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. 132, No. Supplement 1 ( 2018-11-29), p. 1508-1508
    Abstract: Introduction Therapy-related acute myeloid leukemia (t-AML) and de novo AML (dnAML) are a group of aggressive diseases with poor outcome, in spite of recent advance in therapy. However, the alterations in different genes seems to be associated with this dismal prognosis. Recently, different groups have shown that t-AML in adult patients has a mutational profile distinct from dnAML. TP53 mutations were mostly described in t-AML. Next-generation sequencing (NGS) is a fast and accurate method used to demonstrate the presence of multiple alterations in several genes. The comparison of t-AML and dn-AML mutational profiles could be useful for improving our understanding of the molecular genetics events and in the outcome of each group as well. Objectives: 1-To describe the incidence of pathogenic variants in t-AML and dn-AML using a customized NGS panel, 2-to study correlations of these mutations with classical genetic features in each group of AMLs, 3-to compare the mutational profile of both diseases, and 4-to evaluate the prognostic impact of these mutations. Patients and Methods A total of 136 available DNA samples from non-APL dn-AML and 11 t-AML pediatric patients were retrospectively analyzed. The samples were sequenced on a Miseq (Illumina) platform, using a customized amplicon-based NGS panel (Illumina) including 65 genes. A total of 172,360 bp of DNA target regions were covered with 1,207 amplicons with a mean coverage of 1,308. Sequences obtained were analyzed by in-house bioinformatics pipeline, with particular analysis for NPM1, CEBPA and FLT3 genes, using Pindel and ITDseek algorithms. Results At least one pathogenic variant was detected in 100 out of 136 (74%) dn-AML analyzed cases, and 11 out of 11 (100%) t-AML patients. Overall, 317 mutations were found in 49 genes with a mean of 3.2 variants per patient for dn-AML, and 53 mutations in 26 genes with a mean of 4.8 variants per patient for t-AML. The most frequently mutated genes were: BCORL1 (5.4%, p=ns), FLT3 (5.0%, p=ns), KMT2A (5.0%, p=ns), BCOR (4.7%, p=0.0126), RB1 (4.4%, p=ns) for dn-AML, and KDM6A (15.1%, p 〈 0.00001), BCOR (9.4%), CUX1 (9.4%, p=0.0020), BCORL1 (5.7%), FLT3 (5.7%) for t-AML. TP53 mutations were only detected in 5 dnAML cases. In the group of patients with dnAML, the distribution of classical genetic abnormalities was: KMT2A/11q23 (24%), RUNX1-RUNX1T1 (22%), Normal karyotype (19%), CBFB-MYH11 (9%). The most frequent mutations in normal karyotype were: FLT3 (38.1%, p=0.0019), CEBPA (27.3%, p=0.0001), NPM1 (28.6%, p 〈 0.00001) and WT1 (23.8%, p=0.0001). In the group of t-AML, the genetic features were: KMT2A/11q23 (45%, p=ns) and Normal karyotype (9%, p=ns). CBFA2T2 mutation was statistically associated with the presence of RUNX1-RUNX1T1 in dn-AML (p=0.0002). No other significant association between variants and genetic groups was observed. Complete remission was achieved in 92% of cases in dn-AML and 72% in t-AML (median follow-up: 21 and 5 months respectively). Leukemia-free survival probability (LFSp) was 60(5)% [High-Risk: 55(6)%; Low-Risk: 71(10)%] for dn-AML and 13(11)% for t-AML (p=0.0008). In dn-AML group, LFSp(SE) of patients with at least one deleterious variant was 60(6)%, whereas those without any variant showed LFSp(SE): 58(10)(p=ns). In the normal karyotype group, the genotype NPM1/CEBPA-mutated plus FLT3/WT1-wild-type disclosed a LFSp(SE) of 86(13)% while remaining patients showed a LFSp(SE) of 42(14)% (p=ns). Conclusions We describe the incidence of mutations detected by NGS in a series of pediatric t-AML and dn-AML in our country. In spite of the limited number of patients, the mutation profile of t-AML seems to be different from the de novo counterpart. BCOR mutations were significantly associated with dn-AML cases, while KDM6A and CUX1 variants with t-AML patients. In contrast with adult reports, non TP53 mutations were observed in our group of t-AML patients. There was a significantly higher incidence of mutations in FLT3, CEBPA, NPM1 and WT1 within the normal karyotype group of dn-AML. The combined genotype NPM1/CEBPA-mutated plus FLT3/WT1-wild-type showed a trend to a better LFSp in dn-AML. The molecular genetic profile characterization of these heterogeneous diseases allows expansion of our understanding about the leukemogenic mechanisms involved. 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: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 132, No. Supplement 1 ( 2018-11-29), p. 4091-4091
    Abstract: Introduction: Survival of children with acute lymphoblastic leukemia (ALL) has improved in the last decades, achieving approximately 80% in Argentina. However, relapses remain the most frequent adverse event and the identification of patients with higher risk need to be refined. Deletions in IKZF1(IKZF1del) in addition with deletion of CDKN2A, CDKN2B,PAX5 or PAR1 region define a new subgroup of patients (IKZF1plus) with higher relapse rate and poor survival. Objectives: To analyze the characteristics of patients with IKZF1del and IKZF1plus, assessing the impact of the copy number alterations in several genes on survival of pediatric ALL treated with ALLIC strategy. Methods: This is a retrospective analysis performed in the population of patients admitted from October 2009 to May 2018. Samples of 432 patients with diagnosis of ALL were collected and analyzed by MLPA P-335 (MRC-Holland) for copy number alterations of IKZF1, EBF1, JAK2, CDKN2A, CDKN2B, PAX5, ETV6, BTG1, RB1 genes and PAR1 region. IKZF1plus cases were defined as those with IKZF1del with at least one additional deletion in: PAX5, CDKN2A, CDKN2B, PAR1 region. Patients were treated with 2 consecutive ALLIC protocols, according to studies stratification. Patient characteristics were compared with chi-square and Wilcoxon-sum-rank-test. Survival probability was analyzed with Kaplan-Meier calculation and survival results compared with Log-rank-test. Results: IKZF1 was not deleted in 345 cases, IKZF1del was detected in 87 cases and 47 of them were defined as IKZF1plus. Statistically significant higher WBC, MRD+ positivity on day 15, day 33 and week 12, more BCR-ABL+ and high-risk group cases, null response and higher relapse rate were observed in the IKZF1del group (total) when comparing with IKZF1 not del, and also when comparing IKZF1plus vs IKZF1 not del + IKZF1del only. EFS (SE) and DFS (SE) probabilities were: 73 (4)% and 75 (3)% for IKZF1 not del, 66 (9)% and 70 (9)% for IKZF1del, and 20 (10)% and 21 (10)% for IKZF1plus, respectively (p 〈 0.00001).DFS of the standard-risk group was not influenced by the presence of only 1 case of IKZF1del detected in this risk-group of patients. However, DFS of intermediate-risk patients was 41 (11)% for IKZF1plus while 70 (7)% and 73 (4)% were achieved for IKZF1del and IKZF1 not del respectively (p=0,0083). Therefore, high-risk patients with IKZF1plus achieved DFS of 12 (19)% vs 65 (7)% and 50 (21)% for IKZF not del and IKZF1del respectively (p=0.0085). DFS of patients with IKZF1del + CDKN2Adel was 30 (10)% and CDKN2A not deleted 67 (9)% (p=0.0433). DFS of patients with IKZF1del + CDKN2Bdel was 42 (12)% and 66 (9)% for CDKN2B not del. DFS of cases with IKZF1del in addition to deletion of ETV6, BTG1, EBF1 orJAK2 did not show statistically significant differences when comparing with IKZF1del + normal copy number of these genes. In addition, DFS of cases with RB1del was 36 (13)% while cases without RB1del showed 70 (3)% (p=0.0071). Conclusions: 1- Patients with IKZF1del and IKZF1plus disclosed biological features related to poor outcome. 2- IKZF1plus was associated with a poor outcome in intermediate and high-risk groups according to ALLIC stratification. 3- The addition of CDKN2Adel to IKZF1del influence the outcome. However, CDKN2Bdel did not show the same effect. 4- Copy number alterations of ETV6, BTG1, EBF1 or JAK2 did not demonstrate prognostic impact. 5- RB1 showed negative influence in survival. 6- Identification of patients with IKZF1plus at diagnosis could be very useful for improving risk-group stratification of pediatric ALL patients. 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: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Leukemia Research, Elsevier BV, Vol. 71 ( 2018-08), p. 6-12
    Type of Medium: Online Resource
    ISSN: 0145-2126
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2018
    detail.hit.zdb_id: 2008028-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancers, MDPI AG, Vol. 14, No. 13 ( 2022-07-05), p. 3283-
    Abstract: An association of deletions in the IKZF1 gene (IKZF1del) with poor prognosis in acute lymphoblastic leukemia (ALL) has been demonstrated. Additional deletions in other genes (IKZF1plus) define different IKZF1del subsets. We analyzed the influence of IKZF1del and/or IKZF1plus in the survival of children with ALL. From October 2009 to July 2021, 1055 bone marrow samples from patients with ALL were processed by Multiplex ligation-dependent probe amplification (MLPA). Of them, 28 patients died during induction and 4 were lost-in-follow-up, resulting in an eligible 1023 cases. All patients were treated according to ALLIC-BFM-2009-protocol. Patients were classified into three subsets: IKZF1not-deleted (IKZFF1not-del), IKZF1deleted (IKZF1del) and IKZF1del plus deletion of PAX5, CDKN2A, CDKN2B and/or alterations in CRLF2 with ERG-not-deleted (IKZF1plus). The LFSp and SE were calculated with the Kaplan–Meier calculation and compared with a log-rank test. From the 1023 eligible patients, 835 (81.6%) were defined as IKZF1not-del, 94 (9.2%) as IKZF1del and 94 (9.2%) as IKZF1plus. Of them, 100 (9.8%) corresponded to Standard-Risk (SRG), 629 (61.5%) to Intermediate-Risk (IRG) and 294 (28.7%) to High-Risk (HRG) groups. LFSp(SE) was 7 5(2)% for IKZF1not-del, 51 (6)% for IKZF1del and 48 (6)% for IKZF1plus (p-value 〈 0.00001). LFSp(SE) according to the risk groups was: in SRG, 91 (4)% for IKZF1not-del, 50 (35)% IKZF1del and 100% IKZF1plus (p-value = ns); in IRG, 77 (2)% IKZF1not-del, 61 (10)% IKZF1del and 54 (7)% IKZF1plus (p-value = 0.0005) and in HRG, 61 (4)% IKZF1not-del, 38 (8)% IKZF1del and 35 (9)% IKZF1plus (p-value = 0.0102). The IKZF1 status defines a population of patients with a poor outcome, mainly in IRG. No differences were observed between IKZF1del versus IKZF1plus. MLPA studies should be incorporated into the risk-group stratification of pediatric ALL.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2022
    detail.hit.zdb_id: 2527080-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Blood, American Society of Hematology, Vol. 132, No. 3 ( 2018-07-19), p. 264-276
    Abstract: The largest cohort of ambiguous leukemias to date reveals a better prognosis of children who started on lymphoid-directed treatment. Myeloid-type primary treatment correlated with dismal outcomes in CD19+ leukemias.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2018
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 3656-3656
    Abstract: Background: Early T precursor ALL has been defined as a poor prognosis subtype, characterized phenotypically by absence of CD1a and CD8, CD5 weak expression and presence of at least one myeloid and progenitor cell marker. Objectives: Our aims were to identify Early T-ALL among our T ALL cases, based on the characteristic phenotype and thus evaluate the prevalence, biological characteristics and outcome of this particular subset of T-ALL Methods: From April ’94 to December ’13, 197 T-ALL cases were diagnosed and 20 of them showed the typical Early T marker profile by flow cytometry (FC). Conventional cytogenetics, FISH and RT-PCR studies were performed according to standard techniques. Clonality assessment for TRG, TRB, TRD, IGH and IGK was performed by PCR (Biomed-2), heteroduplex and sequencing. Results: Sex distribution was: 16 males and 4 females; median of age: 7.9 (range: 0.3-16.6) years; median WBC: 14.9 (range: 0.6-254.0) x109/L. All but one case expressed CD34, 70% HLA-DR, 61% CD117 and 50% TdT. The most frequently expressed myeloid markers were: CD33 (85%) and CD13 (55%). Three cases expressed g/d TCR. Chromosomal abnormalities were detected in 14 of 16 evaluated cases, in 5 of them compromising 12p13 region. TCR/IgH rearrangements were detected in 63% (10/16). FLT3-ITD mutations were found in 14 % (2/14). Patients were treated with BFM-based ALL schedules and were stratified as Intermediate Risk ALL (n: 7) and High Risk ALL (n: 13) according to protocol criteria. Early response to treatment was poor in 16 cases: 6 presented poor response to prednisone (day 8), 3 MRD 〉 10%, 5 presented bone marrow M2-M3 and 2 non response. Seventeen cases (85%) achieved CR on day 33 and 2 achieved CR later. Five patients underwent HSCT in first CR. Three patients relapsed at 5, 6 and 65 months from diagnosis and one showed lineage switch to M5-AML at 12 months from CR. Three pts died in CR (2 after HSCT and 1 patient with primary immunodeficiency due to pneumonia) and one is in palliative care. Twelve patients remain in CR with a median follow-up of 54 (r: 8-144) months. Conclusions: The prevalence of Early T precursor phenotype within T ALL was 10.5% in our setting. The most frequent progenitor marker was CD34 and CD33 among myeloid markers. Of note, translocations involving 12p13 region were found in 5 patients. Sixty percent of patients remain disease free, although longer follow-up is needed in order to define prognosis of this group in our cohort of patients. The lineage switch case supports the notion of the myeloid differentiation potential of these blasts. 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: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 1858-1858
    Abstract: Background: High-Dose chemotherapy is necessary one of the most important tools for improving response rates and for decreasing relapse rate in childhood B-cell malignancies. However, deaths during induction or in Complete Remission (CR) in children with higher disease burden had adversely influenced pEFS in our setting, and for this reason an accomplished clinical support is mandatory for achieving these better results. Objective: Our aim was to diminish morbidity and mortality of patients with B-cell malignancies with the administration of a reduced intensity BFM-based induction phase, plus the addition of rituximab in a pilot study. We also retrospectively analyzed the outcome of this group of patients. Methods: This is a prospective study, single arm, non-randomized local trial for treatment of B-cell malignancies. Between December-2008 and March-2016, 81 pts consecutive ( 〈 16 years/old) (52:M/29:F) were enrolled in a protocol HPG-09, and 70 of them resulted evaluable. Patients were stratified according stage and DHL level in Risk-1, Risk-2, Risk-3 and Risk-4. Risk-4 included patients with stage IV and stage III with DHL levels higher than 1,000 U/dl. The pathology, flow-cytometry and cytogenetic findings disclosed 60 cases of Burkitt Lymphoma, 11 diffuse large B cell Lymphomas, 9 mature-B acute lymphoblastic leukemia, and 1 Precursor-B ALL with with t(8;14). The treatment locally-none BFM-counterpart included 4 doses of Rituximab administered to Risk-4 cases during early phases of treatment and intensified chemotherapy with HDMTX (24 hr) 5 g/m2, plus reduced intensity induction with pre-phase followed by A1 block (MTX 1 g/m2 4 hr infusion), in order to decrease morbidity and early mortality rates. Results: Distribution of patient according to risk-groups was: Risk-1 (n=2) 2%, Risk-2 (n=9) 11%, Risk-3: (n=13) 16% and Risk-4 (n=46) 57% of cases. Median DHL= 3,642 (range: 396-33,673) UI/dl, CNS+ cases was observed in 6 cases. Complete remission (CR) was achieved in 95.1% of the cases and 4.9% of patients died in early phases of treatment. From patient who achieve CR, 6 adverse events were observed (all of them in Risk-4 group): 2 relapses and 4 death in CR (1 Steven´s-Johnson Syndrome, 2 tumor lysis syndrome plus CID and 1 patient died after extensive initial surgery) The pEFS for R1, R2, and R3 was 100%. With a median follow-up of 48 (range:6-61) months, the pEFS (SE) for all risks groups was 91 (4)% and for Risk-4 85 (6)%. Conclusions: Risk-4 group accounted for 66% of the total population of patients, defining a more aggressive disease prevalence in our setting. Locally-adapted strategy based on BFM experience was successfully implemented, reducing induction intensity and with the incorporation of 4 doses of Rituximab. The outcome of this group of patients was very good. Reduction of induction intensity and addition of Rituximab resulted in low mortality rate and excellent survival outcome. 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: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 952-952
    Abstract: Introduction: Acute Megakaryoblastic Leukemia (AMKL) is an unusual AML subtype in childhood, being the most frequent AML in Down syndrome (DS) patients. AMKL corresponds to 19.3% of AML in our Hospital and some special characteristics have been observed. Objective: To analyze characteristics of AMKL and to compare patients with or without DS. Patients and Methods:From January-1990 to June-2014 AMKL was diagnosed in 77 cases: 57 in non-DS and 20 in DS children. Fivecases were secondary leukemias and 3 cases had been previouslytreated with chemotherapy due to wrong diagnosis. One patient was on induction at the time of analysis. These 9 cases were not evaluable for follow-up. Forty-eight non-DS and 20 DS caseswere evaluable. Megakayocyte lineage commitment was confirmed in all cases by fluorescence microscopy. MPO was analyzedboth by flow cytometry and cytochemistry. G-banding and RT-PCR were performed according to standard techniques. Four successive BFM-based protocols were administeredwith schedule modification for DS patients. Results: Mean age was:2 (range: 2 mo-13 y) years of age and mean WBC count was: 27,500 (range: 1,600-250,000)/mm3. No differences were observed in age or hematological findings at diagnosis. Extramedullar compromise was detected in 10 (17.5%) of non-SD cases. Immunophenotype defined ambiguous lineage leukemia in 3 (5.0%) of the non-DS cases, with compromise of T-lineage. Interestingly, in 4 cases the megakaryoblasts also co-expressed MPO.Cytogenetic findings showed hyperdiploidy (n=7), t(1;22)(p13;q23) (n=3), t(16;21)/FUS-ERG (n=2), abnormalities in chromosome 7 (n=5) and complex karyotype (n=9). Complete remission (CR) was achieved in 51 (33 non-DS and 18 DS) (75.0%) of 68 evaluable cases;null-responses observed in7(only non-DS, 10.3%). Early deaths occurred in 10 (14.7%) during induction phase, 2 of them before treatment onset. From the 51 patients who achieved CR, 16 (14 non-DS and 2 DS) presented relapses(p: 0.04) and 9 (6 in non-DS and 3 in DS) died in CR. EFS-probability for the total population was 34 (6)%, for non-DS 21 (6)% and for DS 63 (11)%(p-value: 0.0026). LFS-probability was 45 (7)% for the total population, for non-DS 29 (8)% and for DS 70 (11)% (p-value: 0.0117). Median follow-up: 148 (range: 2-194) months. Conclusions: 1- Extramedullar compromise and T/myeloid mixed lineage or co-expression of MPO were observed only in non-DS AMKL, in 17.5% and 12.0% respectively. 2- Null-response was observed only in non-SD patients. 3- The most frequent event of non-DS was relapse, and toxic deaths in DS. 4- The significantly better outcome of patients with DS was confirmed in our setting. 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: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Blood, American Society of Hematology, Vol. 128, No. 22 ( 2016-12-02), p. 584-584
    Abstract: Acute leukemia (AL) of ambiguous lineage (AMBI-L) comprises up to 5% of AL cases in both children and adults. Although several definitions exist, a general treatment guideline has been missing. Single country studies usually report fewer than 50 cases of children or adults. Accordingly, the international iBFM AMBI2012 Study/Registry collected 275 AMBI-L cases in patients 〈 18y from Australia, Austria, Brazil, Czechia, Germany, Greece, Israel, Italy, Netherlands, NOPHO (Denmark, Estonia, Finland, Norway, Sweden, Iceland, Latvia and Lithuania), PINDA (Chile), Poland, SAHOP (Argentina), Slovakia, St. Jude's Children Research Hospital (USA), Texas Children's Cancer Center (USA), Ukraine and United Kingdom. Each center/country reported all consecutive patients with AMBI-L from a 2 to 13 year period ending May 31, 2015. Apart from the study itself, the central database served also as a basis for consulting individual patients during the diagnostic workup. Preliminary results of this study were first introduced in ASH 2015 and now the complete detailed analysis of updated findings including significance of immunophenotype, molecular genetics, blast clearance and transplant are shown. In total, 275 patients were included in the study. Of these, 240 fulfilled the definitions of biphenotypic/mixed phenotype AL, partially overlapping with cases in whom two clones had been identified (n=68) and 15 cases presented with undifferentiated AL. Most patients started their treatment with an ALL-type protocol (n=161), 79 with AML therapy, 27 with a combined regimen, including the Interfant protocols, 2 patients were not treated, 2 received other treatment, and in 4 patients such information was missing. The 5yEFS of the entire cohort was 56±3.7% and 5y overall survival was 67±3.3%. Patients treated by ALL-type protocols had superior 5 year event free survival (5yEFS) (70±4.6%, n=158) compared to those who started AML-type treatment (5yEFS: 40±6.4%, n=78) or hybrid ALL/AML treatment (5yEFS: 50±11%, n=27). Although protocol selection was likely biased, we recommend ALL treatment, when diagnostic findings, including molecular genetics, fail to indicate AML therapy. Although myeloperoxidase (MPO) has been used as the ultimate marker of myeloid lineage, patients who started with ALL-type treatment demonstrated a better prognosis even among cases classified as MPOpos/part pos (Fig. 1). These differences by initial choice of treatment are most prominent when CD19pos/part pos cases are analyzed regardless of the overall lineage (Fig. 2). This shows that at least for CD19pos/part pos cases in the absence of RUNX1/RUNX1T1 fusion, treatment should not start with current AML-type protocols. Until week 12, patients with higher leukemia burden were slightly overrepresented compared to non-AMBI ALL patients (data not shown). In addition, patients with higher residual disease had a much poorer prognosis. Thus, Prednisone poor and good responders (based on day 8 blood blast counts) had a 5yEFS of 50±9.7%, n=38 and 81±5.8%, n=82, respectively (p=0.005). By day 15 bone marrow (BM), only cutoffs of 10-4 and 10-3 were analyzed and neither showed significant associations with EFS. At the end of induction, patients with BM residual disease ≥10-3 had a 5yEFS of 51±10%, n=49 compared to 90±4.3% for those with lower levels, n=75 (p=0.0002). Especially higher residual disease at week 12 was associated with an extremely poor EFS (Fig. 3). Early identification of patients with inadequate response and designing alternative treatment for them is our important challenge. No overall benefit of transplantation was seen in patients who started on ALL treatment or hybrid ALL/AML treatment. Again, this may be caused by a biased selection of more severe cases for transplant. In patients who started with AML treatment, transplant appeared to improve prognosis (Fig. 4). This study provides the basis for improved treatment of future patients with AMBI-L, with more accurate diagnostics. OH, AL, IJ, EM and JS were supported by Czech Health Research Council 15-28525A. Disclosures Bleckmann: JazzPharma: Other: financial support of travel costs. Moricke:JazzPharma: Honoraria, Other: financial support of travel costs. Inaba:Arog: Research Funding. Kattamis:Novartis: Honoraria, Research Funding; ApoPharma: Honoraria. Reinhardt:Boehringer Ingelheim: Membership on an entity's Board of Directors or advisory committees; Celgene: Research Funding; Jazz Pharma: Other: Travel Accomodation; Celgene: Membership on an entity's Board of Directors or advisory committees; Pfizer: Membership on an entity's Board of Directors or advisory committees.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2016
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Blood, American Society of Hematology, Vol. 126, No. 23 ( 2015-12-03), p. 252-252
    Abstract: Up to 5% of patients with acute leukemia (AL) are diagnosed as AL of ambiguous lineage. The ambiguous lineage ALs consist of mixed phenotype AL (MPAL, or biphenotypic AL, BAL), bilineal AL, switching AL and rare, undifferentiated ALs. From a molecular genetic point of view, they overlap with several molecular genetic subsets such as AL with MLL rearrangements or early T precursor AL. As no general treatment strategy exists, these patients have been variably treated with lymphoblastic (ALL)- , myeloid (AML)- or combined (hybrid) therapy, with or without stem cell transplant. They are often unreported as they are excluded from standard protocols. So far, attempts to shed more light on these patients has largely focused on definitions of ambiguous lineage AL. Only limited therapeutic observations have been possible in studies on this AL subset, usually reporting 50 or fewer pediatric/adult patients. In order to facilitate more detailed analyses, we have created an international study "iBFM AMBI2012 Study/Registry". In this study, patients under 18 years at diagnosis are eligible. Each center/country was asked to report all consecutive patients with ambiguous lineage AL, from a 2- to 13-year period ending May 31, 2015. The definitions included those with WHO and EGIL criteria and remained unchanged throughout the study. Complete information on type of treatment, follow up and immunophenotype was requested. Where available (n=101 at the time of this abstract uploading), raw cytometric FCS data files were stored centrally for review. Apart from the study itself, the central database served also as a basis for consulting individual patients during the diagnostic workup. Furthermore, data on fusion genes, cytogenetics, treatment response and availability of specimens for collateral studies were also collected. In total, 247 patients from Australia, Austria, Brazil, Czechia, Germany, Greece, Israel, Netherlands, NOPHO (Denmark, Estonia, Finland, Norway, Sweden, Iceland and Lithuania), PINDA (Chile), Poland, SAHOP (Argentina), Slovakia, St. Jude Children's Research Hospital (USA), Ukraine and United Kingdom are reported. Among those, 222 fulfilled the definitions of MPAL/BAL, partially overlapping with cases in whom two clones had been identified (n=47) and 14 cases presented with undifferentiated AL. Most of them, consistent with our general treatment guideline (Figure 1), started their treatment with an ALL type of protocol (n=150), 60 patients started on AML therapy, 8 patients received a combined regimen including the Interfant protocols, 2 patients were not treated, 13 received other treatment, and this information is missing in 9 patients (additional 5 pts. started on ALL treatment but their follow up information is incomplete). The 5 year event free survival of the entire cohort was 55±4% and its separation by first type of treatment is shown in Figure 2. In a collateral study, we set up a qPCR array on 90 genes that are characteristic of the lymphoid or myeloid lineages and/or are thought to be involved in their regulation. Using this array, sorted cells of granulocytic, monocytic, T, and B lineages at various stages of development (17 stages total) were analyzed and compared to samples of AL including 6 samples of MPAL of precursor B/myeloid phenotype. Although this array did not show a general deregulation in the MPAL genome compared to that in AL or healthy cells, subtle changes were seen such as decrease of CEBPE and LILRA2 gene expression, in comparison to classical B precursor ALL. Overall, our data shows that the general treatment guideline (Figure 1), which favors ALL treatment is justified by the outcome. However, although this study is larger than those published, caution is needed during its interpretation due to variations in diagnostics and treatment among the participating countries. Therefore, our data should be viewed as a basis for non-ambiguous treatment guidelines that will direct each patient to either ALL or AML treatment. These guidelines will be tested prospectively. In addition, the framework of this study is being used as a basis of consulting new AL cases with diagnostic uncertainties. Furthermore, it serves as a data resource for biologic studies. Supported by AZV 15-28525A, UNCE 204012, NT/14534-3, NT/13462-4, P302/12/G101. Disclosures Kattamis: Novartis: Research Funding, Speakers Bureau; ApoPharma: Speakers Bureau.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages