Pediatric Treatment Guidelines for Philadelphia Positive (Ph+) Acute Lymphoblastic Leukemia (ALL): What Are They in today’s Imatinib Era?

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4393-4393
Author(s):  
Michael Burke ◽  
Jennifer Willert ◽  
Sunil J. Desai ◽  
Richard Kadota

Abstract Background: The treatment of pediatric Philadelphia positive (Ph+) leukemias in the era of the tyrosine kinase inhibitors (TKI) continues to evolve with the role of allogeneic hematopoietic cell transplantation (allo-HCT) in these high-risk patients becoming more controversial. Ph+ acute lymphoblastic leukemia (ALL) prior to imatinib in both pediatric and adult patients has often involved intensive chemotherapy, including consolidative allo-HCT. Whether treatment strategies in 2008 have changed for these Ph+ leukemias in pediatrics, from heavily allo-HCT based to TKI based medical therapy, is presently unclear. Methods: Thirty-two pediatric centers across the United States and Canada were surveyed regarding current treatment practices for Ph+ ALL in order to explore treatment practices in 2008. The survey targeted primary pediatric oncologists and bone marrow transplant physicians regarding their treatment approach for Ph+ ALL in terms of upfront therapy, utility of allo-HCT, use of TKI (including their role in the post-HCT setting) and how response to therapy was monitored. Results: Twenty-three of the thirty-two centers completed the survey to provide a completion rate of 72% (Table 1). Twenty-two of the 27 physicians (81%) reported they do not classify patients by risk group according to age and presenting WBC (e.g. low-, intermediate- or high-risk) but rather use response to therapy to identify high risk patients, initially treating all Ph+ ALL patients the same. Eighty-one percent of survey responders recommended allo-HCT in first remission, when a matched sibling donor was available, for Ph+ ALL. Regarding the use of TKI in the post-HCT setting, 13 of 27 (48%) physicians reported using imatinib as maintenance therapy post-HCT as a means to prevent relapse. All physicians reported using PCR techniques for bcr-abl of either bone marrow, peripheral blood or both to monitor treatment response with frequencies ranging from monthly to every six months. Conclusion: Treatment of pediatric Ph+ ALL appears variable and center dependent. Classifying patients into low-, intermediate- or high-risk disease based on age and presenting WBC was not shown to be standard practice but rather using treatment response to identify high-risk patients. This survey identified a trend toward less allo-HCT in 2008 for Ph+ ALL compared to years past. Despite the trend toward less HCT, the treatment consensus in 2008 for pediatric Ph+ ALL remains MSD allo-HCT when available. Use of imatinib was recognized by all survey responders as standard of care in upfront therapy for Ph+ ALL, but the use of imatinib or other TKI in the post-HCT setting as maintenance therapy remains in question. Prospective pediatric clinical trials will be necessary to determine the optimal strategy for the Ph+ diseases. Table 1. Pediatric Centers British Columbia’s Children’s Hospital Children’s Hospital of Pittsburgh Children’s Memorial Medical Center–Northwestern Cincinnati Children’s Hospital Medical Center City of Hope Columbia Presbyterian College of Phys & Surgeons Doernbecher Children’s Hospital-OHSU Duke University Medical Center Mayo Clinic Medical College of Wisconsin Nationwide Children’s Hospital Schneider Children’s Hospital St. Jude Children’s Research Hospital Stollery Children’s Hospital–Edmonton Texas Children’s Cancer Center at Baylor College of Medicine The Children’s Hospital of Philadelphia The University of Chicago Comer Children’s Hospital University of California at San Diego/Rady Children’s Hospital San Diego UCSF School of Medicine University of Florida University of Michigan–C.S. Mott Children’s Hospital University of Minnesota Children’s Hospital, Fairview Washington University–St. Louis

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 4399-4399
Author(s):  
Michael Burke ◽  
Jennifer Willert ◽  
Sunil J. Desai ◽  
Richard Kadota

Abstract Background: The treatment of pediatric Philadelphia positive (Ph+) chronic myelogenous leukemia (CML) in the era of the tyrosine kinase inhibitors (TKI) continues to evolve with the role of allogeneic hematopoietic cell transplantation (allo-HCT) in these patients becoming more controversial. Imatinib has completely replaced allo-HCT for adult CML patients presenting in first chronic phase, reserving HCT for TKI resistant and/or advanced stage patients (accelerated phase and blast crisis). Whether treatment strategies in 2008 have changed for CML in pediatrics, from heavily allo-HCT based to TKI based medical therapy, is presently unclear. Methods: Thirty-two pediatric centers across the United States and Canada were surveyed regarding current treatment practices for CML in order to explore treatment practices in 2008. The survey targeted primary pediatric oncologists and bone marrow transplant physicians regarding their treatment approach for CML in terms of upfront therapy, utility of allo-HCT, use of TKI (including their role in the post-HCT setting) and how response to therapy was monitored. Results: Twenty-three of the thirty-two centers completed the survey to provide a completion rate of 72% (Table 1). Sixty-three percent of survey responders recommended allo-HCT, when a matched sibling donor was available, for patients with CML in first chronic phase. Regarding the use of TKI in the post-HCT setting, 9 of 27 (33%) physicians reported using imatinib as maintenance therapy post-HCT as a means to prevent relapse. All physicians reported using PCR techniques for bcr-abl of either bone marrow, peripheral blood or both to monitor treatment response with frequencies ranging from monthly to every six months. Conclusion: Treatment of pediatric CML appears variable and center dependent. This survey identified a trend toward less allo-HCT for CML in 2008 compared to years past. Despite the trend toward less HCT, the pediatric treatment consensus in 2008 for CML remains MSD allo-HCT when available. Use of imatinib was recognized by all survey responders as standard of care in upfront therapy, but the use of imatinib or other TKI in the post-HCT setting as maintenance therapy remains in question. Prospective pediatric clinical trials will be necessary to determine the optimal strategy for CML in children. Table 1. Pediatric Centers British Columbia’s Children’s Hospital Children’s Hospital of Pittsburgh Children’s Memorial Medical Center–Northwestern Cincinnati Children’s Hospital Medical Center City of Hope Columbia Presbyterian College of Phys & Surgeons Doernbecher Children’s Hospital-OHSU Duke University Medical Center Mayo Clinic Medical College of Wisconsin Nationwide Children’s Hospital Schneider Children’s Hospital St. Jude Children’s Research Hospital Stollery Children’s Hospital–Edmonton Texas Children’s Cancer Center at Baylor College of Medicine The Children’s Hospital of Philadelphia The University of Chicago Comer Children’s Hospital University of California at San Diego/Rady Children’s Hospital San Diego UCSF School of Medicine University of Florida University of Michigan–C.S. Mott Children’s Hospital University of Minnesota Children’s Hospital, Fairview Washington University–St. Louis


2021 ◽  
Author(s):  
Evert F.s. van Velsen ◽  
Robin P. Peeters ◽  
Merel T. Stegenga ◽  
F.j. van Kemenade ◽  
Tessa M. van Ginhoven ◽  
...  

Objective Recent research suggests that the addition of age improves the 2015 American Thyroid Association (ATA) Risk Stratification System for differentiated thyroid cancer (DTC). The aim of our study was to investigate the influence of age on disease outcome in ATA High Risk patients with a focus on differences between patients with papillary (PTC) and follicular thyroid cancer (FTC). Methods We retrospectively studied adult patients with High Risk DTC from a Dutch university hospital. Logistic regression and Cox proportional hazards models were used to estimate the effects of age (at diagnosis) and several age cutoffs (per five years increment between 20 and 80 years) on (i) response to therapy, (ii) developing no evidence of disease (NED), (iii) recurrence, and (iv) disease specific mortality (DSM). Results We included 236 ATA High Risk patients (32% FTC) with a median follow-up of 6 years. Age, either continuously or dichotomously, had a significant influence on having an excellent response after initial therapy, developing NED, recurrence, and DSM for PTC and FTC. For FTC, an age cutoff of 65 or 70 years showed the best statistical model performance, while this was 50 or 60 years for PTC. Conclusions In a population of patients with High Risk DTC, older age has a significant negative influence on disease outcomes. Slightly different optimal age cutoffs were identified for the different outcomes, and these cutoffs differed between PTC and FTC. Therefore, the ATA Risk Stratification System may further improve should age be incorporated as an additional risk factor.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1954-1954 ◽  
Author(s):  
Anna Maria Testi ◽  
Maria Grazia Valsecchi ◽  
Valentino Conter ◽  
Marco Vignetti ◽  
Francesca Paoloni ◽  
...  

Abstract Progress in the treatment of acute lymphoblastic leukaemia (ALL) has led to better survival rates; however, children have had a greater benefit from improved treatment modalities than adolescent who show an overall lower event-free survival (EFS) compared to younger patients. Some differences in the clinical and biologic characteristics of adolescents compared to childhood ALL may partly account for the different outcome, but adolescents treated on pediatric ALL trials seem to have a significantly better EFS than those treated on adult trials. We retrospectively compared the results obtained in a series of 245 patients ranging in age from 14 to 18 years diagnosed and enrolled in specific Italian children and adult ALL trials, between 4/1996 and 10/2003. One hundred and fifty patients, from 30 pediatric centers, underwent the childhood AIEOP ALL 95 and 2000 protocols; the other 95, from 28 adult centers, were enrolled in the GIMEMA ALL 0496 and 2000 protocols. The AIEOP 95 and 2000 trials are BFM-like protocols with a 7 drug induction followed by risk-modulated post-remission therapy that includes high-dose MTX and reinduction for low and intermediate groups, and intensive blocks (high-dose MTX and cytarabine) for high-risk patients. Standard maintenance therapy is administered up to a total of 2 years. Cranial radiotherapy is limited to high-risk patients. Stem cell transplantation is planned for very high-risk patients. The GIMEMA regimens are instead based on an induction with high-dose anthracyclines (cumulative dose 550 mg/m2), high-dose cytarabine as consolidation and do not include high-dose MTX and the reinduction phase. Standard maintenance with vincristine + daunorubicin/cyclophosphamide pulses is given for 2 years. Cranial radiotherapy is administered to all patients. The main patients characteristics at diagnosis, in the two groups under examination, were comparable except for age: median age was 15 and 16 years, respectively in the AIEOP and GIMEMA trials.Poor risk cytogenetic translocations and T-immunophenotype were equally dinstributed. Adolescents in the AIEOP protocols had a higher CR rate (94% vs 89%) and a lower relapse rate (17% vs 45%) compared to the adolescents enrolled in the GIMEMA trials. The 2-year overall survival rate was 80% in the AIEOP protocols and 71% in the GIMEMA trials. Detailed results according to the different clinical and biologic features of the adolescents analyzed will be presented. The results of our comparative study indicate that adolescents enrolled in pediatric trials have a more favourable clinical outcome.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 881-881
Author(s):  
Amélie Trinquand ◽  
Aline Tanguy-Schmidt ◽  
Raouf Ben Abdelali ◽  
Jérôme Lambert ◽  
Etienne Lengliné ◽  
...  

Abstract Abstract 881 T-cell acute lymphoblastic leukemia (T-ALL) represents a heterogeneous group of acute leukemias, which account for 25% of adult ALL. The GRAALL group recently reported a significant improvement in the outcome of BCR-ABL negative adult ALL using an intensified treatment protocol and a significantly better outcome in T-ALL harbouring NOTCH1 and/or FBXW7 (N/F) mutations compared to unmutated cases. Despite this, a third of N/F mutated T-ALL patients relapse and the identification of a T-ALL subgroup with very favorable outcome remains desirable. In a series of 212 adult T-ALLs included in the multicenter randomized GRAALL-2003 and 2005 trials, we searched for N/K-RAS (exon 1) mutations and PTEN (exon 7 mutations and gene deletion by CGH-array SNP-6 Affymetrix®) defects, which are considered as “type B3” mutations involved in pre-TCR signalling. Overall survival (OS) and event-free survival (EFS) were estimated by the Kaplan-Meier method, and then compared by the log-rank test. NOTCH1 and/or FBXW7 mutations were identified in 143 (67%) of the 212 patients and lack of N/F mutation was associated with a poor prognostic. N-RAS, K-RAS and PTEN mutations were identified in 3/191 (1.6%), 17/191 (8.9%) and 17/175 (9.7%) patients, respectively. PTEN genomic deletions/mutations and N/K-RAS activating mutations were virtually mutually exclusive. N/K-RAS mutations were more frequent in TCR negative phenotype and CNS positive T-ALLs, but did not correlate with other classical parameters, EGIL phenotype, N/F status, or cortico- or chemo-sensitivity. PTEN alterations were more frequent in mature TCR expressing, SIL-TAL+, N/F unmutated cases with high leukemic bulk tumors, but did not significantly differed with respect to age, gender, CNS involvement, cortico- or chemo-sensitivity. When analyzed separately, N/K-RAS mutations or PTEN genomic abnormalities demonstrated trends to a worse outcome. We then analyzed the effect of N/K-RAS mutations and/or PTEN genomic abnormalities on the good prognosis associated with N/F mutations by a multivariate Cox model for EFS and OS, entering the two N/F and RAS/PTEN covariates, as well as an interaction term. The prognostic significance of N/F mutations was still observed (HR, 0.26 [95% CI, 0.15–0.46] and 0.26 [95% CI, 0.14–0.49] with P<0.0001 for EFS and OS, respectively), with a significant interaction between N/F and RAS/PTEN mutations (P=0.03 and 0.05 for EFS and OS, respectively. In other terms, the favorable impact of N/F mutation was still observed in, and was restricted to patients without RAS/PTEN abnormalities. These observations led us to propose a new T-ALL oncogenetic classifier defining low-risk patients as those with N/F mutation but no RAS/PTEN mutation (97 out of 189 [51%] patients in the present cohort) and all other patients (49%, including 13% N/F and RAS/PTEN mutated cases) as high-risk patients (Figures 1A and 1B). Comparing this refined oncogenetic classifier to the simple N/F classification, hazard ratios for high-risk patients increased from 2.6 (95% CI, 1.7–4.0) to 3.25 (95% CI, 2.0–5.3) for EFS and from 2.5 (95% CI, 1.5–4.0) to 3.3 (95% CI, 1.9–5.8) for OS. When adjusting the effect of the new N/F-RAS-PTEN classifier to age (using the 35-year cutoff) and WBC (using the 100.109/L cut-off), the oncogenetic classifier remained the only significant prognostic covariate (HR= 3.2 (95% CI, 1.9–5.15) and 3.2 (95% CI, 1.9–5.6); P<0.0001 and <0.0001, for EFS and OS, respectively). The prognostic impact was maintained when GRAALL-2003 and GRAALL-2005 patients were analysed separately. Taken together, these data demonstrate that detection of RAS and PTEN mutations add significant prognostic value to assessment of N/F status, allowing identification of nearly 50% very good prognosis T-ALL adults. Figure 1A Figure 1A. Figure 1B Figure 1B. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3780-3780 ◽  
Author(s):  
Valentino Conter ◽  
Maria Grazia Valsecchi ◽  
Barbara Buldini ◽  
Rosanna Parasole ◽  
Franco Locatelli ◽  
...  

Abstract Aim: To evaluate the outcome of the Italian Association of Pediatric Hematology and Oncology (AIEOP) patients diagnosed with Early T-cell Precursor (ETP) Acute Lymphoblastic Leukemia (ALL) in the period September 2000-December 2011 and treated in the context of the AIEOP-BFM ALL 2000 Study and the subsequent - very similar – AIEOP ALL R2006 Study. Patients and methods: ETP-ALL was defined by staining negative for CD1a and CD8, mildly positive or negative for CD5 and positive for at least one of CD34, CD117, HLADR, CD13, CD33, CD11b, or CD65 antigens. Treatment consisted of BFM protocol I either with dexamethasone or prednisone in phase IA, 4 HDMTX courses (5 g/sqm over 24 hrs) in non-high risk patients or 3 poly-chemotherapy blocks in high risk patients, delayed intensification based on protocol(s) II or III, protracted intrathecal therapy or cranial radiotherapy, maintenance therapy for a total of 2 years of treatment. Hematopoietic stem cell transplantation (HSCT) was indicated for patients with poor treatment response assessed either morphologically at day +8 (Prednisone Poor Response, PPR) or day +33 (no Complete Remission, CR) or by PCR at day +78 (high-level Minimal Residual Disease, HR-MRD; ≥10-3). Results: Of the 439 T-ALL eligible patients, 34 had ETP ALL. The incidence (7.7%) may be underestimated since the full set of the data needed was not available for all patients. Out of the 34 patients with ETP ALL 14 were at high risk due to PPR; of them, 3 were HR-MRD and 5 did not achieve CR after Phase IA of induction therapy (including one with HR-MRD). Of the remaining 20 patients (all prednisone good responders), 3 patients were at high risk due to resistance to Phase IA and HR-MRD (n=1) or HR-MRD only (n=2). 17/30 patients could not be monitored by MRD due to death in Induction (n=1), or absent (n=10) or inadequate PCR markers (n=6). Of the17 patients monitored by MRD, 13 had HR-MRD at day 33 and 6 of them also at day +78. One patient died during induction therapy. The remaining 33 achieved morphological CR: 27 after phase IA and 6 (those resistant to phase IA) after phase IB of protocol I. Of 12 patients who underwent HSCT, 3 died of HSCT-related complications and 3 relapsed. With a median follow-up of 6.6 years, the 5-year event-free survival (EFS) and Survival in the 34 ETP ALL patients were of 60.9%(SE 8.5) and 66.6%(8.3), versus 71.2%(2.3) and 77.1%(2.1) respectively in the non-ETP patients. The overall cumulative incidence of relapse was 27.3%(7.8) and 22.2%(2.1) in ETP and non-ETP T-ALL patients, respectively (p=0.52). EFS in ETP vs non-ETP patients was respectively 81.9%(9.5) vs 83.8%(2.4) in non high risk patients (p=0.87) and 41.2%(11.9) vs 53.2%(4.0) in high risk patients (p=0.24). Conclusions: The outcome of T-ALL patients treated with BFM-type therapy is comparable in ETP and non-ETP for those with good response to initial chemotherapy; it was slightly, but not significantly, inferior in ETP-ALL patients with poor initial response. Phase IB treatment element is very effective in ETP-ALL, suggesting that intensification with antimetabolite and alkylating agents may be beneficial, while the benefit of HSCT in first CR needs to be further investigated. Disclosures No relevant conflicts of interest to declare.


2019 ◽  
Vol 6 (Supplement_2) ◽  
pp. S833-S833
Author(s):  
Steven W Johnson ◽  
David H Priest ◽  
Shannon V Brown

Abstract Background Studies suggest oral vancomycin prophylaxis may be effective in preventing Clostridioides difficile infection. These studies are limited by their retrospective design, reliance on local clinical practice patterns, lack of intervention standardization, and limited risk stratification. We sought to evaluate the effectiveness of oral vancomycin for the prevention of healthcare facility-onset CDI (HCFO-CDI) in high-risk patients. Methods We conducted a randomized, prospective, open-label study at Novant Health Forsyth Medical Center in Winston-Salem, North Carolina between October 2018 and April 2019. Admitted high-risk patients (defined as: ≥ 60 years of age, hospitalized ≤ 30 days prior to the index hospitalization and received systemic antibiotics during that prior hospitalization and currently receiving systemic antibiotics) were randomized 1:1 to either oral vancomycin (dosed at 125 mg once daily while receiving systemic antibiotics and continued for 5 days post completion of systemic antibiotics [OVP]), or no prophylaxis. The primary endpoint was incidence of HCFO-CDI. Secondary endpoints included incidence of community-onset healthcare facility-associated CDI (CO-HCFA-CDI), development of VRE colonization after receiving OVP, and adverse effects and cost of OVP. Results A total of 100 patients were evaluated, 50 patients in each group. Baseline and hospitalization characteristics were similar in each group. No incidents of HCFO-CDI were diagnosed in the OVP group compared with 6 (12%) in the no prophylaxis group (P = 0.03). CO-HCFA-CDI was not observed in either group. No patients developed new VRE colonization with only 1 patient reporting mild gastrointestinal side-effects to OVP. A total of 600 doses of OVP were given during the study, with each patient receiving an average of 12 doses. Total acquisition cost of OVP was $728.25, $60.69 per patient. Conclusion OVP was highly effective in preventing HCFO-CDI. OVP was well tolerated with no apparent risk for VRE colonization. Further prospective investigation is warranted to determine the impact and cost-effectiveness of routine use of OVP in high-risk patients. Disclosures All authors: No reported disclosures.


Sign in / Sign up

Export Citation Format

Share Document