Gene Expression Pathways That Distinguish Diagnosis and Relapse in Childhood Acute Lymphoblastic Leukemia.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 848-848
Author(s):  
Deepa Bhojwani ◽  
Elizabeth Raetz ◽  
Naomi Moskowitz ◽  
Hokyung Lee ◽  
Bret Sohn ◽  
...  

Abstract In contrast to the excellent outcomes for children with newly diagnosed acute lymphoblastic leukemia (ALL), outcomes following ALL marrow relapse have remained poor despite incremental increases in the intensity of therapy. Those children whose relapse occurs after therapy is completed (late relapse; >36 months from diagnosis) have a much better outcome than those who relapse during therapy (early relapse; ≤36 months). To discover differences in the underlying biological mechanisms of treatment failure we have extended our study of gene expression profiling to patients who experienced their first bone marrow relapse in whom matched pairs of marrow samples from both initial diagnosis and initial marrow relapse were available (30 pairs). These children were treated on contemporary cooperative group studies over the past six years. Twenty patients had relapsed early while ten had relapsed late. Affymetrix U133A microarrays were used and data was normalized, filtered and analyzed. In an unsupervised analysis (hierarchical clustering), we observed that the diagnosis and relapse samples of individual patients who relapsed early tended to cluster together (median correlation coefficient = 0.38) while the diagnosis and relapse samples from the patients who relapsed late were more divergent (median correlation coefficient = 0.03). Using Significance Analysis of Microarrays (SAM) many genes were identified that were commonly deregulated at relapse compared to initial diagnosis (459 probe sets with a false discovery rate (FDR)< 10%). Strikingly, a number of cell cycle genes were found to be up-regulated at the time of relapse consistent with the known increased proliferation rate of ALL cells at relapse, which often have defects in negative regulators of cell cycle progression such as p16. Key differences in gene expression were validated on an independent set of samples obtained at diagnosis (29 samples) and relapse (19 samples) by real time quantitative PCR. These included BIRC5, TOP2A, CCNB1 and PTTG1. We were able to identify many common differences in early relapse pairs (221 probe sets, FDR< 10%). Many of these genes are involved in DNA replication and repair (FEN1, CHAF1A, ORC6L). To date we have been unable to identify genes uniformly deregulated at late relapse compared to initial diagnosis. Thus late relapse mechanisms may be more diverse although the smaller number of late pairs may preclude the identification of common pathways. The more divergent nature of late relapse pairs suggests that late relapse may more commonly be the result of additional transforming events that take place in a reservoir of premalignant leukemic stem cells, while early relapse represents selective outgrowth of resistant cells from the fully leukemic clone present at diagnosis. Analysis of matched diagnosis and relapse pairs have identified candidate pathways that may mediate drug resistance and suggests that agents that target the cell cycle regulatory pathway may have particular efficacy in ALL cases that relapse early. Documentation of their direct role in this process by modulation of expression in preclinical model systems will identify opportunities for rationale design of new therapeutic approaches to prevent and treat relapse.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3345-3345
Author(s):  
Deepa Bhojwani ◽  
Jinhua Wang ◽  
Jun J Yang ◽  
Debra Morrison ◽  
Meenakshi Devidas ◽  
...  

Abstract The outcome for childhood acute lymphoblastic leukemia (ALL) following marrow relapse remains bleak in spite of numerous approaches to further intensify therapy. Understanding the biological basis of relapse and chemoresistance, as well as identifying and validating potential new targets, are the goals of our study. Previously we determined global gene expression patterns of matched diagnosis and relapse leukemic blasts in 32 patients (64 samples) with childhood B-precursor ALL using Affymetrix U133A arrays (Blood2006;108(2):711–7). We now have extended this analysis to 60 patients (120 samples). Thirty-six patients relapsed early (within 36 months of initial diagnosis), while 24 patients relapsed late. Within the TEL/AML1 subset (n=12 patients), time to relapse was inversely proportional to the correlation co-efficient of expression profiles of the diagnosis and relapse matched pair samples, suggesting that the later the relapse, the more distinct the relapse clone is from the diagnostic clone. A supervised pairwise analysis in all 60 patients identified 292 probesets that were differentially expressed between diagnosis and relapse (FDR < 10%). In a relative enrichment analysis, multiple genes mediating cell death were down-regulated at relapse (p=0.00003), suggesting that the leukemia cells had evolved mechanisms to enhance survival. These included p21, TNFPAI3, RIPK2, BCLAF1, STK17B. In concert, DNA replication genes were up-regulated at relapse (p=0.00002). Differences in pathways leading to early vs. late relapse were evident. Early relapse was characterized by an over-expression of cell cycle genes reflecting a proliferative state. At the time of relapse, a marked over-representation of genes involved in the progression through the M phase of the cell cycle was observed in early relapse compared to late relapse (p=1.3E-08). Late relapse was characterized by the over-expression of genes involved in nucleoside biosynthesis, particularly targets of antifolates (DHFR, MTHFD1, TYMS). A small number of gene expression patterns were common to both early and late relapse, including up-regulation at relapse of BIRC5 (survivin): an attractive target for therapeutic intervention. In conclusion, analysis of an expanded cohort of matched diagnosis/relapse pairs has validated and extended our previous findings that early relapse is associated with a proliferative gene expression signature. In addition we have now identified pathways operative in late relapse. Targeting these individual genes and pathways may lead to innovative strategies to treat or prevent relapsed ALL.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 174-174
Author(s):  
Mary Eapen ◽  
Elizabeth Raetz ◽  
Mei-Jie Zhang ◽  
Catherine Muehlenbein ◽  
Meenakshi Devidas ◽  
...  

Abstract The best treatment for children with B-precursor acute lymphoblastic leukemia (ALL) in second remission after a marrow relapse is controversial. To address this question, we compared outcomes in 192 patients enrolled on COG chemotherapy trials 9110 (n=109), 9310 (n=59) and 9411 (n=29) and 189 HLA-matched sibling transplant recipients reported to the CIBMTR. Patients received treatment between 1991 and 1997. Patients with isolated extra-medullary relapse were excluded. Median follow-up was 103 (range 34–159) and 101 (16–160) months after chemotherapy and transplantation, respectively. Groups were similar with respect to sex, leukemia sub-type, leukocyte count at diagnosis and duration of first remission. Chemotherapy recipients were younger at diagnosis (5 vs. 8 years) and less likely to have combined marrow and extra-medullary site relapse (12% vs. 30%). To adjust for time-to transplant bias, we used left-truncated Cox regression models to examine treatment outcomes. The relative efficacy of chemotherapy and transplantation depended on the duration of first clinical remission and transplant conditioning regimen as shown in the Table below. Comparing children with early relapse (&lt;36 months from diagnosis) the 5-year probabilities of leukemia-free survival (LFS) were 23%, 44% and 8% after chemotherapy alone, transplantation with a total body radiation (TBI)-containing regimen and a non-TBI regimen, respectively. In contrast to these findings, relapse and treatment failure were similar after transplantation with a TBI-containing regimen or chemotherapy after a late relapse (≥36 months from diagnosis). The 5-year probabilities of LFS were 61% and 60% after chemotherapy alone and a TBI-containing regimen, respectively and significantly lower at 30% after a non-TBI regimen. These data support HLA-matched related donor transplantation using a TBI-containing regimen in second remission for patients with ALL and early relapse. Early relapse (&lt;36 months) Late relapse ≥36 months) ( Outcome N Relative Risk (95% CI) P-value N Relative Risk (95% CI) P-value Leukemia recurrence Chemotherapy 115 1.00 81 1.00 TBI-containing regimen 92 0.50 (0.27–0.68) 0.0004 61 1.02 (0.62–1.68) 0.91 Non-TBI containing regimen 19 1.36 (0.81–2.31) 0.24 14 2.51 (1.23–5.16) 0.01 Treatment failure Chemotherapy 115 1.00 81 1.00 TBI-containing regimen 92 0.53 (0.37–0.75) 0.0003 61 0.93 (0.63–1.37) 0.70 Non-TBI containing regimen 19 1.49 (0.93–2.37) 0.09 14 2.87 (1.60–5.13) 0.0004


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1733-1733
Author(s):  
Laura Hogan ◽  
Deepa Bhojwani ◽  
Jinhua Wang ◽  
Debra Morrison ◽  
Jun J Yang ◽  
...  

Abstract Abstract 1733 Poster Board I-759 Introduction Outcomes for relapsed pediatric acute lymphoblastic leukemia (ALL) are poor overall but depend on the timing of relapse, with lower survival rates for patients that relapse early (defined as < 36 months from diagnosis). Previously we sought to discover underlying biological pathways that mediate relapse by analyzing gene expression profiles in a large cohort of diagnosis/relapse paired bone marrow samples on the Affymetrix U133A arrays (Bhojwani et al, ASH 2008). We determined that early relapse was characterized by over-expression of cell cycle genes reflective of a proliferative state but late relapse was characterized by genes involved in nucleotide biosynthesis including targets of anti-folate agents. Given the potential therapeutic implications of these results we sought to validate these findings in an independent set of samples. Patients and Methods Validation of the first 60 pairs was performed using 26 additional pairs analyzed on Affymetrix U133Plus2 arrays. This validation set consisted of 17 early and 9 late diagnosis/relapse pairs. All patients had B precursor ALL and were at first relapse. Probesets differentially expressed between early and late relapse were identified in a supervised pair-wise analysis using an FDR cutoff of <10%. Expression of several genes found to be up-regulated at relapse by array expression were validated using real-time quantitative PCR. Results Evaluation of the last 26 pairs once again revealed distinct gene signatures that could distinguish between early and late relapse and many genes that were significant in the original 60 pairs were validated. Overall, thirty percent of the genes that distinguish diagnosis from relapse were validated in this smaller cohort. Importantly, genes involved in nucleotide metabolism that are targets of anti-folates such as thymidylate synthetase (TYMS), dihydrofolate reductase (DHFR) and methylenetetrahydrofolate dehydrogenase (MTHFD1) were validated as up-regulated at late relapse. TYMS was elevated at late relapse in the original (p=0.004) and validation (p=0.02) cohorts. MTHFD1 and DHFR also showed increased expression at late relapse (p=0.01 and p=0.09 for MTHFD1; and p=0.01and p=0.07 for DHFR) in both the discovery and validation cohorts respectively. Increased expression of DHFR and TYMS in late relapse was also confirmed by real-time quantitative PCR comparing 12 early and 12 late relapse pairs. Median expression at late relapse was 2 fold higher than diagnosis for both DHFR (p= 0.03) and TYMS (p= 0.01). Conclusion Increased expression of anti-folate target genes is seen in clinical samples from patients whose disease recurs late, indicating possible selection during maintenance where the bulk of anti-folate exposure occurs. Therapeutic strategies to augment folate antagonism may prevent late recurrences of ALL. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Author(s):  
Tatsiana Aneichyk ◽  
Stefan Schmidt ◽  
Daniel Bindreither ◽  
Armin Kroesbacher ◽  
Nikola S Mueller ◽  
...  

Glucocorticoids (GCs) are an essential component of acute lymphoblastic leukemia (ALL) therapy. To identify genes mediating the anti-leukemic GC effects in vivo, we performed gene expression profiling of lymphoblasts from 46 children during the first 6-24h of systemic GC mono-therapy. Differential gene expression analysis across all patients revealed a considerable number of GC-regulated genes (190 induced, 179 repressed at 24h). However, when 4 leukemia subtypes (T-ALL, ETV6-RUNX1+, hyperdiploid, other preB-ALLs) were analyzed individually only 17 genes were regulated in all of them showing subtype-specificity of the transcriptional response. Cell cycle-related genes were down-regulated in the majority of patients, while no common changes in apoptosis genes could be identified. Surprisingly, none of the cell cycle or apoptosis genes correlated well with the reduction of peripheral blasts used as parameter for treatment response. These data suggest that (a) GC effects on cell cycle are independent of the cell death response and (b) GC-induced cell death cannot be explained by a single transcriptional pathway conserved in all subtypes. To unravel more complex, potentially novel pathways, we employed machine learning algorithms using an iterative elastic net approach, which identified gene expression signatures that correlated with the clinical response.


Blood ◽  
2020 ◽  
Vol 135 (1) ◽  
pp. 41-55 ◽  
Author(s):  
Benshang Li ◽  
Samuel W. Brady ◽  
Xiaotu Ma ◽  
Shuhong Shen ◽  
Yingchi Zhang ◽  
...  

Li and colleagues report the genomic landscape of over 100 patients with relapsed acute lymphoblastic leukemia. Analysis of diagnosis-relapse-remission trios suggest that whereas early relapse is mediated by retained subclones, late relapse is driven by mutations induced by and conferring resistance to chemotherapy.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 336-336
Author(s):  
Olga Sala-Torra ◽  
Holly M. Gundacker ◽  
Derek L. Stirewalt ◽  
Paula A. Ladne ◽  
Era L. Pogosova-Agadjanyan ◽  
...  

Abstract Outcome of adult acute lymphoblastic leukemia (ALL) continues to be poor, and parameters to better discriminate patients with distinct prognosis are necessary. In studies comparing global gene expression differences between normal hematopoietic cells (whole bone marrow, peripheral blood, CD34+, and CD22+ sorted populations) and ALL cells using microarrays we found that the B-ALL cells showed evidence of increased expression of Connective Tissue Growth Factor (CTGF). The median log 2 transformed signal intensity of CTGF was 4.26 (range 3.95–4.76) in normal hematopoietic cells, and 6.59 (range: 3.85–10.62) in all leukemic samples; this difference in signal intensity is equivalent to a 5-fold increase in median expression of CTGF in leukemic cells. Therefore, we hypothesized that expression level of CTGF may have prognostic significance in adult ALL. Using real-time RT-PCR assays for CTGF we examined the expression of CTGF in 79 diagnostic ALL patients from SWOG protocol S9400 (28 bone marrow and 51 peripheral blood samples). Patients with L3 ALL were excluded from the study. The median age of patients was 35 (range 17–64), with the median WBC 23,400/ul (range 600–396,600), and peripheral blood blasts 56% (range: 0–98). Fifty patients had B-ALL (63%), 13 (16%) had T-ALL and lineage was unknown for 16 (20%). When treated as a continuous variable in a logistic regression model, the level of CTGF expression was significantly associated with inferior OS and DFS (p=0.007 and p=0.0012, respectively). When controlled for WBC and cell lineage, the association of CTGF with OS and DFS remained statistically significant. We then sub-grouped the ALL patients into three equal groups (tertiales) based on CTGF expression. This subgroup analysis found that the OS for patients in the highest tertile (highest CTGF expression) was approximately 11% (95% CI 0–24) at 5 years, as compared to 42% (95% CI 23–61) and 58% (95% CI 38–78) for patients with middle and low CTGF expression respectively (figure). In sub-analysis of patients with B-lineage ALL (n=50), the association of CTGF expression with OS and DFS was still statistically significant (p=0.009 and p=0.005) when treated as a continuous variable. This report is an example where a gene expression study detected a gene differentially expressed in leukemia, with clear clinical value. Moreover, this is the first report that correlates the level of expression of CTGF with outcome in ALL patients. We are actively pursuing the biological and clinical significance of CTGF in other ALL patients and model systems. Figure Figure


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1850-1850
Author(s):  
Naveed I. Khan ◽  
Kenneth Francis Bradstock ◽  
Linda J. Bendall

Abstract Wnt proteins are important bone marrow-derived growth factors known to support normal hematopoietic progenitor and stem cell development. Here we report that B cell progenitor acute lymphoblastic leukemia (pre-B ALL) cells express Wnt proteins, including Wnt-2b in 33%, Wnt-5a in 42%, Wnt-10b in 58% and Wnt-16b in 25% of cases. The Wnt receptors, Frizzled (Fz)-7 and -8 were also expressed in most cases while Fz-3, -4 and -9 were occasionally detected. Stimulation of pre-B ALL cells with Wnt-3a activated canonical Wnt signaling with increased expression and nuclear translocation of β-catenin. This resulted in a 1.7 to 5.3-fold increase in cell proliferation, which was associated with enhanced cell cycle entry. Wnt-3a also significantly increased the survival of pre-B ALL cells under conditions of serum deprivation. To determine the mechanisms involved we examined the effects of Wnt-3a on gene expression using the leukemic pre-B ALL cell line NALM6 and a cancer specific microarray (GEArray® OHS-802), which contains 440 known cancer genes. Expression of 83 genes (19%) could be detected on the array. Exposure to Wnt-3a for 24 hours resulted in increased (>1.5 fold) expression of 29 genes and reduced (<50% of control) expression of 3 genes. The most highly regulated genes in response to Wnt-3a were MYBL2, E2F1, CD10, VDAC1, CDC25B (upregulated) and TRAIL-R2 (downregulated). Using qRT-PCR, we confirmed regulation of these genes in NALM6 cells and/or in another leukemic cell line LK63. These genes play important roles in the control of cell cycle (MYBL2, E2F1 and CDC25B), apoptosis (VDAC1 and TRAIL-R2) and motility (CD10) in cancer cells. Our results suggest that Wnt signalling regulates cell growth and proliferation in leukemic cells by modulating the expression of a number of genes. To our knowledge this is the first study examining the gene expression profile following Wnt stimulation in leukemic cells and potentially identifies new therapeutic targets for treatment.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1522-1522
Author(s):  
Barbara Meissner ◽  
Cornelia Eckert ◽  
Anja Moericke ◽  
Arend von Stackelberg ◽  
Renate Kirschner-Schwabe ◽  
...  

Abstract Despite considerable improvements regarding treatment outcome about 20% of children with acute lymphoblastic leukemia (ALL) still suffer from recurrent disease. Especially patients with an early relapse occurring before 6 months of treatment cessation have a poor prognosis and, therefore, urgently need therapy optimization. As an initial step, detection of genetic aberrations that are associated with resistant disease and/or early relapse might be of great value as they could serve as prognostic markers to predict disease recurrence and, in addition, may increase our understanding of mechanisms underlying treatment resistance and early relapse. This knowledge may also help to identify new therapeutic strategies including targeted approaches. Surprisingly, almost no information is available on the genetic evolution from initial diagnosis to early relapse. As a first aim in the process of uncovering the pathomechanism of early relapse we wanted to find out if early relapse of childhood ALL reflects primarily resistant disease with little or no additional aberrations at relapse or represents evolution and selection of a new and more resistant clone possibly triggered through chemotherapy. Recently, high throughput technologies (single nucleotide polymorphism [SNP]–Arrays) have been developed to screen the whole genome with high resolution for submicroscopic, genetic alterations by simultaneously analyzing the SNP genotype and determining the copy number state. This prompted us to perform SNP-array analysis encompassing more than 100.000 SNPs in 20 childhood ALL samples collected at initial diagnosis and to compare them to their counterparts obtained at relapse. The analyzed patients belonged to the intermediate or high risk treatment group; 16 exhibited B-precursor and 4 T-precursor ALL. Exclusion criteria were translocations t(12;21), t(9;22) and (4;11) as well as pre-existing conditions (e.g. Down syndrome). At initial diagnosis, the leukemic sample of all patients already displayed a complex karyotype with multiple genetic lesions (average numbers per patient: 5.15 deletions, 1.35 amplifications, and 0.65 copy number neutral LOH (CNN-LOH)). Most alterations remained stable from initial diagnosis to relapse (99 of 103 (96%) initially observed deletions as well as 24 of 27 (89%) amplifications). Four deletions occurring in 3 different patients at initial diagnosis disappeared at relapse. Nevertheless, these patients had additional stable copy number alterations. Newly occurring aberrations at relapse were less frequent (average numbers per patient: 1.8 new deletions, 0.75 new amplifications, and 0.15 new CNN-LOH). In 3 patients genetic alterations at diagnosis and relapse were entirely identical. The most commonly affected chromosome was 9p (75% of samples). Fourteen patients had either heterozygous or homozygous deletions of CDKN2A at initial diagnosis and relapse with 2 patients progressing from heterozygous to homozygous deletions. PAX5 deletions (mainly heterozygous) were detectable in 9 patients at initial diagnosis and 10 patients at relapse. Other sides of recurrent deletions – mainly detected at relapse – included multiple areas of chromosome 7p (p21.3, p15.3, p14.3, p14.3-14.1, p14.1-p13) as well as chromosome 1q (deletion: q42.12-q44, amplification: q25.2-q31.1), chromosome 17p (deletion: p13.3-p11.2) and 17q (amplification: q21.2-q25.1). Potential pathogenetic implications of specific recurrent genetic lesions will be discussed. In conclusion, our results demonstrate a high degree of genomic stability from initial diagnosis to early relapse of childhood ALL. These results suggest that – at least in some patients – early relapse might already be predetermined by a resistant leukemic clone at the time of initial diagnosis.


Sign in / Sign up

Export Citation Format

Share Document