Putten WL, De Greef GE et al. Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype. J Clin Oncol. 2008;26(29):4791-4797 15 Döhner H, Estey EH, Amadori S 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;115(3):453-474. 16 Mrózek K, Marcucci G, Nicolet D et al. Prognostic Significance of the European LeukemiaNet Standardized System for Reporting Cytogenetic and Molecular Alterations in Adults With Acute Myeloid Leukemia. J Clin Oncol. 2012;30(36):4515-4523 17 Röllig C, Bornhäuser M, Thiede C et al. Long-term prognosis of acute myeloid leukemia according to the new genetic risk classification of the European LeukemiaNet recommendations: evaluation of the proposed reporting system. J Clin Oncol. 2011;29(20):2758-2765. 18 Allen C, Hills RK, Lamb K et al. The importance of relative mutant level for evaluating impact on outcome of KIT, FLT3 and CBL mutations in core-binding factor acute myeloid leukemia. Leukemia. 2013;27(9):1891-1901 19 Rücker FG, Schlenk RF, Bullinger L et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome. Blood. 2012;119(9):2114-2121. 20 Rausch T, Jones DT, Zapatka M et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell. 2012;148(1-2):59-71. 21 Klco JM, Spencer DH, Miller CA et al. Functional heterogeneity of genetically defined subclones in acute myeloid leukemia. Cancer Cell. 2014;25(3):379-392.

22 Löwenberg B, Pabst T, Vellenga E et al. Cytarabine dose for acute myeloid leukemia. N Engl J Med. 2011;364(11):1027-1036. 23 Medeiros BC, Othus M, Appelbaum FR. Cytarabine dose for acute myeloid leukemia. N Engl J Med. 2011;364(22):2167-2168

Table 1. Associations between baseline characteristics of AML patients with clonal heterogeneity Characteristics 1 abnormal clone 2 or more abnormal clones p-value (n=519) (n=164) Age (years) (range) 57 (15, 84) 60 (19, 89) 0.025 Age quartiles (N)(%) 0.03 15-40 years 127 (80) 32 (20) 40-60 years 180 (78) 50 (22) 60-70 years 126 (74) 45 (26) 70-90 years 86 (70) 37 (30) WBC at diagnosis (x10 3 ) (range) 10 (0, 545) 8 (1, 137) 0.33 Platelet count (x10 3 ) (range) 48 (3, 8300) 50 (2, 1200) 0.88 Bone Marrow Blasts (%) 67 (0, 100) 62 (0,99) 0.4 Peripheral Blood Blasts (%) 30 (0, 99) 24 (0,97) 0.1 Female Gender (N) (%) 217 (42) 69 (42) 1 ECOG Performance Status (N) (%) 0.43 0-1 411 (81) 126 (78) 2+ 98 (19) 36 (22) Karyotype (N) (%) <0.001 Favorable 104 (20) 19 (12) Intermediate 70 (13) 7 (4) Unfavorable 254 (49) 124 (76) Unknown 91 (18) 14 (9) Secondary AML (N) (%) 54 (13) 27 (20) 0.048 Abbreviations: WBC: White blood cell count; ECOG: Eastern Cooperative Group.

Table 2. Multivariate Analysis of Impact of Risk Factors on response and survival outcomes. CR Rate EFS RFS OS Factors OR 95% CI p- value HR 95% CI p- value HR 95% CI p- value HR 95% CI p- value Clonal 0.96 0.82, 1.13 0.66 1.05 0.98, 1.14 0.17 1.18 1, 1.39 0.054 1.09 1, 1.18 0.039 Heterogeneity Age (years) 0.98 0.97, 0.99 <0.001 1.02 1.01, 1.02 <0.001 1.02 1.01, 1.03 <0.001 1.03 1.03, 1.04 <0.001 ECOG PS 0.45 0.28, 0.72 <0.001 1.64 1.32, 2.03 <0.001 1.45 0.99, 2.13 0.055 1.65 1.32, 2.06 <0.001 Karyotype (unfavorable risk used as reference) Favorable 6.44 3.7, 11.22 <0.001 0.34 0.26, 0.45 <0.001 0.47 0.33, 0.68 <0.001 0.28 0.2, 0.38 <0.001 Intermediate 2.09 1.2, 3.66 0.0096 0.71 0.53, 0.94 0.016 0.8 0.53, 1.23 0.32 0.68 0.5, 0.9 0.0085 Unknown 1.85 1.14, 2.99 0.013 0.59 0.45, 0.76 <0.001 0.58 0.38, 0.86 0.0076 0.49 0.37, 0.64 <0.001 CR: Complete remission; EFS: Event free survival; RFS: Relapse free survival; OS: Overall survival; OR: Odds ratio; HR: Hazard ratio; CI: Confidence interval; ECOG: Eastern Cooperative Group; PS: Performance status. - Logistic regression models included for the following prognostic cofounders: Clonal heterogeneity, age, white blood cell and platelet counts at presentation, percentage of bone marrow and peripheral blood blasts at diagnosis, gender, performance status (PS) (0-1 versus 2+) and karyotypic risk group (unfavorable used as reference)

Figure Legends A. Overall survival for all patients with abnormal karyotype according to clonal heterogeneity status B. Overall survival for patients with unfavorable risk cytogenetics according to clonal heterogeneity status

Outcome definitions and statistical methods Complete remission (CR) was defined as bone marrow blasts < 5%, the absence of blasts with Auer rods, the absence of extramedullary disease, absolute neutrophil count > 1.0 10 9 /L (1000/μL), platelet count > 100 10 9 /L (100 000/μL), and independence of red cell transfusions. Overall survival (OS) was measured from the patient's date of entry onto the clinical trial until death from any cause, with observations censored at the date of last contact for patients last known to be alive. Event-free survival (EFS) was defined as the time from patient's date of entry onto the clinical trial until the first of completion of protocol therapy without documentation of CR, relapse from CR, or death from any cause. Patients last known to be alive and in CR were censored at date of last contact. Relapse free survival (RFS) was defined for patients who achieved a CR as the time from date of CR to date of relapse from CR or death from any cause. Patients last known to be alive and in CR were censored at the date of last contact. Wilcox-rank sum test were used to test associations between quantitative covariates and Fisher s exact test was used to test associations between categorical covariates. The Kaplan-Meier method was used to estimate survival curves. i Log-rank tests were used to assess differences between survival curves. Cox regression ii was used to evaluate the endpoints OS, EFS, and RFS and logistic regression was used to evaluate CR rate. Regression models adjusted for potential confounders including clonal heterogeneity, the quantitative covariates age, white blood cell and platelet counts at presentation, percentage of bone marrow and peripheral blood blasts at diagnosis, and the categorical covariates gender, performance status (PS) (0-1 versus 2+) and karyotypic risk group (unfavorable used as reference). For these analyses, patients with one cytogenetic clone were compared to patients with two or more cytogenetic clones. All P values are 2-sided with a significance level of 0.05. REFERENCES i Kaplan EL, Meier P. Nonparametric estimation from incomplete observations. J Am Stat Assoc 1958;52(282):457-481. ii Cox DR. Regression models and life-tables (with discussion). J R Stat Soc, Series B 1972;34(2):187-220.

Supplemental Table 1. Logistic regression model demonstrating lack of interaction between clonal heterogeneity and cytogenetic risk group. CR Rate EFS RFS OS Factors OR 95% CI p-value HR 95% CI p-value HR 95% CI p-value HR 95% CI p-value Clonal Heterogeneity 0.78 0.5, 1.22 0.27 1.39 1.11, 1.73 0.0037 1.91 1.3, 2.81 <0.001 1.44 1.15, 1.8 0.0016 Karyotype (unfavorable risk used as reference) Favorable 5.65 3.31, 9.63 <0.001 0.34 0.26, 0.46 <0.001 0.44 0.3, 0.64 <0.001 0.23 0.16, 0.32 <0.001 Intermediate 1.8 1.06, 3.07 0.031 0.81 0.61, 1.07 0.13 1.03 0.68, 1.56 0.9 0.79 0.59, 1.05 0.1 Unknown 1.69 1.04, 2.74 0.033 0.65 0.5, 0.84 0.0012 0.67 0.44, 1.02 0.063 0.58 0.44, 0.76 <0.001 Karyotype/Clonal heterogeneity interaction Favorable 1.3 0.36, 4.67 0.69 0.72 0.37, 1.42 0.34 0.52 0.21, 1.27 0.15 0.88 0.41, 1.87 0.74 Intermediate 1.45 0.28, 7.4 0.66 0.73 0.31, 1.74 0.48 0.67 0.19, 2.34 0.53 0.57 0.22, 1.47 0.25 Unknown 20.8 0.59, 7.26 0.25 0.69 0.35, 1.34 0.27 0.65 0.25, 1.69 0.37 0.51 0.25, 1.07 0.074 Abbreviations: CR: Complete remission; EFS: Event free survival; RFS: Relapse free survival; OS: Overall survival; OR: Odds ratio; HR: Hazard ratio; CI: Confidence interval.