scholarly journals Integration of genetic and clinical risk factors improves prognostication in relapsed childhood B-cell precursor acute lymphoblastic leukemia

Blood ◽  
2016 ◽  
Vol 128 (7) ◽  
pp. 911-922 ◽  
Author(s):  
Julie A. E. Irving ◽  
Amir Enshaei ◽  
Catriona A. Parker ◽  
Rosemary Sutton ◽  
Roland P. Kuiper ◽  
...  

Key Points Chromosomal abnormalities predict outcome after relapse in BCP-ALL, and high-risk cytogenetics takes precedence over clinical risk factors. Patients with mutations or deletions targeting TP53, NR3C1, BTG1, and NRAS were associated with clinical high risk and an inferior outcome.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 584-584
Author(s):  
Susanna Ranta ◽  
Nadine Gretenkort Andersson ◽  
Ulf R. Tedgard ◽  
Tony Frisk ◽  
Maria Winther Gunnes ◽  
...  

Abstract Introduction Cerebral sinus venous thrombosis (CSVT) is potentially life-threatening thrombosis with mortality around 10%. Venous thromboembolism (VTE) is a common complication in children with cancer. These children have several thrombotic risk factors such as the malignancy itself, severe infections, prothrombotic medication and immobilization. The treatment of acute lymphoblastic leukemia (ALL) includes steroids and asparaginase (ASP), raising the VTE risk. In children with ALL the central nervous system (CNS) is a common localization for VTE. However, retrospective studies on small numbers of patients, larger studies and population-based data in children are scarce. The five Nordic countries, Estonia and Lithuania have a common treatment protocol for children with ALL between 1 and 18 years of age with prospective registration of toxicities, including CSVT offering a unique opportunity to study CSVT in this patient group. This is to our knowledge the largest report of children with ALL and CSVT describing the incidence, symptoms, treatment and the effect of CSVT on ALL treatment. Methods We assessed the symptoms, treatment, clinical risk factors and outcome of all children between ages 1 and 17 years at diagnosis of B-cell precursor or T-cell ALL between June 2008 and July 2013 and with CSVT. Data were collected from the patients’ medical records and the NOPHO leukemia registry. Results In total, 20 (1.9%) of the 1038 children with ALL treated according to the NOPHO ALL 2008 protocol developed CSVT. The cumulative incidence of CSVT was 2.0%. All the thromboses occurred within the first 5 months of treatment. The most common symptoms at the diagnosis of CSVT were headache, convulsions, weakness/fatigue and cerebral nerve palsy/hemiparesis/hemiplegia. The most frequent localizations for CSVT were sinus sagittalis (n=16) and sinus transversus (n=10). However, in most cases multiple cerebral veins were involved ( 70%). Median D-dimer at time of the CSVT diagnosis was 0.85 mg/L (range 0.19-4.7 mg/L) with 5 patients having normal D-dimer. We could not identify any clinical risk factors for CSVTs. CSVT was associated with steroids (treatment within 2 weeks before the diagnosis of CSVT) in 16/20 and with Pegylated asparaginase in 16/20. Fifteen patients were later screened for the inherited thrombophilic factors; one child had heterozygous prothrombin G20110A mutation and another heterozygous factor V (R506Q) Leiden mutation. Most patients (19/20) were treated with anticoagulants: mostly low molecular weight heparin (LMWH). The median treatment with LMWH was 26 weeks (range 14-119 weeks). No bleeding complications were observed in connection with LMWH. Two deaths were directly related to CSVT. Asparaginase was omitted from the treatment in 7 and delayed or reduced in 5 of the cases raising the risk for subsequent suboptimal leukaemia treatment. Of the surviving 18 patients, follow-up imaging revealed complete recanalization in 7 and partial recanalization in 7 cases. No imaging was available for the remaining 4 patients. Conclusions The incidence of CSVT in children with ALL was approximately 2%. No statistically significant clinical predictors for CSVT were identified. The mortality related to CSVT was 10%. Anticoagulation with LMWH was the treatment of choice in most cased and was well tolerated. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 125 (11) ◽  
pp. 1759-1767 ◽  
Author(s):  
Shannon L. Maude ◽  
Sibasish Dolai ◽  
Cristina Delgado-Martin ◽  
Tiffaney Vincent ◽  
Alissa Robbins ◽  
...  

Key Points ETP-ALL, a high-risk subtype of T-ALL, is characterized by aberrant activation of the JAK/STAT signaling pathway. The JAK1/2 inhibitor ruxolitinib demonstrates robust activity in patient-derived xenograft models of ETP-ALL.


Blood ◽  
2016 ◽  
Vol 127 (15) ◽  
pp. 1863-1869 ◽  
Author(s):  
Nitin Jain ◽  
Audrey V. Lamb ◽  
Susan O’Brien ◽  
Farhad Ravandi ◽  
Marina Konopleva ◽  
...  

Key Points Adult patients with ETP-ALL/LBL have poor long-term outcomes. Novel therapies are urgently needed for adult patients with ETP-ALL/LBL.


2010 ◽  
Vol 30 (1) ◽  
pp. 58-73 ◽  
Author(s):  
Paul T. Seed ◽  
Lucy C. Chappell ◽  
Michael A. Black ◽  
Katrina K. Poppe ◽  
Yuan-Chun Hwang ◽  
...  

2021 ◽  
pp. 1-9
Author(s):  
Anna Płotka ◽  
Krzysztof Lewandowski

<b><i>Background:</i></b> <i>BCR/ABL1</i>-like acute lymphoblastic leukemia is a newly recognized high-risk subtype of ALL, characterized by the presence of genetic alterations activating kinase and cytokine receptor signaling. This subtype is associated with inferior outcomes, compared to other B-cell precursor ALL. <b><i>Summary:</i></b> The recognition of <i>BCR/ABL1</i>-like ALL is challenging due to the complexity of underlying genetic alterations. Rearrangements of <i>CRLF2</i> are the most frequent alteration in <i>BCR/ABL1</i>-like ALL and can be identified by flow cytometry. The identification of <i>BCR/ABL1</i>-like ALL can be achieved with stepwise algorithms or broad-based testing. The main goal of the diagnostic analysis is to detect the underlying genetic alterations, which are critical for the diagnosis and targeted therapy. <b><i>Key Messages:</i></b> The aim of the manuscript is to review the available data on <i>BCR/ABL1</i>-like ALL characteristics, diagnostic algorithms, and novel, molecularly targeted therapeutic options.


2020 ◽  
Vol 41 (21) ◽  
pp. 1988-1999 ◽  
Author(s):  
Neal A Chatterjee ◽  
Jani T Tikkanen ◽  
Gopi K Panicker ◽  
Dhiraj Narula ◽  
Daniel C Lee ◽  
...  

Abstract Aims To determine whether the combination of standard electrocardiographic (ECG) markers reflecting domains of arrhythmic risk improves sudden and/or arrhythmic death (SAD) risk stratification in patients with coronary heart disease (CHD). Methods and results The association between ECG markers and SAD was examined in a derivation cohort (PREDETERMINE; N = 5462) with adjustment for clinical risk factors, left ventricular ejection fraction (LVEF), and competing risk. Competing outcome models assessed the differential association of ECG markers with SAD and competing mortality. The predictive value of a derived ECG score was then validated (ARTEMIS; N = 1900). In the derivation cohort, the 5-year cumulative incidence of SAD was 1.5% [95% confidence interval (CI) 1.1–1.9] and 6.2% (95% CI 4.5–8.3) in those with a low- and high-risk ECG score, respectively (P for Δ &lt; 0.001). A high-risk ECG score was more strongly associated with SAD than non-SAD mortality (adjusted hazard ratios = 2.87 vs. 1.38 respectively; P for Δ = 0.003) and the proportion of deaths due to SAD was greater in the high vs. low risk groups (24.9% vs. 16.5%, P for Δ = 0.03). Similar findings were observed in the validation cohort. The addition of ECG markers to a clinical risk factor model inclusive of LVEF improved indices of discrimination and reclassification in both derivation and validation cohorts, including correct reclassification of 28% of patients in the validation cohort [net reclassification improvement 28 (7–49%), P = 0.009]. Conclusion For patients with CHD, an externally validated ECG score enriched for both absolute and proportional SAD risk and significantly improved risk stratification compared to standard clinical risk factors including LVEF. Clinical Trial Registration https://clinicaltrials.gov/ct2/show/NCT01114269. ClinicalTrials.gov ID NCT01114269.


Blood ◽  
2015 ◽  
Vol 125 (16) ◽  
pp. 2486-2496 ◽  
Author(s):  
Nathalie Dhédin ◽  
Anne Huynh ◽  
Sébastien Maury ◽  
Reza Tabrizi ◽  
Kheira Beldjord ◽  
...  

Key Points SCT in first complete remission is associated with 69.5% 3-year overall survival in high-risk ALL adult patients treated with intensified pediatric-like protocol. Poor early MRD response is a powerful tool to select patients who may benefit from SCT in first complete remission.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1182-1182 ◽  
Author(s):  
Marion K Mateos ◽  
Toby N. Trahair ◽  
Chelsea Mayoh ◽  
Pasquale M Barbaro ◽  
Rosemary Sutton ◽  
...  

Abstract Venous thromboembolism (VTE) is an unpredictable and life-threatening toxicity that occurs early in acute lymphoblastic leukemia (ALL) therapy. The incidence is approximately 5% in children diagnosed with ALL [Caruso et al. Blood. 2006;108(7):2216-22], which is higher than in other pediatric cancer types [Athale et al. Pediatric Blood & Cancer. 2008;51(6):792-7]. Clinical risk factors for VTE in children during ALL therapy include older age and the use of asparaginase. We hypothesized that there may be additional risk factors that can modify VTE risk, beyond those previously reported [Mitchell et al. Blood. 2010;115(24):4999-5004]. We sought to define early predictive clinical factors that could select a group of children at highest risk of VTE, with possible utility in an interventional trial of prophylactic anticoagulation. We conducted a retrospective study of 1021 Australian children, aged 1-18 years, treated between 1998-2013 on successive BFM-based ALL therapies. Patient records were reviewed to ascertain incidence of VTE; and to systematically document clinical variables present at diagnosis and during induction/consolidation phases of therapy. The CTCAE v4.03 system was used for grading of VTE events. Multivariate logistic and cox regression were used to determine significant clinical risk factors associated with VTE (SPSS v23.0). All P values were 2-tailed, significance level <.05. The incidence of on-treatment VTE was 5.09% [96% ≥Grade 2 (CTCAE v4.0)]. Age ≥10 years [P =.048, HR 1.96 (95% confidence interval= 1.01-3.82)], positive blood culture in induction/consolidation [ P =.009, HR 2.35 (1.24-4.46)], extreme weight at diagnosis <5th or >95th centile [ P =.028, HR 2.14 (1.09-4.20)] and elevated peak gamma-glutamyl transferase (GGT) >5 x upper limit normal in induction/consolidation [ P =.018, HR 2.24 (1.15-4.36)] were significantly associated with VTE in multivariate cox regression modeling. The cumulative incidence of VTE, if all 4 clinical risk factors in our model were present, was 33.33%, which is significantly greater than the incidence of VTE for a patient without any risk factors (1.62%, P <.001). These 4 clinical factors could be used as a basis for assigning thromboprophylaxis in children with ALL. Our model detected 80% (42/52) of all VTE events by incorporating one or more risk factors. An equal proportion of patients eventually developing VTE could be predicted by weight and age ≥10 years; or later bacteremia and elevated GGT. Bacteremia, when present as a risk factor, preceded VTE in 80% of cases (20/25 cases) at a median of 29 days before VTE (range 3-668 days). The negative predictive value (NPV), specificity and sensitivity for the 4 risk factor model were 98.38%, 98.70% and 28.57% respectively. If 3 specified risk factors were included in the algorithm, such as 2 baseline and one treatment-related variable, the incidence of VTE was ≥25%, NPV 98.38%, specificity ≥96.19% and sensitivity 80%. The high NPV and high specificity mean the model can successfully exclude children who are not at increased risk of VTE. The challenge is to balance unnecessary exposure to anticoagulation against the risk of development of VTE. We have identified novel clinical risk factors in induction/consolidation - positive blood culture, hepatic enzymatic elevation and extreme weight at diagnosis- that may highlight risk mechanisms related to VTE pathogenesis. Our predictive model can define a group at highest risk of VTE who may benefit from randomized trials of prophylactic anticoagulation in childhood ALL therapy. Acknowledgments: The authors acknowledge support from the Kids Cancer Alliance (a Translational Cancer Research Centre of Cancer Institute NSW), Cancer Institute New South Wales, Royal Australasian College of Physicians - Kids Cancer Project Research Entry Scholarship, Cancer Therapeutics CRC (CTx) PhD Clinician Research Top-Up Scholarship, The Kids Cancer Project, Australian and New Zealand Children's Haematology Oncology Group, ASSET study members, data managers and clinical research associates at each site. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document