GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 826-826
    Abstract: Using gene expression profiling, we and others identified a novel subgroup of B-precursor acute lymphoblastic leukemia (B-ALL) with a gene expression signature similar to Philadelphia (Ph) chromosome (BCR-ABL1)-positive ALL. Termed “Ph-like” or “BCR-ABL1-like” ALL, this subgroup constitutes 10-15% of pediatric and 25% of adolescent/young adult ALL cases and is associated with a very poor clinical outcome. Using next generation sequencing, we have shown that Ph-like ALL is characterized by a highly heterogeneous spectrum of activating mutations or gene fusions targeting genes regulating cytokine receptor and tyrosine kinase signaling (JAK1/2, ABL1/2, PDGFRB, EPOR, CSF1R, AKT2, STAT5B, CRLF2, IL7R, SH2B3). As Ph-like ALLs may be sensitive to tyrosine kinase inhibitors (TKIs) in vivo, incorporating TKIs into therapy may significantly improve clinical outcomes. Here we report the development and validation of a robust gene expression classifier that can prospectively identify Ph-like ALL patients for therapeutic intervention. Methods Supervised learning methods were applied to gene expression profiles (Affymetrix U133_Plus_2.0; RMA normalized) generated from pre-treatment leukemic samples from 811 B-ALL patients accrued to COG High-Risk ALL Trials P9906 and AALL0232. Patients were partitioned into a training (P9906: n=207; AALL0232: n=278) and an independent test set (AALL0232: n=325). Next generation sequencing was used to identify Ph-like ALL-associated genomic lesions in these cohorts. The 54,675 Affymetrix probe sets were evaluated using Prediction Analysis of Microarrays (PAM), applying the method of nearest shrunken centroids to identify those probe sets best distinguishing Ph-like ALL. These probe sets were then distilled by 100 iterations of 10-fold cross-validation using three optimization criteria (overall error, average error, and ROC accuracy), leading to the identification of the 64 most predictive probe sets (derived from 38 unique genes). Quantitative RT-PCR assays were developed for each of the 38 genes by selecting optimized primer/probe sets and assays were run on 384-well Low Density Microarray (LDA) cards; 780/811 cases had residual material for LDA testing. LDA data were remodeled in the training set using double loop cross validation, resulting in a best and final predictive model and statistical algorithm containing 15 of the 38 genes (IGJ, SPATS2L, MUC4, CRLF2, CA6, NRXN3, BMPR1B, GPR110, CHN2, SEMA6A, PON2, SLC2A5, S100Z, TP53INP1, IFITM1). The sensitivity and specificity of the predictor was then evaluated in the independent test set. Results The 15 gene LDA classifier was able to predict Ph or Ph-like ALL in the test set with a high degree of sensitivity (93.0%) and specificity (89.7%) and identified the heterogeneous genomic lesions associated with Ph-like ALL with very high frequency (Table 1). When compared to non-Ph-like ALL, Ph-like cases had a significantly poorer event-free survival (HR 3.58; p 〈 .0001) (Fig. 1, left). A second predictive classifier modeled on the same training/test sets but with true BCR-ABL1 cases excluded yielded a virtually identical performance (97.2% sensitivity, 87.1% sensitivity; HR: 2.9; p 〈 .0001). Strikingly, Ph-like ALL cases with IKZF1 deletions had a significantly worse outcome when compared to ALL cases with IKZF1 deletions alone, emphasizing the clinical importance of the Ph-like signature (Fig. 1, right). Concordance between the LDA predictor and our previously reported PAM method (NEJM 360:470, 2009) was 87.2%, with the largest difference being additional CRLF2 lesions identified by LDA. Conclusions We have developed and validated a highly robust gene expression classifier for the prospective identification of Ph-like ALL. Rapidly screened patients will then undergo targeted sequencing to confirm the presence of specific genomic lesions. This approach will facilitate the therapeutic targeting of Ph-like ALL patients to novel clinical trials, hopefully leading to improved outcomes. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 115, No. 26 ( 2010-07-01), p. 5312-5321
    Abstract: Gene expression profiling of 207 uniformly treated children with high-risk B-progenitor acute lymphoblastic leukemia revealed 29 of 207 cases (14%) with markedly elevated expression of CRLF2 (cytokine receptor-like factor 2). Each of the 29 cases harbored a genomic rearrangement of CRLF2: 18 of 29 (62%) had a translocation of the immunoglobulin heavy chain gene IGH@ on 14q32 to CRLF2 in the pseudoautosomal region 1 of Xp22.3/Yp11.3, whereas 10 (34%) cases had a 320-kb interstitial deletion centromeric of CRLF2, resulting in a P2RY8-CRLF2 fusion. One case had both IGH@-CRLF2 and P2RY8-CRLF2, and another had a novel CRLF2 rearrangement. Only 2 of 29 cases were Down syndrome. CRLF2 rearrangements were significantly associated with activating mutations of JAK1 or JAK2, deletion or mutation of IKZF1, and Hispanic/Latino ethnicity (Fisher exact test, P 〈 .001 for each). Within this cohort, patients with CRLF2 rearrangements had extremely poor treatment outcomes compared with those without CRLF2 rearrangements (35.3% vs 71.3% relapse-free survival at 4 years; P 〈 .001). Together, these observations suggest that activation of CRLF2 expression, mutation of JAK kinases, and alterations of IKZF1 cooperate to promote B-cell leukemogenesis and identify these pathways as important therapeutic targets in this disease. This trial was registered at www.clinicaltrials.gov as #NCT00005603.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 1449-1449
    Abstract: The use of risk adapted therapy has improved outcome for children with ALL and assessment of early response (ER) has been used to identify a subset of patients who benefit from augmented treatment. In spite of these advances, the majority of treatment failures occur in a “good risk” subset indicating the critical need for better predictors of outcome. We performed oligonucleotide microarrays on 99 B-precursor patients treated on CCG 1961 for high risk ALL to identify: 1) genetic pathways that play a role in early response to therapy (42 day 7 rapid (R)ER, 40 slow (S)ER) and 2) a signature that would predict event free survival (28 pts & gt; 4 yr CCR, 31 relapses & lt; 3 yrs). RNA was extracted and amplified before hybridization to Affymetrix U133 Plus 2.0 microarrays representing over 47,000 transcripts. Data were normalized, filtered and then analyzed by both unsupervised and supervised methods. We identified a robust signature that predicted SER at day 7 (79% of cases in a test set) but not RER thus limiting its utility in risk stratification. Furthermore, we also identified a highly significant set of genes that correlated with SER (M2/M3) at day 14. Importantly, expression of 47 probe sets were significantly different in patients who were in CCR for more than 4 years compared to cases that experienced a relapse within the first 3 years (FDR ≤0.05%). Logistic regression indicated that each of the 47 probe sets has significant prognostic value beyond that contained in the clinical covariates (including gender, age and WBC count). A major subset of these genes were involved in signal transduction (MAP3K3, YWHAZ, PCTK2, TGFBR1, PTPRE, IFNGR2, HSPA8). In a separate validation analysis, subsets of genes were selected using logistic regression with backward, forward and stepwise variable selection and the models proved to be significantly predictive of response in the independent expression data set of 220 patients generated previously using a smaller probe set with U95Av2 microarrays (Mosquera-Caro et al, Blood 2003, 102(11)). Interestingly, the genetic models superseded clinical co-variates such as age, WBC and gender. Finally, to assess further the predictive accuracy of these gene signatures, we randomly divided the data set into a training set (2/3 cases) that was used to develop a signature predictive of relapse that was tested for accuracy in the remaining cases (1/3 cases). This was repeated three times. On average, 77% of patients in CCR and 73% of patients who relapsed were accurately classified in the test set. With further validation these signatures of outcome (not early response) will allow for more precise identification of patients who are at very high risk for relapse and may provide a mechanistic understanding of why patients fail therapy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Society of Hematology ; 2005
    In:  Blood Vol. 106, No. 11 ( 2005-11-16), p. 1457-1457
    In: Blood, American Society of Hematology, Vol. 106, No. 11 ( 2005-11-16), p. 1457-1457
    Abstract: Background: A novel sequence, NM_hypothetical protein FLJ20154, has been identified in our previous microarray study as one of the most powerful predictors of CCR in B precursor acute lymphoblastic leukemia (ALL). We have cloned, characterized, and named this gene as Outcome Predictor in Acute Leukemia 1(OPAL1). Our preliminary data indicates that OPAL1 distinguishes those ALL patients with t(12;21) who will achieve CCR, and predicts for CCR in children who have adverse prognostic features (normal karyotype, higher age, higher white blood cell count at presentation). To further investigate the role of OPAL1 in ALL, we first determined OPAL1 expression in cells from different hematopoietic lineages, compared the OPAL1 mRNA expression levels of normal hematopoietic cells with ALL patients with t(12;21) or t(1;19). Methods: Peripheral blood mononuclear cells from 46 healthy individuals (age range: 2–18 years) were collected and sorted into three groups: B enriched (CD19+), T enriched (CD3+) and others (mostly were nutrophil cells); along with pre-treatment ALL samples with high blast count ( & gt;90%) from patient with t(12;21) or t(1;19) were used for study. mRNA expression levels of OPAL1 were determined by real-time quantitative PCR. The OPAL1 expression values were calculated by using the formula: Value=1/2^ΔCt, ΔCt=Cttarget−Ctcontrol.. The generated values were grouped into four multitude ranges categories: −10−2, 10−1, 100, 101. Results: Most of the relative expression ratio of OPAL1 (normalized to GAPDH) for the B enriched (29/45, 64.5%) or T enriched cells (6/12, 50%) in healthy individuals fell in the 100 level, while 75% (9/12) of the other residual mononuclear cells (mostly neutrophils cells), their OPAL1 relative expression ratio were in the 10−1 level which is ten times lower than those seen in normal T and B cells. OPAL1 expression ratio were significantly decreased in ALL patients with t(12;21) and t(1;19) compared with normal B enriched cells, p=0.0079 and p= 0.0006, respectively. Forty five percent (9/20) of t(12;21) patients their OPAL1expression ratio were at 10−1 level, and 40% (8/20) at 10−2 level. As for patients with t(1;19) or higher risk patients group (three patients in this group were bcr/abl positive), 60% (9/15) of the OPAL1 expression ratio were at 10−2 level. These results strongly suggested that the correlation of higher OPAL1 expression is associated with “good risk” genetic subtypes of ALL Summary: Although currently the biologic functions of OPAL1 remained to be determined, the high expression level of OPAL1 in normal T or B lymphocytes compares with non-T, non-B cells; low expression in ALL leukemia blast cells suggested that OPAL1 may play a role in lymphocyte differentiation, especially in the development and maturation of the normal B cell. The reverse relationship between OPAL1 expression level and the patient genetic risk factors indicates that OPAL1 might be another strong predictor for treatment outcome along with other well known clinical indicators. Hence, the levels of OPAL1 may be useful in identifying ALL patients with a potential good outcome, who may be able to benefit from less intensive treatment regimens and still achieve long term remission.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2005
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 121, No. 3 ( 2013-01-17), p. 485-488
    Abstract: One recently identified subtype of pediatric B-precursor acute lymphoblastic leukemia (ALL) has been termed BCR-ABL1–like or Ph-like because of similarity of the gene expression profile to BCR-ABL1 positive ALL suggesting the presence of lesions activating tyrosine kinases, frequent alteration of IKZF1, and poor outcome. Prior studies demonstrated that approximately half of these patients had genomic lesions leading to CRLF2 overexpression, with half of such cases harboring somatic mutations in the Janus kinases JAK1 and JAK2. To determine whether mutations in other tyrosine kinases might also occur in ALL, we sequenced the tyrosine kinome and downstream signaling genes in 45 high-risk pediatric ALL cases with either a Ph-like gene expression profile or other alterations suggestive of activated kinase signaling. Aside from JAK mutations and 1 FLT3 mutation, no somatic mutations were found in any other tyrosine kinases, suggesting that alternative mechanisms are responsible for activated kinase signaling in high-risk ALL.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 530-530
    Abstract: Abstract 530 Although cure rates of childhood acute lymphoblastic leukemia (ALL) have improved dramatically, a substantial portion of children still relapse and the prognosis of relapsed ALL is extremely poor. Therefore, a better understanding of molecular determinants of drug resistance in ALL is imperative for the development of more efficacious and individualized therapy, particularly in the context of relapsed disease. In a recent genome-wide association study of 2,534 children with ALL, we identified that genetic variation in PDE4B– phosphodiesterase 4B–strongly influenced the risk of ALL relapse across various ALL treatment regimens (Nat Genet 2011: 43:237). While PDE4B is the predominant phosphodiesterase in lymphoid tissue and a major regulator of cyclic AMP, its role in ALL pathobiology is largely unknown. To this end, we sought to characterize the molecular mechanisms by which PDE4B modulates antileukemic drug sensitivity in ALL. We first characterized PDE4B expression in ALL blasts at diagnosis and its relationship with drug response in vivo in 3 independent cohorts of children with ALL. In 191 children with newly diagnosed ALL enrolled on the COG P9906 protocol, PDE4B expression in ALL blasts was positively correlated with minimal residual disease status at the end of remission induction (P=0.0096). Higher PDE4B expression was also associated with slower early response to induction therapy in COG 1961 (N=82, P=0.019). In 275 children with newly-diagnosed ALL enrolled on the Shanghai Children's Medical Center ALL05 study, we determined that PDE4B2 was the predominant isoform of PDE4B in ALL blasts (P 〈 0.0001); there was also a trend that children with poor in vivo response to the upfront single-agent prednisone treatment had higher PDE4B2 expression in the diagnostic blasts (P=0.042). In parallel, shRNA-mediated knock-down of PDE4B in a glucocorticoid-sensitive (i.e., Nalm6) and a glucocorticoid-resistant (i.e., UOCB1) ALL cell line significantly potentiated cytotoxic effects of prednisolone, whereas Nalm6 and CEM ALL cells over-expressing PDE4B2 were significantly more resistant to prednisolone compared to cells transduced with empty vectors. Sensitization to glucocorticoid was further amplified by forskolin, a stimulator of cAMP synthesis, and was concomitant with activation of PKA as determined by CREB phosphorylation, suggesting that the effects of PDE4B on glucocorticoid sensitivity involve signaling of the cAMP-PKA cascade. Importantly, PDE4B knockdown by shRNA and pharmacologic inhibition by rolipram in Nalm6 and UOCB1 cells consistently led to upregulation of BIM, a key apoptosis regulator and a critical mediator of glucocorticoid sensitivity in lymphoid cells. In both cells lines, BIM upregulation following PDE4B inhibition was enhanced by forskolin but suppressed by PKA inhibitor H89, indicating that BIM might act as a downstream effector of cAMP-PKA signaling in ALL. Finally, we evaluated effects of small molecule regulators of the PDE-cAMP pathway (namely, forskolin and rolipram) on glucocorticoid response in primary ALL cells (N=23) in vitro. Measuring IC50 by MTT assay, increased cytotoxicity was observed in 16 (70%), 8 (35%), and 17 (74%) cases, when forskolin, rolipram, or both were added to prednisolone, respectively. In conclusion, tumor expression of PDE4B at diagnosis was associated with poorer early treatment response in ALL, particularly resistance to glucocorticoids in vivo. Inhibition of PDE4B sensitized both cultured and primary ALL cells to glucocorticoids via activating the cAMP-PKA pathway and subsequent upregulation of BIM. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 116, No. 23 ( 2010-12-02), p. 4874-4884
    Abstract: To resolve the genetic heterogeneity within pediatric high-risk B-precursor acute lymphoblastic leukemia (ALL), a clinically defined poor-risk group with few known recurring cytogenetic abnormalities, we performed gene expression profiling in a cohort of 207 uniformly treated children with high-risk ALL. Expression profiles were correlated with genome-wide DNA copy number abnormalities and clinical and outcome features. Unsupervised clustering of gene expression profiling data revealed 8 unique cluster groups within these high-risk ALL patients, 2 of which were associated with known chromosomal translocations (t(1;19)(TCF3-PBX1) or MLL), and 6 of which lacked any previously known cytogenetic lesion. One unique cluster was characterized by high expression of distinct outlier genes AGAP1, CCNJ, CHST2/7, CLEC12A/B, and PTPRM; ERG DNA deletions; and 4-year relapse-free survival of 94.7% ± 5.1%, compared with 63.5% ± 3.7% for the cohort (P = .01). A second cluster, characterized by high expression of BMPR1B, CRLF2, GPR110, and MUC4; frequent deletion of EBF1, IKZF1, RAG1-2, and IL3RA-CSF2RA; JAK mutations and CRLF2 rearrangements (P 〈 .0001); and Hispanic ethnicity (P 〈 .001) had a very poor 4-year relapse-free survival (21.0% ± 9.5%; P 〈 .001). These studies reveal striking clinical and genetic heterogeneity in high-risk ALL and point to novel genes that may serve as new targets for diagnosis, risk classification, and therapy.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. 1514-1514
    Abstract: Accurate risk stratification constitutes the fundamental paradigm of treatment in acute lymphoblastic leukemia (ALL), allowing the intensity of therapy to be tailored to the patient’s risk of relapse. We have recently identified 2 known (E2A-PBX1, MLL) and 6 novel cluster groups with varying prognostic significance within a high risk ALL cohort (COG P9906) using gene expression profiling. Key genes predicted membership in these novel cluster groups, including MUC4, GPR110, IGJ (poorest outcome cluster) and CENTG2 and PTPRM (most favorable outcome cluster). The poor outcome cluster had a 4-year relapse free survival (RFS) of 20.9%, while the favorable outcome group had an RFS of 94.7% -- both significantly different from the RFS of 61% for the overall cohort. In the current study, we perform quantitative RT- PCR for expression of these genes (MUC4, GPR110, IGJ, CENTG2 and PTPRM) in tandem with gene expression microarray analysis on a new cohort (n=85) of high risk ALL patients enrolled in COG 0232 to validate the findings of our prior microarray classification, and identify a manageable subset of discriminatory genes amenable to RT-PCR analysis, thus permitting the detection of these prognostically significant groups in a clinical setting. Evaluation of PBX1 expression by RT-PCR was included for further validation. RT-PCR assays were designed to simulate as closely as possible testing conditions consistent with a clinical assay; thus, RT-PCR was performed using 10 ng of RNA from each sample, run in duplicate in a quantitative TaqMan assay, and normalized to EEF2 expression. The expression of the analyzed genes by RT-PCR showed excellent correlation with the data generated by expression microarray analysis; using receiver operator curve (ROC) analysis and employing the microarray clusters as a gold standard, the analyzed genes produced areas under the curve (AUC) ranging from 0.88 to 1.0 (a finding characteristic of useful clinical tests) and highly desirable sensitivity and specificity for the identification of poor and favorable outcome groups (see table). Cluster analysis using only RT-PCR expression of these six genes reproduced the significant cluster assignments determined by the microarray analysis. RT-PCR expression of the genes showed a wide dynamic range, a valuable feature for a potential clinical assay. Moreover, using a decision tree approach, we established a step-wise algorithm using the RT-PCR data that permits accurate classification of patients into the poor, favorable and neutral outcome groups. Finally, PBX1 was significantly expressed exclusively in patients found to have E2A-PBX1 by cytogenetic or standard PCR analyses. These results confirm our prior microarray findings and demonstrate that the previously-described prognostic subgroups within high-risk pediatric ALL may be identified in a real-time, clinical setting with a robust quantitative RT-PCR assay for expression of as few as two or three genes. Identification of these prognostic subgroups may improve risk classification in ALL, enhance therapeutic targeting, and thereby improve overall outcome. Gene Prognostic group Area under ROC curve p value for ROC curve Sensitivity (%) Specificity (%) MUC4 Poor 0.88 & lt;0.0001 87.5 92.2 GPR110 Poor 0.89 & lt;0.0001 100 80.5 IGJ Poor 0.89 & lt;0.0002 87.5 87 CENTG2 Good 0.97 & lt;0.0001 100 83.3 PTPRM Good 0.97 & lt;0.0001 85.7 100 PBX1 E2A-PBX 1.00 & lt;0.0001 100 100
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 112, No. 11 ( 2008-11-16), p. sci-8-sci-8
    Abstract: With the progressive intensification of chemotherapy, the majority of children with ALL now achieve long-term survival. In parallel, a number of molecular subtypes of ALL have been identified that are associated with treatment outcomes, which are either excellent (TEL-AML1 or trisomy of chromosomes 4, 10, and 17), intermediate (MLL rearrangements or E2a-PBX), or very poor (BCR-ABL or hypodiploidy). Yet, the underlying genetic abnormalities in the majority of children with ALL, such as those with “high-risk” disease who remain resistant to current therapies, remain to be discovered. Supported by the NCI SPECS and TARGET Initiatives, the Children’s Oncology Group (COG), and The Leukemia & Lymphoma Society, we are using comprehensive genomic technologies (i.e., expression profiling, genome-wide analyses of DNA copy number abnormalities [CNAs] and germline polymorphisms, and direct gene sequencing) to develop molecular classifiers for outcome prediction that can be used to discover novel underlying genetic abnormalities and therapeutic targets in ALL. Our work has focused on a cohort of 220 children with “high-risk” ALL registered to COG Trial 9906. Using supervised learning methods on gene expression profiles, molecular classifiers predictive of relapse-free survival (RFS) and minimal residual disease (MRD) at end-induction have been developed. A 38-gene molecular risk classifier predictive of RFS (MRC-RFS) can distinguish two groups of high-risk ALL patients with different relapse risks: low (4 yr RFS: 81%, n=109) vs. high (4 yr RFS: 50%, n=98) (P 〈 0.0001). In multivariate analysis, the best predictor combines MRC-RFS and end-induction flow MRD, classifying children into low- (87% RFS), intermediate- (62% RFS), or high-risk (29% RFS) groups (P 〈 0.0001). A 21-gene molecular classifier predictive of MRD can effectively substitute for end-induction MRD, yielding a combined classifier that similarly distinguishes three risk groups at pre-treatment (low: 82% RFS; intermediate: 63% RFS; and high: 45% RFS) (P 〈 0.0001). This combined molecular classifier was further validated on an independent cohort of 84 children with high-risk ALL registered to COG Trial 1961 (P = 0.006). Using unsupervised clustering methods, 8 distinct cluster groups based on gene expression were identified, 6 of which were entirely novel. Two of the novel clusters were associated with strikingly different outcomes (95% 4-year RFS vs 20% 4-year EFS). Novel underlying genetic abnormalities and genes that may represent novel therapeutic targets have been identified in each of these clusters. Interestingly, children of Hispanic ethnicity were disproportionately represented in the poorest outcome clusters. CNAs were revealed in genes regulating B lymphoid development in 50.2% of cases (PAX5 in 30.7% and IKZF1 in 24.9%). In addition, recurring CNAs were detected in a number of other genes known to play roles in transformation, including CDKN2A/B, RB1, BTG1, IL3RA, NRAS, KRAS, NR3C2, and ERG. CNAs in IKZF1, EBF, and BTLA were strongly associated with the poorest outcome clusters defined by gene expression profiling. Deletion of IKZF1 was particularly associated with negative outcome (p=0.002). These ongoing studies demonstrate that molecular classifiers can be used to distinguish distinct prognostic groups within high-risk ALL, significantly improving risk classification schemes and the ability to prospective identify children who will respond to or fail current therapies. These classifiers are now being integrated into the design of COG clinical trials. The discovery of novel cluster groups and underlying genetic abnormalities is being exploited to develop new therapeutic targets for this disease.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 116, No. 21 ( 2010-11-19), p. 412-412
    Abstract: Abstract 412 ALL arising in infants is a highly refractory disease. Overall event-free survival (EFS) remains poor and infants with MLL rearrangements (MLL-R) or those 〈 90 days of age are known to have particularly poor outcomes. To identify genes predictive of event-free survival (EFS) that might serve as new diagnostic and therapeutic targets, we completed gene expression profiling (Affymetrix HG_U133Plus2) in 97 infant ALL cases registered to COG Clinical Trial P9407. Of these 97 infants, 78 were most recently uniformly treated on P9407 cohort 3. In the 97 cases, median age at diagnosis was 166 days (range 1–365) and increased age at diagnosis was significantly associated with improved EFS (P = 0.001). 89/97 infants had MLL-R, of which 49 had an AF4 partner gene (MLL-AF4 (AFF1)). Infants 〈 90 days of age (P=.0001) and those with MLL-R (MLL-AF4, MLL-ENL (MLLT1), MLL-Other) had a significantly decreased EFS, while infants with MLL-AF9 (MLLT3) or cases lacking MLL-R had a significantly better EFS (P=0.014). From modeling expression profiles and multivariate analyses, a number of genes were identified that had a significant effect on EFS and were independent of patient age or MLL-R status, including: TACC2 and IRX2 (from modeling the entire cohort of 97 cases); RORA, IGJ, ZEB1, YES1 (cohort 3 modeled alone); and IRX1, IRX2, ST3GAL6, HLA-DQB1, STAB1, NEGR1, IRX5 (MLL-AF4 cases modeled alone). The significant effect of MEIS1 and KCNK12 expression on EFS was lost after consideration of MLL-R status, while the significance of many genes (particularly in the HOXA family) was not independent of patient age in multivariate analyses. Assessment of the expression levels of two genes alone at diagnosis: TACC2 and IRX2 in the entire cohort of 97 cases (P 〈 0.0001; Fig. A), or, NEGRI and IRX2 in the MLL-AF4 cases (P 〈 0.0001; Fig. B), were highly predictive of outcome on current treatment regimens. Distinctive and strikingly different gene expression profiles were also seen in infant ALL cases 〈 90 days of age vs. 〉 90 days of age (in the overall cohort and in the MLL-AF4 cases). Specifically evaluating the impact of patient age treated as a continuous variable revealed a striking transition in expression profiles at 90 days with a differential expression pattern involving many genes encoding histone-related, heat shock family, or immune response regulators (including HLA-DRB4, IL1R2, HSPA1A///1B). These distinctive profiles may reflect different transformed stem/precursor cells or susceptibilities to leukemic transformation at different patient ages, altered marrow microenvironments, or altered immune status; high expression of the heat shock proteins in particular among the youngest infants may reflect a more limited immune surveillance capacity. Given the rarity of infant ALL, this study represents one of the largest uniformly treated groups of infant leukemia to undergo gene expression profiling. In these studies we have identified genes that are highly predictive of outcome at diagnosis, in all infant ALL and in MLL-AF4 cases. Further analysis of these expression profiles, coupled with validation studies in other infant ALL cohorts, may allow for the identification of novel therapeutic targets among the genes discovered herein and ultimately for the development of more effective therapies. Disclosures: Felix: None: Patent not licensed.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2010
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...