• No results found

VU Research Portal

N/A
N/A
Protected

Academic year: 2021

Share "VU Research Portal"

Copied!
17
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Clinical aspects in pediatric acute myeloid leukemia

Klein, K.

2020

document version

Publisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA)

Klein, K. (2020). Clinical aspects in pediatric acute myeloid leukemia.

General rights

Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain

• You may freely distribute the URL identifying the publication in the public portal ? Take down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

E-mail address:

(2)
(3)

General introduction and outline of this thesis

Parts of the introduction are adapted from:

Klein K, de Haas V, Kaspers GJL. Clinical Challenges in de Novo Pediatric Acute Myeloid Leukemia. Expert Rev Anticancer Ther. 2018 Mar;18(3):277-293.

(4)
(5)

Although the overall incidence in children is low, cancer is the most frequent non-accidental cause of death during childhood in most developed countries. The global age-standardized incidence rate is 140.6 per million person-year in children aged 0-14 years. Incidences vary between and within cancer type, age, sex, specific regions, and racial and ethnic group. The most common forms of childhood cancer are leukemias, followed by tumors of the central nervous system (CNS), and lymphomas.1 In the

Netherlands, 500-600 children are diagnosed with cancer every year.2 Figure 1.1 shows

an overview of the distribution of childhood cancers in the Netherlands.

Figure 1.1 | Distribution of childhood cancers in The Netherlands

Approximately 150 of these patients (25%) are diagnosed with de novo leukemia. The far majority (80%) concerns acute lymphoblastic leukemia (ALL). Acute myeloid leukemia (AML) accounts for approximately 15% of the pediatric leukemias. Both types of leukemia arise from the hematopoietic stem cell, however at different levels (see Figure 1.2). Chronic forms of leukemia are rare in children.

(6)

Chapter 1

Over the past decades the overall survival (OS) of children with leukemia has improved substantially.3 This is partly contributable to the increasing interest in scientific research

and availability of international achievements. Basic and biological studies are needed in order to understand disease etiology and to identify potential targetable pathways in leukemic cells. Translational research bridges bench to bed, which contributes to deliberate decision making on how to proceed to novel clinical trials. Lastly, clinical research is of invaluable importance to proof concepts and to enable the development of evidence-based guidelines in clinical practice.

Figure 1.2 | Cell lines involved in acute myeloid leukemia and acute lymphoblastic leukemia

Epidemiology and outcome of AML

Nowadays, survival rates of ALL exceed 90%4, whereas long-term OS in pediatric AML

is 75%3,5 at best. However, specific subgroups of AML are associated with very good or

very poor survival.6 Inferior outcomes mainly derives from a high relapse rate.3,6 With an

incidence of approximately 0.8 : 100 000, AML is a rare disease in childhood.7

In the Netherlands, all cases of pediatric AML are registered by the Dutch Childhood Oncology Group (DCOG). The DCOG functions as a central reference laboratory for diagnostics of all childhood leukemias, and in a centralized way, they register all childhood leukemia patients (as well as patients with other childhood malignancies) of whom they receive diagnostic and follow-up material for review. Furthermore, as

(7)

the trial office for pediatric oncology studies in the Netherlands, they are involved in the development and monitoring of almost all childhood cancer treatment protocols. In the Netherlands approximately 20 to 25 children with AML are diagnosed annually. Population-based incidence rates, survival rates, and epidemiological data over time are, however, not publically available for Dutch patients (this thesis).

Etiology

A series of changes in the DNA of the hematopoietic precursor stem cell alter normal hematopoietic cell growth and differentiation. Class I mutations confer proliferative and/or survival advantages, whereas class II mutations primarily impair differentiation.8

However, pathogenesis is much more complicated, as recent studies identified many novel (epi)genetic alterations and driver-mutations aside from the previously well-known large chromosomal events.3,9-12 A combination of these events usually results in

the accumulation of abnormal, immature myeloid cells in the bone marrow, followed by accumulation in the peripheral blood. Malignant blasts can also accumulate in the cerebral spinal fluid or extramedullary as gingiva hyperplasia or chloromas of the skin. Contrary to in adults, pediatric AML mainly involves de novo disease. AML secondary to myelodysplastic syndrome is rare. Some germline affecting diseases, e.g., Fanconi anemia or Li-Fraumeni syndrome, are associated with an increased risk of AML, as is Down syndrome. However, in the majority of the children the exact underlying etiology or cause of AML remains unknown.

Clinical aspects and diagnostics

Suspicion of AML could yet arise from a thorough history and physical exam. Clinical patterns derive from accumulation of malignant blasts in the bone marrow and subsequent repression of healthy cell lines. Common clinical signs include fatigue, paleness, fever, bleeding tendency, and joint/bone pains. Swollen lymph nodes, an enlarged liver and/or spleen, and cutaneous manifestations may underline the diagnosis. In high-income countries, diagnosis is based on morphological assessment, immunophenotype, tumor cytogenetics, and molecular biology of bone marrow and/ or peripheral blasts. Final diagnosis is made by confirmation of at least 20% malignant myeloid blasts in the peripheral blood and/or bone marrow, and/or (in case of lower percentages) the presence of well-defined AML-specific chromosomal and/or molecular abnormalities.6,13 Cerebrospinal fluid is studied for the presence of CNS involvement of

(8)

Chapter 1

Cytomorphology

Initially, AML was classified according to the French-American-British (FAB) classification, based on cytomorphology and –to some extent– immunophenotype profiles of proliferating blasts.14-16 This classification system has been replaced by the World Health

Organization (WHO) classification, based on recurrent cytogenetic and molecular abnormalities.13,17 However, in case of absent recurrent abnormalities (“AML not further

specified") descriptive definitions based on morphological features are still used.13 The

suspicion of acute promyelocytic leukemia (APL) is usually based on morphology as well. Furthermore, there are other associations between morphology and cytogenetics.

Immunophenotyping

Multiparameter flow cytometry is used to distinguish between AML and ALL, to determine the leukemic cell differentiation and maturation, and to assess the leukemia associated immunophenotype (LAIP), useful for both diagnosis and follow-up. According to the WHO classification, lineage assignment criteria include MPO-positivity or monocytic differentiation with at least two of the following: nonspecific esterase cytochemistry, CD11c, CD14, CD64, lysosome. Aside from diagnostic purposes, the LAIP can be used for response measurements of minimal residual disease (MRD) after the onset of treatment and for detection of an emerging relapse.

Cytogenetics and molecular aberrations

Numerous studies have shown the importance of cytogenetic characterization as an independent prognostic factor in both adults and children with AML.18-25 The WHO

classification13,17 and pediatric-specific recommendations6 include several predefined,

recurrent cytogenetic subgroups at diagnosis, useful for risk-group stratification. Core-binding factor (CBF) leukemias t(8;21)(q22;q22)/RUNX1-RUNX1T1 [t(8;21)] and inv(16) (p13.1q22)/t(16;16)(p13.1;q22)/CBFB-MYH11 [inv(16)/t(16;16)] are associated with favorable prognosis.6,13,21,22 However, more specific risk-group stratification within these

subgroups might be in place (this thesis). On a molecular level, AML with mutations in the nucleophosmin 1 gene (NPM1)26 or CEBPα gene27 were more recently acknowledged

as favorable aberrations. Approximately 30-40% of the pediatric AML patients harbor favorable aberrations.6 Acute promyelocytic leukemia (APL), typically characterized by

FAB-type M3 and t(15;17)(q22;q21)/PML-RARA, is also considered favorable-risk, but is considered a distinct disease entity within the group of myeloid malignancies.6,28 APL

requires different treatment strategies and was beyond the scope of this thesis.

Aberrations t(6;9)(p23;q34), monosomy 5/del(5q), monosomy 7, t(9;11)(p22;q23), and t(10;11)(p12;q23) KMT2A (previously known as MLL) rearrangements are typically associated with poor outcome.6,13,21,22 However, not all of these adverse aberrations

(9)

are included as risk-stratificating aberration for treatment by different international study groups (this thesis). On a molecular level, internal duplications of the FLT3-gene (FLT3-ITD)29,30 and WT1 mutations31,32 are associated with a high relapse risk and poor

outcome. The prognostic impact of FLT3-ITD is influenced by the allelic ratio, with higher ratios being associated with poorer outcomes.29,30 Contrary, low allelic burden and

concomitant NPM1 mutations are associated with intermediate outcome.33,34

Altogether, AML is a molecularly heterogeneous disease, which becomes more and more evident with next-generation sequencing technologies. These novel techniques provide numerous of new insights into the biology of AML, with the potential to find targets for therapeutic intervention.

Risk-based treatment in de novo AML

Risk-group stratification followed by risk-based treatment is applied by all international pediatric AML groups. Defining parameters include cytogenetics, a few molecular aberrations, and treatment response. Most international study groups use

FLT3-ITD as a high-risk marker for treatment adjustments. However, some groups include

concomitant NPM1 and/or allelic ratio, whereas others do not. With regard to other well-known adverse aberrations, consequences for treatment are less uniform.

Apart from cytogenetic and molecular aberrations, response and MRD detection early during therapy are the most important predictors for outcome. Traditionally, morphological response after one or two courses of chemotherapy, i.e., achieving complete remission (CR), was and still is considered the most important outcome predictor. CR is defined as less than 5% malignant blasts in the bone marrow, absence of blasts with Auer rods, absence of extramedullary disease, combined with an absolute neutrophil count of at least 1.0×109/L (1000/μL), platelets above 80×109 (80,000/μL),

and independence of red blood cell transfusions.6 More recently, the LAIP defined by

multiparameter flow cytometry was added to routine response assessment in several protocols. The development in current flow technologies allows for the detection of MRD at a sensitivity of at least 1×10-3 in over 90% of the patients. Multiple study groups

have proven the predictive value of flow-MRD in respect to relapse risk and subsequent outcome.35-40 Furthermore, the leukemic stem cell burden at diagnosis has recently

been reported as a potential marker to identify young AML patients at risk of treatment failure.41

Due to the heterogeneity of the disease and the intensity of the treatment with subsequent side-effects and persistent unsatisfactory relapse rates, working in the field of pediatric AML is full of clinical challenges (this thesis). In general, pediatric AML treatment consists of four or five courses of chemotherapy with high doses of cytarabine

(10)

Chapter 1

and anthracyclines as backbone, complemented by other agents.6 Potential additional

agents include etoposide, 6-thioguanine, amsacrine, and gemtuzumab ozogamicin (GO). However, the additive value of most of these agents continues to be an important point of discussion. Classic agents like anthracyclines are now available in liposomal forms, such as liposomal daunorubicin. These liposomal forms may be administered in higher doses with good effect and similar, or even reduced, toxicity (this thesis).5 Although

major efforts are being undertaken to unravel targetable pathways for individualized therapy, the few novel agents that have been studied in phase I/II trials have not shown the results we would hope for.3 Subsequently, the far majority of pediatric AML

patients is still being treated with cytarabine and anthracyclines, sometimes followed by allogeneic hematopoietic stem cell transplantation (allo-HSCT) in first CR (CR1).3

However, the role of allo-HSCT in CR1 has been a point of debate for decades now. 42-44 Although the anti-leukemic effect of HSCT has been established in multiple studies,

efficacy needs to be balanced against HSCT-related mortality and morbidity, especially when considering the relatively good results obtained with chemotherapy alone.45-48

Relapsed AML

Despite intensified treatment, still 25-35% of the pediatric patients with AML relapses.3,49

Although most relapsed patients respond to reinduction therapy, many patients have a second relapse or die due to treatment or HSCT-related toxicity. This results in OS rates after relapse of at best 40%.50 Most important risk factors for relapsed patients include

time to relapse, early treatment response after reinduction therapy, and the cytogenetic profile at time of relapse.51 Whereas complicated risk stratification is applied at initial

diagnosis, most relapse protocols lack risk-based treatment approaches. However, molecular and cytogenetic changes between diagnosis and relapse, explained by either clonal evolution or clonal selection of preexisting minor clones, are frequent events and potentially of prognostic interest (this thesis).52,53

Toxicity and Supportive Care

Aside from leukemia-related death, approximately 3-5% of the patients dies while being in CR54,55 and this number is even higher when including treatment with

allo-SCT in CR148. Main causes of death in CR include infection or bleeding.54,55 Furthermore,

treatment-related morbidity has an important effect on the quality of life of all patients. The most frequently reported forms of severe toxicity include transfusion dependency, febrile neutropenia and infections, gastrointestinal toxicity (including mucositis), and cardiotoxicity. However, up-to-date information on toxicity and treatment-related mortality (TRM) is lacking for Dutch pediatric AML patients (this thesis). Internationally, TRM has improved over time due to the increased interest in supportive care, but

(11)

overall, published studies on supportive care are often limited by retrospective designs and/or small patients numbers. Subsequently, available guidelines mainly involve a more general population of children with cancer, without detailed specifications per disease.56,57 Chemo-induced profound neutropenia, disruption of skin and mucosal

barriers, multiple invasive procedures, the use of a central venous line, and hospitalization in general all make pediatric AML patients at high risk for severe infections. Whereas pharmacological prophylaxis with fluoroquinolones and antifungal agents is generally recommended for adult patients with AML58, the role of prophylactic antibiotics in

pediatric AML remains uncertain.59 Despite appeals by experts and working groups in

the field to harmonize guidelines, many divergent regimens among hospitals and study groups seem to exist (this thesis).60-62 Prospective randomized trials in this field are very

much needed (this thesis).

Late effects in survivors

Like many anticancer regimens, AML treatment is associated with late effects that can impair both physical and psychological health at an older age. Up to half of the childhood AML survivors report a chronic medical condition in adulthood.63 Important

late effects among AML survivors include neurocognitive and endocrine abnormalities. Especially high doses of anthracyclines are associated with (late) cardio-toxicity. With the increasing survival, the interest in childhood cancer survivor research has increased substantially. However, long-term follow-up is yet often lacking.

Outline of this thesis

International collaborations are needed for high-quality research on all aspects of pediatric AML, acknowledging the importance of biological, translational, clinical and quality of life studies in this vulnerable population. Several of the studies described in this thesis have been carried out within the international Berlin-Frankfurt-Munster (I-BFM) study group (SG).

This thesis includes several translational and clinical projects related to cytogenetics and clinical aspects of pediatric AML. The aim of all projects outlined in this thesis was to provide more insight in either risk stratification and/or clinical aspects of AML treatment. The first part of this thesis focusses on epidemiology, outcomes, and mortality of children with AML in the Netherlands. Chapter 2 describes the incidence and outcome of pediatric AML in the Netherlands over the past 26 years. Findings described in Chapter

3 are the results of the first project as part of a large national study on toxicity among

children recently treated for AML in the Netherlands, and includes incidence and causes of treatment-related mortality.

(12)

Chapter 1

The second part of this thesis focusses on clinical outcome among different cytogenetic subgroups of pediatric AML. Chapters 4-7 focus on the prognostic impact of different cytogenetic aberrations. Initially, risk-group stratification was based on the cytomorphology-based FAB classification.14-16

FAB type AML-M4 with eosinophilia (M4eo) has been associated with better outcome compared to other subgroups in children.64 M4eo morphology has been associated

with the presence of inv(16)/t(16;16) in adults.65 In Chapter 4 the prognostic and clinical

consequences of the presence of eosinophilia morphology in relation to inv(16)/t(16;16) is explored in children with AML-M4.

Aberrations inv(16) and t(16;16) result in the same fusion of the CBFβ subunit and the Myosin-11 heavy chain gene (MYH11).66 This fusion results in a subtype of myeloid

leukemias associated with a favorable prognosis.6,67,68 However, CBFβ-MYH11 fusion

transcripts are heterogeneous and may vary in biological activity.69,70 A few risk factors

have been identified in inv(16)/t(16;16) adult cohorts, but little is known about pediatric t(16;16)-AML patients specifically. The international cohort study described in Chapter

5 evaluated potential prognostic features and outcome in this specific subgroup of

CBF-leukemia patients.

Chapter 6 describes the largest international cohort of pediatric AML patients

with a t(8;21)-aberration, generally classified as favorable-risk. The importance of additional cytogenetic aberrations and the role of different doses of the mainly used chemotherapeutic agents are addressed.

The biological aspects and prognostic impact of cytogenetics and molecular aberrations at diagnosis have been well studied, but little is known about the prognostic significance of cytogenetics, chromosomal instability, and clonal evolution at the time of relapse in pediatric AML. Chapter 7 describes an international effort that aims to provide more insight in the prognostic impact of cytogenetic aberrations at time of relapse.

The third part of this thesis focusses on toxicity and supportive care. With increasing survival rates, toxicity, impact on quality of life, and late effects of initial treatment become of increasing importance.

One of the most important types of myelosuppressive agents concerns anthracyclines. Similar to many drugs used in the treatment of oncologic disease, anthracyclines also inflict damage to many other healthy cells. One of the most important side effects includes cardiotoxicity.71,72 In order to reduce especially this dose-limiting toxicity,

liposomal forms of anthracyclines have been developed. Chapter 8 reviews several aspects of one of the most frequently used liposomal forms of anthracyclines in pediatric AML: liposomal daunorubicin.

(13)

Glucocorticoids (GCs) are mainly being used as anti-leukemic drugs in the treatment of ALL, since these cells are much more sensitive to glucocorticoid (GC)-induced cell kill than AML cells.73,74 However, some study groups still use GCs as an anti-leukemic agent75,

whereas other physicians prescribe the GC dexamethasone as supportive anti-emetic drug. Furthermore, GCs are sometimes used as anti-inflammatory drugs during AML treatment, or as part of post-SCT regimens. The aim of the study described in Chapter 9 was to confirm in vitro resistance patterns in pediatric AML, but also to identify potential risks of increased blast proliferation associated with GC exposure.

Intensification of treatment protocols and high doses of chemo have led to an increase of treatment-related complications, with severe infections being one of the most important risks. Despite appeals by experts and working groups in the field to harmonize infection prophylactic guidelines, many divergent regimens among hospitals and study groups seem to exist.60-62 We evaluated the currently available infection prophylaxis

guidelines among international pediatric AML study groups affiliated with the I-BFM-SG, as well as hospital-based guidelines among hospitals affiliated to the NOPHO-DBH AML Study Group. An overview of the results of this survey are presented in Chapter 10. During our repeated discussions on the use of antibiotic prophylaxis and the emerging need for prospective randomized trials, a novel study was set up. The aim of this study is to evaluate whether the use of the glycopeptic antibiotic teicoplanin reduces the number of specific bacterial infections, i.e., Viridans Group Streptococci, in children treated for AML. The concept synopsis of this trial design is presented in Chapter 11. Finally, Chapter 12 provides an overview of the clinical challenges when treating patients with AML, including cytogenetic and molecular diagnostics, the current risk-group stratification, and the fragile balance between efficacy and toxicity.

In the general discussion (Chapter 13) the results of the studies described in this thesis are summarized and discussed in relation to current literature, concluded by future perspectives.

(14)

Chapter 1

REFERENCES

1. Steliarova-Foucher E, Colombet M, Ries LAG, et al. International incidence of childhood cancer, 2001-10: a population-based registry study. Lancet Oncol. 2017;18(6):719-731.

2. Dutch Childhood Oncology Group. Dutch Childhood Oncology Annual Report 2016 (SKION jaarverslag). 2016.

3. Zwaan CM, Kolb EA, Reinhardt D, et al. Collaborative Efforts Driving Progress in Pediatric Acute Myeloid Leukemia. J Clin Oncol. 2015;33(27):2949-2962.

4. Pieters R, de Groot-Kruseman H, Van der Velden V, et al. Successful Therapy Reduction and Intensification for Childhood Acute Lymphoblastic Leukemia Based on Minimal Residual Disease Monitoring: Study ALL10 From the Dutch Childhood Oncology Group. J Clin Oncol. 2016;34(22):2591-2601.

5. Creutzig U, Zimmermann M, Bourquin JP, et al. Randomized trial comparing liposomal daunorubicin with idarubicin as induction for pediatric acute myeloid leukemia: results from Study AML-BFM 2004.

Blood. 2013;122(1):37-43.

6. Creutzig U, van den Heuvel-Eibrink MM, Gibson B, et al. Diagnosis and management of acute myeloid leukemia in children and adolescents: recommendations from an international expert panel. Blood. 2012;120(16):3187-3205.

7. Surveillance Research Program NCI. 2018. Fast Stats An interactive tool for access to SEER cancer statistics, http://seer.cancer.gov/faststats <http://seer.cancer.gov/faststats >. Accessed 2018 May 17. 8. Kelly LM, Gilliland DG. Genetics of myeloid leukemias. Annu Rev Genomics Hum Genet. 2002;3:179-198. 9. Cancer Genome Atlas Research N, Ley TJ, Miller C, et al. Genomic and epigenomic landscapes of adult de

novo acute myeloid leukemia. N Engl J Med. 2013;368(22):2059-2074.

10. Metzeler KH, Herold T, Rothenberg-Thurley M, et al. Spectrum and prognostic relevance of driver gene mutations in acute myeloid leukemia. Blood. 2016;128(5):686-698.

11. Papaemmanuil E, Gerstung M, Bullinger L, et al. Genomic Classification and Prognosis in Acute Myeloid Leukemia. N Engl J Med. 2016;374(23):2209-2221.

12. Bullinger L, Dohner K, Dohner H. Genomics of Acute Myeloid Leukemia Diagnosis and Pathways. J Clin

Oncol. 2017;35(9):934-946.

13. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391-2405.

14. Bennett JM, Catovsky D, Daniel MT, et al. Proposed revised criteria for the classification of acute myeloid leukemia. A report of the French-American-British Cooperative Group. Ann Intern Med. 1985;103(4):620-625.

15. Bennett JM, Catovsky D, Daniel MT, et al. Criteria for the diagnosis of acute leukemia of megakaryocyte lineage (M7). A report of the French-American-British Cooperative Group. Ann Intern Med. 1985;103(3):460-462.

16. Bennett JM, Catovsky D, Daniel MT, et al. Proposal for the recognition of minimally differentiated acute myeloid leukaemia (AML-MO). Br J Haematol. 1991;78(3):325-329.

17. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937-951.

18. 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.

19. Grimwade D, Walker H, Oliver F, et al. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1,612 patients entered into the MRC AML 10 trial. The Medical Research Council Adult and Children’s Leukaemia Working Parties. Blood. 1998;92(7):2322-2333.

(15)

20. Byrd JC, Mrozek K, Dodge RK, et al. Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461). Blood. 2002;100(13):4325-4336.

21. von Neuhoff C, Reinhardt D, Sander A, et al. Prognostic impact of specific chromosomal aberrations in a large group of pediatric patients with acute myeloid leukemia treated uniformly according to trial AML-BFM 98. J Clin Oncol. 2010;28(16):2682-2689.

22. Harrison CJ, Hills RK, Moorman AV, et al. Cytogenetics of childhood acute myeloid leukemia: United Kingdom Medical Research Council Treatment trials AML 10 and 12. J Clin Oncol. 2010;28(16):2674-2681. 23. Weltermann A, Fonatsch C, Haas OA, et al. Impact of cytogenetics on the prognosis of adults with de

novo AML in first relapse. Leukemia. 2004;18(2):293-302.

24. Kumar CC. Genetic abnormalities and challenges in the treatment of acute myeloid leukemia. Genes

Cancer. 2011;2(2):95-107.

25. 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.

26. Hollink IH, Zwaan CM, Zimmermann M, et al. Favorable prognostic impact of NPM1 gene mutations in childhood acute myeloid leukemia, with emphasis on cytogenetically normal AML. Leukemia. 2009;23(2):262-270.

27. Ho PA, Alonzo TA, Gerbing RB, et al. Prevalence and prognostic implications of CEBPA mutations in pediatric acute myeloid leukemia (AML): a report from the Children’s Oncology Group. Blood. 2009;113(26):6558-6566.

28. Mi JQ, Li JM, Shen ZX, Chen SJ, Chen Z. How to manage acute promyelocytic leukemia. Leukemia. 2012;26(8):1743-1751.

29. Zwaan CM, Meshinchi S, Radich JP, et al. FLT3 internal tandem duplication in 234 children with acute myeloid leukemia: prognostic significance and relation to cellular drug resistance. Blood. 2003;102(7):2387-2394.

30. Meshinchi S, Alonzo TA, Stirewalt DL, et al. Clinical implications of FLT3 mutations in pediatric AML.

Blood. 2006;108(12):3654-3661.

31. Hollink IH, van den Heuvel-Eibrink MM, Zimmermann M, et al. Clinical relevance of Wilms tumor 1 gene mutations in childhood acute myeloid leukemia. Blood. 2009;113(23):5951-5960.

32. Ho PA, Zeng R, Alonzo TA, et al. Prevalence and prognostic implications of WT1 mutations in pediatric acute myeloid leukemia (AML): a report from the Children’s Oncology Group. Blood. 2010;116(5):702-710.

33. Pratcorona M, Brunet S, Nomdedeu J, et al. Favorable outcome of patients with acute myeloid leukemia harboring a low-allelic burden FLT3-ITD mutation and concomitant NPM1 mutation: relevance to post-remission therapy. Blood. 2013;121(14):2734-2738.

34. Shamaa S, Laimon N, Aladle DA, et al. Prognostic implications of NPM1 mutations and FLT3 internal tandem duplications in Egyptian patients with cytogenetically normal acute myeloid leukemia.

Hematology. 2014;19(1):22-30.

35. Sievers EL, Lange BJ, Buckley JD, et al. Prediction of relapse of pediatric acute myeloid leukemia by use of multidimensional flow cytometry. J Natl Cancer Inst. 1996;88(20):1483-1488.

36. Sievers EL, Lange BJ, Alonzo TA, et al. Immunophenotypic evidence of leukemia after induction therapy predicts relapse: results from a prospective Children’s Cancer Group study of 252 patients with acute myeloid leukemia. Blood. 2003;101(9):3398-3406.

37. van der Velden VH, van der Sluijs-Geling A, Gibson BE, et al. Clinical significance of flowcytometric minimal residual disease detection in pediatric acute myeloid leukemia patients treated according to the DCOG ANLL97/MRC AML12 protocol. Leukemia. 2010;24(9):1599-1606.

(16)

Chapter 1

38. Rubnitz JE, Inaba H, Dahl G, et al. Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: results of the AML02 multicentre trial. Lancet Oncol. 2010;11(6):543-552.

39. Loken MR, Alonzo TA, Pardo L, et al. Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia: a report from Children’s Oncology Group. Blood. 2012;120(8):1581-1588.

40. Tierens A, Bjorklund E, Siitonen S, et al. Residual disease detected by flow cytometry is an independent predictor of survival in childhood acute myeloid leukaemia; results of the NOPHO-AML 2004 study. Br J

Haematol. 2016;174(4):600-609.

41. Hanekamp D, Denys B, Kaspers GJL, et al. Leukaemic stem cell load at diagnosis predicts the development of relapse in young acute myeloid leukaemia patients. Br J Haematol. 2018;183(3):512-516.

42. Creutzig U, Reinhardt D. Current controversies: which patients with acute myeloid leukaemia should receive a bone marrow transplantation?--a European view. Br J Haematol. 2002;118(2):365-377. 43. Chen AR, Alonzo TA, Woods WG, Arceci RJ. Current controversies: which patients with acute myeloid

leukaemia should receive a bone marrow transplantation?--an American view. Br J Haematol. 2002;118(2):378-384.

44. Hasle H. A critical review of which children with acute myeloid leukaemia need stem cell procedures. Br

J Haematol. 2014;166(1):23-33.

45. Bleakley M, Lau L, Shaw PJ, Kaufman A. Bone marrow transplantation for paediatric AML in first remission: a systematic review and meta-analysis. Bone Marrow Transplant. 2002;29(10):843-852.

46. Burnett AK, Hills RK, Milligan DW, et al. Attempts to optimize induction and consolidation treatment in acute myeloid leukemia: results of the MRC AML12 trial. J Clin Oncol. 2010;28(4):586-595.

47. Hasle H, Kaspers GJ. Strategies for reducing the treatment-related physical burden of childhood acute myeloid leukaemia - a review. Br J Haematol. 2017;176(2):168-178.

48. Niewerth D, Creutzig U, Bierings MB, Kaspers GJ. A review on allogeneic stem cell transplantation for newly diagnosed pediatric acute myeloid leukemia. Blood. 2010;116(13):2205-2214.

49. Kaspers G. How I treat paediatric relapsed acute myeloid leukaemia. Br J Haematol. 2014;166(5):636-645. 50. Kaspers GJ, Zimmermann M, Reinhardt D, et al. Improved outcome in pediatric relapsed acute myeloid

leukemia: results of a randomized trial on liposomal daunorubicin by the International BFM Study Group. J Clin Oncol. 2013;31(5):599-607.

51. Creutzig U, Zimmermann M, Dworzak MN, et al. The prognostic significance of early treatment response in pediatric relapsed acute myeloid leukemia: results of the international study Relapsed AML 2001/01.

Haematologica. 2014;99(9):1472-1478.

52. Bachas C, Schuurhuis GJ, Hollink IH, et al. High-frequency type I/II mutational shifts between diagnosis and relapse are associated with outcome in pediatric AML: implications for personalized medicine.

Blood. 2010;116(15):2752-2758.

53. Kim Y, Jang J, Hyun SY, et al. Karyotypic change between diagnosis and relapse as a predictor of salvage therapy outcome in AML patients. Blood Res. 2013;48(1):24-30.

54. Creutzig U, Zimmermann M, Reinhardt D, Dworzak M, Stary J, Lehrnbecher T. Early deaths and treatment-related mortality in children undergoing therapy for acute myeloid leukemia: analysis of the multicenter clinical trials AML-BFM 93 and AML-BFM 98. J Clin Oncol. 2004;22(21):4384-4393.

55. Slats AM, Egeler RM, van der Does-van den Berg A, et al. Causes of death--other than progressive leukemia--in childhood acute lymphoblastic (ALL) and myeloid leukemia (AML): the Dutch Childhood Oncology Group experience. Leukemia. 2005;19(4):537-544.

56. Lehrnbecher T, Phillips R, Alexander S, et al. Guideline for the management of fever and neutropenia in children with cancer and/or undergoing hematopoietic stem-cell transplantation. J Clin Oncol. 2012;30(35):4427-4438.

57. Loeffen EA, Kremer LC, Mulder RL, et al. The importance of evidence-based supportive care practice guidelines in childhood cancer-a plea for their development and implementation. Support Care Cancer. 2017;25(4):1121-1125.

(17)

58. Gafter-Gvili A, Fraser A, Paul M, et al. Antibiotic prophylaxis for bacterial infections in afebrile neutropenic patients following chemotherapy. Cochrane Database Syst Rev. 2012;1:CD004386.

59. Alexander S, Nieder M, Zerr DM, Fisher BT, Dvorak CC, Sung L. Prevention of bacterial infection in pediatric oncology: what do we know, what can we learn? Pediatr Blood Cancer. 2012;59(1):16-20. 60. Lehrnbecher T, Ethier MC, Zaoutis T, et al. International variations in infection supportive care practices

for paediatric patients with acute myeloid leukaemia. Br J Haematol. 2009;147(1):125-128.

61. Lehrnbecher T, Aplenc R, Rivas Pereira F, et al. Variations in non-pharmacological anti-infective measures in childhood leukemia--results of an international survey. Haematologica. 2012;97(10):1548-1552. 62. Loeffen EA, Mulder RL, van de Wetering MD, et al. Current variations in childhood cancer supportive care

in the Netherlands. Cancer. 2016;122(4):642-650.

63. Mulrooney DA, Dover DC, Li S, et al. Twenty years of follow-up among survivors of childhood and young adult acute myeloid leukemia: a report from the Childhood Cancer Survivor Study. Cancer. 2008;112(9):2071-2079.

64. Creutzig U, Ritter J, Niederbiermann-Koczy G, Harbott J, Schellong G. [Prognostic significance of eosinophilia in children with acute myeloid leukemia in the studies AML-BFM-78 and -83]. Klin Padiatr. 1989;201(4):220-226.

65. Le Beau MM, Larson RA, Bitter MA, Vardiman JW, Golomb HM, Rowley JD. Association of an inversion of chromosome 16 with abnormal marrow eosinophils in acute myelomonocytic leukemia. A unique cytogenetic-clinicopathological association. N Engl J Med. 1983;309(11):630-636.

66. Liu P, Tarle SA, Hajra A, et al. Fusion between transcription factor CBF beta/PEBP2 beta and a myosin heavy chain in acute myeloid leukemia. Science. 1993;261(5124):1041-1044.

67. Larson RA, Williams SF, Le Beau MM, Bitter MA, Vardiman JW, Rowley JD. Acute myelomonocytic leukemia with abnormal eosinophils and inv(16) or t(16;16) has a favorable prognosis. Blood. 1986;68(6):1242-1249.

68. Hossain MJ, Xie L, Caywood EH. Prognostic factors of childhood and adolescent acute myeloid leukemia (AML) survival: evidence from four decades of US population data. Cancer Epidemiol. 2015;39(5):720-726.

69. Shurtleff SA, Meyers S, Hiebert SW, et al. Heterogeneity in CBF beta/MYH11 fusion messages encoded by the inv(16)(p13q22) and the t(16;16)(p13;q22) in acute myelogenous leukemia. Blood. 1995;85(12):3695-3703.

70. Costello R, Sainty D, Lecine P, et al. Detection of CBFbeta/MYH11 fusion transcripts in acute myeloid leukemia: heterogeneity of cytological and molecular characteristics. Leukemia. 1997;11(5):644-650. 71. Floyd JD, Nguyen DT, Lobins RL, Bashir Q, Doll DC, Perry MC. Cardiotoxicity of cancer therapy. J Clin

Oncol. 2005;23(30):7685-7696.

72. Kremer LC, van Dalen EC, Offringa M, Voute PA. Frequency and risk factors of anthracycline-induced clinical heart failure in children: a systematic review. Ann Oncol. 2002;13(4):503-512.

73. Kaspers GJ, Pieters R, Klumper E, De Waal FC, Veerman AJ. Glucocorticoid resistance in childhood leukemia. Leuk Lymphoma. 1994;13(3-4):187-201.

74. Zwaan CM, Kaspers GJ, Pieters R, et al. Cellular drug resistance profiles in childhood acute myeloid leukemia: differences between FAB types and comparison with acute lymphoblastic leukemia. Blood. 2000;96(8):2879-2886.

75. Hicsonmez G. The effect of steroid on myeloid leukemic cells: the potential of short-course high-dose methylprednisolone treatment in inducing differentiation, apoptosis and in stimulating myelopoiesis.

Referenties

GERELATEERDE DOCUMENTEN

By dual-color live- cell imaging experiments, we show that unencapsulated pneumococci adhere significantly better to human lung epithelial cells than encapsulated strains, in

established firms. Cultures with dominant internal process model characteristics are changing towards other cultures. This may explain the presence of business model change. In

Furthermore, the initial presentation of this patient with jaundice and marked liver biochemistry abnormalities; in the absence of a pancreatic mass, bile duct obstruction,

Disease relapse remains the major cause of treat- ment failure in adults with acute myeloid leukaemia (AML) receiving allogeneic stem cell transplantation (allo-SCT) or

Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute

Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute

Hogere verliezen, tussen 5 en 15 % zijn te verwachten als, ervan uitgaande dat er geen vorst in de kuil optreedt, de buitentemperatuur relatief hoog is (gem.

Rehabilitatie bij mensen met (zeer) ernstige verstandelijke en visuele beperking. Zelfredzaamheid, kracht en balans bij mensen