Identification of Gene Expression Signatures Accurately Predicting Cytogenetic Subtypes in Pediatric Acute Myeloid Leukemia.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1509-1509 ◽  
Author(s):  
Brian V Balgobind ◽  
Marry M van den Heuvel-Eibrink ◽  
Renee X Menezes ◽  
Dirk Reinhardt ◽  
Iris H.I.M. Hollink ◽  
...  

Abstract Pediatric acute myeloid leukemia (AML) is a heterogeneous disease, which is classified according to the WHO classification, based on morphology, immunophenotyping and non-random genetic aberrations. AML is hypothesized to arise from two different types of genetic aberrations, i.e. type-I (proliferation enhancing) mutations and type-II (differentiation impairing) mutations. To detect genetic aberrations multiple techniques such as conventional karyotyping, FISH and RT-PCR are being used. In addition to conventional karyotyping, the latter two techniques revealed a higher frequency of aberrations. Still, failures or false negative results should be taken into account. Recent studies have focused on the potential of gene expression profiling (GEP) to classify acute leukemias. To study the clinical value of classification by GEP, we first used a double-loop cross validation (CV) to avoid over-fitting of GEP data and, subsequently, addressed whether the identified GEP was suitable to classify pediatric AML cases in a second independent group of cases. Affymetrix Human Genome U133 plus 2.0 microarrays were used to generate gene expression profiles of 257 children with AML, with high blast counts, if necessary, after enrichment (~80% or more) and good quality RNA. Probe set intensities were normalized using the variance-stabilizing normalization (VSN) implemented in R (version 2.2.0). The patient group was divided into a test cohort (n=170) and an independent validation cohort (n=87). The test cohort was used to construct the classifier using two levels of CV: the minimum number of predictive genes was estimated using a 10-fold CV on random subsets of about 113 (~2/3 of total) patients; the accuracy of the obtained classifier is estimated on the remaining 57 (~1/3) patients. Candidate genes to represent the GEP in the classifier were those genes that discriminated AML subtypes according to an empirical Bayes linear regression model (Bioconductor package: Limma). To construct a reliable classifier it was sufficient to use 75 probe sets, representing the top 15 discriminating probe sets for MLL-gene rearranged AML, t(8;21), inv(16), t(15;17) and t(7;12). These subtypes represented ~50% of the included patients. The remaining patients either had normal cytogenetics, random aberrations or no data available (cytogenetic failure). Due to the heterogeneity of these remaining groups discriminative probe sets were not found. This classifier could reliably predict the 5 subtypes with a median accuracy of 93%. Validation of the classifier on the independent cohort confirmed that the sensitivity and accuracy was more than 99%. No gene expression signatures could be found for the molecular aberrations NPM1, CEBPa, MLL-PTD, FLT3, C-KIT, RAS or PTPN1, possibly due to the small number of cases. However, specific gene expression signatures were found for FLT3-ITD within the subset of cases with t(15;17) or normal cytogenetics. Importantly, a high expression of HOXB-cluster related genes was found in cases with FLT3-ITD and normal cytogenetics. In conclusion, GEP can correctly predict several important cytogenetic subtypes of pediatric AML, including cases that are currently classified using different cytogenetic techniques and cases with failed cytogenetic analysis. Prospective studies are needed to validate the use of GEP in the classification of pediatric AML, especially to provide information on its utility in clinical practice. Increasing numbers in rare subtypes may result in the discovery of genes discriminative for them, and may foster GEP as a new diagnostic tool.

Haematologica ◽  
2010 ◽  
Vol 96 (2) ◽  
pp. 221-230 ◽  
Author(s):  
B. V. Balgobind ◽  
M. M. Van den Heuvel-Eibrink ◽  
R. X. De Menezes ◽  
D. Reinhardt ◽  
I. H. I. M. Hollink ◽  
...  

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2516-2516
Author(s):  
Eva A Coenen ◽  
C. Michel Zwaan ◽  
Christine J Harrison ◽  
Oskar A. Haas ◽  
Valerie de Haas ◽  
...  

Abstract Abstract 2516 Introduction: In pediatric acute myeloid leukemia (AML) cytogenetic abnormalities are important for prognosis and treatment stratification. Some recurring cytogenetic abnormalities occur so rarely that large collaborative studies are required to define their prognostic impact. t(8;16)(p11;p13) is an aberration reported in <1% of pediatric AML patients. Recently, Haferlach et al. (Leukemia, 2009) reported that t(8;16)(p11;p13) represents a distinct clinical and biological subgroup in adult AML, with dismal clinical outcome (median overall survival (OS) 4.7 months). In pediatric AML case reports are available, but studies of larger series are lacking. Methods: We collected data on patients with t(8;16)(p11;p13) (n=39) from 18 collaborative study groups belonging to the International Berlin-Frankfurt-Münster AML-Committee. Data collection included clinical characteristics, karyotype, morphology and immunophenotype, which were centrally reviewed by the co-authors. In addition, the literature was screened for reports on pediatric t(8;16)(p11;p13), which found an additional 23 cases. Results: A total of 62 pediatric AML patients with t(8;16)(p11;p13) were identified, diagnosed between 1978–2010, including 2 secondary AML cases. They had a median age of 1.2 years (range 0–17 years) and 56% of the patients were younger than 2 years old. Median white blood cell count was 21.3 × 109/l (n= 59, range 1.1–173 × 109/l). FAB type was M4 or M5 in 95% of the cases and immunophenotypes showed monocytic differentiation. Erythrophagocytosis (23/33, 70%), leukemia cutis (21/36, 58%) and disseminated intravascular coagulation (DIC, 15/38, 39%) occurred frequently. At initial diagnosis, 35/57 (61%) of the patients showed t(8;16)(p11;p13) as the sole aberration (5 incomplete karyotypes were excluded). In the remaining 22 patients, no recurrent additional cytogenetic aberrations were found. In patients diagnosed after January 1st1993, and treated with chemotherapy at initial diagnosis (n= 34), the CR rate was 85% (29/34), 5-year OS was 59% (±9%), event free survival (EFS) was 57% (±9%) and the cumulative incidence of relapse (CIR) was 28% (±8%). The median follow-up time of survivors was 4 years. Outcome was is comparable to a reference cohort of AML-BFM patients (n= 543) treated in the same era, with OS of 62% (±2%), EFS of 50% (±2%) and CIR of 42% (±2%). Most striking was that in 7 neonates (<1 month of age) diagnosed with AML and t(8;16)(p11;p13), spontaneous complete remission was achieved. Three of these infants survived without recurrence of disease (follow-up 0.9–4.0 years). For those who relapsed (0.7–4 years after initial diagnosis), chemotherapy treatment was successful in 2/4 patients. Gene expression data, using the Affymetrix Human Genome U133 plus 2.0 array, were available from 297 pediatric AML samples, of which 8 were t(8;16)(p11;p13). The gene expression signature of t(8;16)(p11;p13) AML clustered close to but distinct from MLL-rearranged AML using unsupervised analysis. Highly expressed genes included HOXA11, HOXA10, RET, PERP and GGA2, indicating a pathway in common with MLL-rearranged AML with distinct features. PERP is a direct target gene of p53 and acts as an effector of apoptosis, GGA2 codes for a protein that regulates protein trafficking within the cell. PERP and GGA2 knockdown in high expressing cell lines did not result in significant changes in cell proliferation or apoptosis, thus their function in leukemic blasts remains unknown. DNA methylation profiling was performed using Agilent 244K Human CpG Island arrays on 167 pediatric AML samples of which 12 had t(8;16)(p11;p13). Differential methylation of the TRIM59 promoter, an oncogene described in prostate cancer located on chromosome 3, was confirmed by pyrosequencing, with subsequent mRNA overexpression, validated by RT-qPCR. Conclusion: This study shows the distinct nature of t(8;16)(p11;p13) pediatric AML with remarkable clinical features such as high percentage of leukemia cutis and DIC. The 5-year OS of this pediatric cohort was intermediate (59%), and therefore different from the dismal prognosis reported in adult cases. In neonatal t(8;16)(p11;p13) AML, spontaneous remissions were reported. Profiling studies have shed light on potential oncogenic mechanisms involved in AML with t(8;16)(p11;p13), including HOX-gene upregulation similar to MLL-rearranged AML. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 1002-1002
Author(s):  
Yusuke Hara ◽  
Norio Shiba ◽  
Kentaro Ohki ◽  
Myoung-ja Park ◽  
Akira Shimada ◽  
...  

Abstract Background: Although pediatric acute myeloid leukemia (AML) still has a poor prognosis, advanced risk-stratified chemotherapy and effective supportive care have improved the outcome, with the long-term survival rate of 60–70%. Comprehensive molecular analysis using next-generation sequencing identified the heterogeneity of this disease, suggesting that it would be difficult to increase the effectiveness of risk stratification. FLT3-ITD, accounting for 12–15% of pediatric AML, is a well-known predictor of poor prognosis and is used for risk stratification. However, recent studies have reported frequent co-occurrence of FLT3-ITD with other prognostic factors such as NUP98-NSD1, MLL-PTD, NPM1, and DEK-NUP214. These findings prompted us to reconsider the significance of FLT3-ITD and search for other factors that can identify a poor-prognosis subgroup in pediatric AML patients with FLT3-ITD. Patients and Methods: From 2006 to 2010, 485 de novo pediatric AML patients aged <18 years participated in the Japanese AML-05 study conducted by the Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG). A total of 369 samples were available for this study. All samples were centrally reviewed at diagnosis according to the FAB classification and cytogenetic analysis. FLT3-ITD was found in 47 patients (12.7%). Age and WBC counts were significantly higher in FLT3-ITD-positive than in FLT3-ITD-negative patients (p = 0.0016 and p = 0.0003, respectively). In contrast, the probability of overall survival (pOS) in FLT3-ITD-positive patients was significantly lower than that in FLT3-ITD-negative patients (4-year pOS: 41.3% vs 69.5%, p < 0.0001). Fusion gene and gene mutation analyses were performed for the detection of NUP98-NSD1, NUP98-JARID1A, CBFA2T3-GLIS2, FUS-ERG, DEK-NUP214, RUNX1-RUNX1T1, CBFB-MYH11, KIT, N-RAS, K-RAS, WT1, NPM1, and MLL-PTD. Results and Discussion: Survival analysis in FLT3-ITD-positive patients (n = 47) revealed that patients with FAB subtypes M4 and M5 (M4/5 patients; n = 9 and 11, respectively) had a much poorer prognosis than those with other FAB subtypes (non-M4/5 patients; n = 27; 4-year pOS: 7.5% vs 70.3%, p < 0.0001). Six of 9 FAB-M4 patients and all FAB-M5 patients died, indicating that the M4/5 patients are strongly associated with a poor survival in FLT3-ITD-positive patients. In M4/5 patients (n = 20), age was not statistically different (11.3 years vs 10.9 years, p = 0.5902), but the WBC count was significantly higher than non-M4/5 patients (110,200/ µL vs 26,200/ µL, p = 0.0176). In M4/5 patients, NUP98-NSD1 (n = 6), MLL-PTD (n = 3), DEK-NUP214 (n = 2), KIT mutation (n = 2), and WT1 mutation (n = 3) were repeatedly detected, and all patients with any of these aberrations died. Prognostic independence of NUP98-NSD1 and MLL-PTD remains to be determined because they are associated with a high incidence of FLT3-ITD. Meanwhile, 6 patients did not have any molecular aberrations, and 4 patients had a normal karyotype, suggesting the presence of cryptic fusion genes or unknown gene mutations. On the other hand, favorable genetic aberrations were frequent in non-M4/5 patients (n = 27). RUNX1-RUNX1T1 and NPM1 mutations were found in 3 and 5 non-M4/5 patients, respectively. All of these patients are still alive without a relapse, suggesting that the effects of FLT3-ITD are reversed by some favorable genetic aberrations, although the underlying mechanism remains unknown. Non-M4/5 patients who died (n = 7) included 1 patient each with NUP98-NSD1 and CBFA2T3-GLIS2; thus, further criteria are necessary to not misclassify these patients. Although it is unclear why the poor prognosis associated with M4/5 was not noticed to date, the possibilities are racial difference and the small sample size of Japanese cohort studies. In addition, the problem is that there are no promising treatments at present for patients with a very poor outcome, even if such a distinct subgroup is identified. Conclusions: We identified the prognostic significance of the FAB classification which stratified Japanese pediatric AML patients with FLT3-ITD into a very high-risk group and intermediate-risk group. The poor prognosis associated with NUP98-NSD1, MLL-PTD, and DEK-NUP214 may be worsened with the co-occurrence of FLT3-ITD and FAB-M4/5. We believe that these findings will help to develop not only a risk-stratified therapy, but also an epoch-making treatment for this poor-prognosis subgroup of pediatric AML. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 2021 ◽  
pp. 1-14
Author(s):  
Feng Jiang ◽  
Xin-Yu Wang ◽  
Ming-Yan Wang ◽  
Yan Mao ◽  
Xiao-Lin Miao ◽  
...  

Objective. The aim of this research was to create a new genetic signature of immune checkpoint-associated genes as a prognostic method for pediatric acute myeloid leukemia (AML). Methods. Transcriptome profiles and clinical follow-up details were obtained in Therapeutically Applicable Research to Generate Effective Treatments (TARGET), a database of pediatric tumors. Secondary data was collected from the Gene Expression Omnibus (GEO) to test the observations. In univariate Cox regression and multivariate Cox regression studies, the expression of immune checkpoint-related genes was studied. A three-mRNA signature was developed for predicting pediatric AML patient survival. Furthermore, the GEO cohort was used to confirm the reliability. A bioinformatics method was utilized to identify the diagnostic and prognostic value. Results. A three-gene (STAT1, BATF, EML4) signature was developed to identify patients into two danger categories depending on their OS. A multivariate regression study showed that the immune checkpoint-related signature (STAT1, BATF, EML4) was an independent indicator of pediatric AML. By immune cell subtypes analyses, the signature was correlated with multiple subtypes of immune cells. Conclusion. In summary, our three-gene signature can be a useful tool to predict the OS in AML patients.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 5228-5228
Author(s):  
Genki Yamato ◽  
Hiroki Yamaguchi ◽  
Hiroshi Handa ◽  
Norio Shiba ◽  
Satoshi Wakita ◽  
...  

Abstract Background Acute myeloid leukemia (AML) is a complex disease caused by various genetic alterations. Some prognosis-associated cytogenetic aberrations or gene mutations such as FLT3-internal tandem duplication (ITD), t(8;21)(q22;q22)/RUNX1-RUNX1T1, and inv(16)(p13q22)/CBFB-MYH11 have been found and used to stratify the risk. Numerous gene mutations have been implicated in the pathogenesis of AML, including mutations of DNMT3A, IDH1/2, TET2 and EZH2 in addition to RAS, KIT, NPM1, CEBPA and FLT3in the recent development of massively parallel sequencing technologies. However, even after incorporating these molecular markers, the prognosis is unclear in a subset of AML patients. Recently, NUP98-NSD1 fusion gene was identified as a poor prognostic factor for AML. We have reported that all pediatric AML patients with NUP98-NSD1 fusion showed high expression of the PR domain containing 16 (PRDM16; also known as MEL1) gene, which is a zinc finger transcription factor located near the breakpoint at 1p36. PRDM16 is highly homologous to MDS1/EVI1, which is an alternatively spliced transcript of EVI1. Furthermore, PRDM16 is essential for hematopoietic stem cell maintenance and remarkable as a candidate gene to induce leukemogenesis. Recent reports revealed that high PRDM16 expression was a significant marker to predict poor prognosis in pediatric AML. However, the significance of PRDM16 expression is unclear in adult AML patients. Methods A total of 151 adult AML patients (136 patients with de novo AML and 15 patients with relapsed AML) were analyzed. They were referred to our institution between 2004 and 2015 and our collaborating center between 1996 and 2013. The median length of follow-up for censored patients was 30.6 months. Quantitative RT-PCR analysis was performed using the 7900HT Fast Real Time PCR System with TaqMan Gene Expression Master Mix and TaqMan Gene Expression Assay. In addition to PRDM16, ABL1 was also evaluated as a control gene. We investigated the correlations between PRDM16 gene expression and other genetic alterations, such as FLT3-ITD, NPM1, and DNMT3A, and clarified the prognostic impact of PRDM16 expression in adult AML patients. Mutation analyses were performed by direct sequence analysis, Mutation Biased PCR, and the next-generation sequencer Ion PGM. Results PRDM16 overexpression was identified in 29% (44/151) of adult AML patients. High PRDM16 expression correlated with higher white blood cell counts in peripheral blood and higher blast ratio in bone marrow at diagnosis; higher coincidence of mutation in NPM1 (P = 0.003) and DNMT3A (P = 0.009); and lower coincidence of t(8;21) (P = 0.010), low-risk group (P = 0.008), and mutation in BCOR (P = 0.049). Conversely, there were no significant differences in age at diagnosis and sex distribution. Patients with high PRDM16 expression tended to be low frequency in M2 (P = 0.081) subtype, and the remaining subtype had no significant differences between high and low PRDM16 expression. Remarkably, PRDM16 overexpression patients were frequently observed in non-complete remission (55.8% vs. 26.3%, P = 0.001). Patients with high PRDM16 expression tended to have a cumulative incidence of FLT3-ITD (37% vs. 21%, P = 0.089) and MLL-PTD (15% vs. 5%, P = 0.121). We analyzed the prognosis of 139 patients who were traceable. The overall survival (OS) and median survival time (MST) of patients with high PRDM16 expression were significantly worse than those of patients with low expression (5-year OS, 17% vs. 32%; MST, 287 days vs. 673 days; P = 0.004). This trend was also significant among patients aged <65 years (5-year OS, 25% vs. 48%; MST, 361 days vs. 1565 days, P = 0.013). Moreover, high PRDM16 expression was a significant prognostic factor for FLT3-ITD negative patients aged < 65 years in the intermediate cytogenetic risk group (5-year OS, 29% vs. 58%; MST, 215 days vs. undefined; P = 0.032). Conclusions We investigated the correlations among PRDM16 expression, clinical features, and other genetic alterations to reveal clinical and prognostic significance. High PRDM16 expression was independently associated with non-CR and adverse outcomes in adult AML patients, as well as pediatric AML patients. Our finding indicated that the same pathogenesis may exist in both adult and pediatric AML patients with respect to PRDM16 expression, and measuring PRDM16 expression was a powerful tool to predict the prognosis of adult AML patients. Disclosures Inokuchi: Bristol-Myers Squibb: Honoraria, Research Funding; Novartis: Honoraria; Celgene: Honoraria; Pfizer: Honoraria.


Leukemia ◽  
2018 ◽  
Vol 33 (2) ◽  
pp. 348-357 ◽  
Author(s):  
Nicolas Duployez ◽  
Alice Marceau-Renaut ◽  
Céline Villenet ◽  
Arnaud Petit ◽  
Alexandra Rousseau ◽  
...  

2012 ◽  
Vol 96 (2) ◽  
pp. 234-240 ◽  
Author(s):  
Liang Huang ◽  
Kuangguo Zhou ◽  
Yunfan Yang ◽  
Zhen Shang ◽  
Jue Wang ◽  
...  

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3263-3263
Author(s):  
Luca Lo Nigro ◽  
Laura Sainati ◽  
Anna Leszl ◽  
Elena Mirabile ◽  
Monica Spinelli ◽  
...  

Abstract Background: Myelomonocytic precursors from acute or chronic leukemias can differentiate to dendritic cells in vitro, but leukemias with a dendritic cell immunophenotype are rare, have been reported mainly in adults, and their molecular pathogenesis is unknown. Dendritic cells are classified as Langherans, myeloid and lymphoid/plasmacytoid cells, but leukemias arising from dendritic cells are unclassified in the FAB system. We identified a new entity of pediatric acute myeloid leukemia (AML) presenting with morphologic and immunophenotypic features of mature dendritic cells, which is characterized by MLL gene translocation. Methods and Results: Standard methods were used to characterize the morphology, immunophenotype, karyotype and MLL translocations in 3 cases of pediatric AML. The patients included two boys and one girl diagnosed with AML between 1–6 years old. Their clinical histories and findings included fever, pallor, abdominal and joint pain, adenopathy, hepatosplenomegaly, normal WBC counts but anemia and thrombocytopenia. and no evidence of CNS disease. The bone marrow aspirates were hypocellular and replaced completely by large blasts with irregular nuclei, slightly basophilic cytoplasm, and prominent cytoplasmic projections. There were no cytoplasmatic granules or phagocytosis. Myeloperoxidase and alpha napthyl esterase reactions were negative, excluding FAB M5 AML, and the morphology was not consistent with any standard FAB morphologic diagnosis. The leukemic blasts in all three cases were CD83+, CD86+, CD116+, consistent with differentiated myeloid dendritic cells, and did not express CD34, CD56 or CD117. MLL translocations were identified in all 3 cases. In the first case FISH analysis showed t(10;11)(p12;q23) and RT-PCR identified and a ‘5-MLL-AF10-3’ fusion transcript. In the second case FISH analysis showed t(9;11)(p22;q23) and RT-PCR identified and a ‘5-MLL-AF9-3’ fusion transcript. In the remaining case, the MLL gene rearrangement was identified by Southern blot analysis and RT-PCR showed an MLL-AF9 fusion transcript. The fusion transcripts in all 3 cases were in-frame. Remission induction was achieved with intensive chemotherapy, and all three patients have remained in durable remission for 30–60 months after hematopoietic stem cell transplantation. Conclusions. We have characterized a new pediatric AML entity with features of mature dendritic cells, MLL translocation and an apparently favorable prognosis. The in-frame MLL fusion transcripts suggest that chimeric MLL oncoproteins underlie its pathogenesis. The partner genes in all 3 cases were known partner genes of MLL that encode transcription factors. This study increases the spectrum of leukemias with MLL translocations. Comprehensive morphological, immunophenotypic, cytogenetic and molecular analyses are critical for this diagnosis, and will reveal its frequency and spectrum as additional cases are uncovered.


Sign in / Sign up

Export Citation Format

Share Document