1
|
Recent Advances in Treatment Options for Childhood Acute Lymphoblastic Leukemia. Cancers (Basel) 2022; 14:cancers14082021. [PMID: 35454927 PMCID: PMC9032060 DOI: 10.3390/cancers14082021] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Acute lymphoblastic leukemia is the most common blood cancer in pediatric patients. Despite the enormous progress in ALL treatment, which is reflected by a high 5-year overall survival rate that reaches up to 96% in the most recent studies, there are still patients that cannot be saved. Treatment of ALL is based on conventional methods, including chemotherapy and radiotherapy. These methods carry with them the risk of very high toxicities. Severe complications related to conventional therapies decrease their effectiveness and can sometimes lead to death. Therefore, currently, numerous studies are being carried out on novel forms of treatment. In this work, classical methods of treatment have been summarized. Furthermore, novel treatment methods and the possibility of combining them with chemotherapy have been incorporated into the present work. Targeted treatment, CAR-T-cell therapy, and immunotherapy for ALL have been described. Treatment options for the relapse/chemoresistance ALL have been presented. Abstract Acute lymphoblastic leukemia is the most common blood cancer in pediatric patients. There has been enormous progress in ALL treatment in recent years, which is reflected by the increase in the 5-year OS from 57% in the 1970s to up to 96% in the most recent studies. ALL treatment is based primarily on conventional methods, which include chemotherapy and radiotherapy. Their main weakness is severe toxicity, which prompts dose reduction, decreases the effectiveness of the treatment, and, in some cases, can lead to death. Currently, numerous modifications in treatment regimens are applied in order to limit toxicities emerging from conventional approaches and improve outcomes. Hematological treatment of pediatric patients is reaching for more novel treatment options, such as targeted treatment, CAR-T-cells therapy, and immunotherapy. These methods are currently used in conjunction with chemotherapy. Nevertheless, the swift progress in their development and increasing efficacity can lead to applying those novel therapies as standalone therapeutic options for pediatric ALL.
Collapse
|
2
|
van Binsbergen AL, de Haas V, van der Velden VHJ, de Groot-Kruseman HA, Fiocco MF, Pieters R. Efficacy and toxicity of high-risk therapy of the Dutch Childhood Oncology Group in childhood acute lymphoblastic leukemia. Pediatr Blood Cancer 2022; 69:e29387. [PMID: 34648216 DOI: 10.1002/pbc.29387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 11/12/2022]
Abstract
BACKGROUND Children with acute lymphoblastic leukemia (ALL) and high-risk (HR) features have a poor outcome and are treated with HR blocks, often followed by allogenic stem cell transplantation (SCT). PROCEDURE This article analyses the outcomes of children treated with HR blocks between 2004 and 2017 according to DCOG ALL10/11 protocols. 1297 patients with newly diagnosed ALL were consecutively enrolled, of which 107 met the HR criteria (no complete remission; minimal residual disease (MRD) > 10-3 after consolidation; "MLL-AF4" translocation and in ALL-10 also poor prednisone response). Patients were treated with one induction and consolidation course followed by three HR chemotherapy blocks, after which they received either SCT or further chemotherapy. MRD levels were measured at end of induction, consolidation, and after each HR block. RESULTS At five years, the event-free survival was 72.8% (95% CI, 64.6-82.0), and the cumulative incidence of relapse was 13.0% (95% CI, 6.3-19.8). Patients with only negative or low-positive MRD levels during HR blocks had a significantly lower five-year cumulative incidence of relapse (CIR) of 2.2% (95% CI, 0-6.6) compared with patients with one or more high-positive MRD levels (CIR 15.4%; 95% CI, 3.9-26.9). During the entire treatment protocol, 11.2% of patients died due to toxicity. CONCLUSIONS The high survival with HR blocks seems favorable compared with other studies. However, the limit of treatment intensification might have been reached as the number of patients dying from leukemia relapse is about equal as the number of patients dying from toxicity. Patients with negative or low MRD levels during HR blocks have lower relapse rates.
Collapse
Affiliation(s)
| | - Valérie de Haas
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Hester A de Groot-Kruseman
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Dutch Childhood Oncology Group (DCOG), Utrecht, The Netherlands
| | - Marta F Fiocco
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Leiden University, Mathematical Institute, The Netherlands
| | - Rob Pieters
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
3
|
Ramos-Peñafiel C, Olarte-Carrillo I, Maldonado RC, de la Cruz Rosas A, Collazo-Jaloma J, Martínez-Tovar A. Association of three factors (ABCB1 gene expression, steroid response, early response at day + 8) on the response to induction in patients with acute lymphoblastic leukemia. Ann Hematol 2020; 99:2629-2637. [PMID: 32980890 DOI: 10.1007/s00277-020-04277-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022]
Abstract
Treatment of acute lymphoblastic leukemia (ALL) requires the combination of multiple drugs to integrate a complete remission. The different prognostic factors (age, leukocytes, risk, cytogenetic alterations) allow identifying those patients with a high risk of relapse, but there are few described factors that impact the induction response. The objective was to identify the utility of different risk factors (overexpression of the ABCB1 drug resistance gene, favorable response to steroids (FRS) and early response at day + 8 of treatment) on the percentage of complete remissions and overall survival. This is a prospective, observational study in adult patients with B-ALL without specific cytogenetic alterations, who started induction treatment based on a pretreatment with prednisone and subsequently vincristine (1.6 mg/m2 subcutaneous) plus daunorubicin (45 mg/m2 subcutaneously) on days + 1, + 8, + 15. The ABCB1 resistance gene was evaluated at diagnosis, the FRS at the end of the pretreatment and the early response during day + 8. A total of 53 adult patients diagnosed with ALL Philadelphia negative chromosome (Ph-), with immunophenotype B, with a normal karyotype, were studied. Cases with genetic abnormalities with a poor prognosis were excluded in order to reduce bias. The mean age was 48 years (range 17-68 years). 62.3% of patients were at high risk of relapse. When analyzing the risk factors, 30.2% showed high levels of the ABCB1 resistance gene, without showing an impact on the induction response (OR: 1.218, p = 0.743), but its overexpression was associated with a poor response to steroids as in the absence of early response. Individually, both the FRS (OR: 5.7, p = 0.004) and the absence of early response to day + 8 (OR: 6.42, p = 0.002) showed significance. By combining the different factors, having more than 2 was directly related to a failure (OR: 9.514, p = 0.000). The identification of factors such as FRS such as the persistence of blasts at the end of the first week of treatment is useful to identify patients at risk of failure in induction.
Collapse
Affiliation(s)
- Christian Ramos-Peñafiel
- Servicio de Hematología, Hospital General de México, "Dr. Eduardo Liceaga", 06726, Ciudad de México, Mexico
| | - Irma Olarte-Carrillo
- Laboratorio de Biología Molecular, Servicio de Hematología, Hospital General de México, "Dr. Eduardo Liceaga", Ciudad de México, Mexico
| | - Rafael Cerón Maldonado
- Laboratorio de Biología Molecular, Servicio de Hematología, Hospital General de México, "Dr. Eduardo Liceaga", Ciudad de México, Mexico
| | - Adrián de la Cruz Rosas
- Laboratorio de Biología Molecular, Servicio de Hematología, Hospital General de México, "Dr. Eduardo Liceaga", Ciudad de México, Mexico
| | - Juan Collazo-Jaloma
- Servicio de Hematología, Hospital General de México, "Dr. Eduardo Liceaga", 06726, Ciudad de México, Mexico
| | - Adolfo Martínez-Tovar
- Servicio de Hematología, Hospital General de México, "Dr. Eduardo Liceaga", 06726, Ciudad de México, Mexico.
| |
Collapse
|
4
|
Vega-García N, Perez-Jaume S, Esperanza-Cebollada E, Vicente-Garcés C, Torrebadell M, Jiménez-Velasco A, Ortega M, Llop M, Abad L, Vagace JM, Minguela A, Pratcorona M, Sánchez-Garcia J, García-Calderón CB, Gómez-Casares MT, Martín-Clavero E, Escudero A, Riñón Martinez-Gallo M, Muñoz L, Velasco MR, García-Morin M, Català A, Pascual A, Velasco P, Fernández JM, Lassaletta A, Fuster JL, Badell I, Molinos-Quintana Á, Molinés A, Guerra-García P, Pérez-Martínez A, García-Abós M, Robles Ortiz R, Pisa S, Adán R, Díaz de Heredia C, Dapena JL, Rives S, Ramírez-Orellana M, Camós M. Measurable Residual Disease Assessed by Flow-Cytometry Is a Stable Prognostic Factor for Pediatric T-Cell Acute Lymphoblastic Leukemia in Consecutive SEHOP Protocols Whereas the Impact of Oncogenetics Depends on Treatment. Front Pediatr 2020; 8:614521. [PMID: 33614543 PMCID: PMC7892614 DOI: 10.3389/fped.2020.614521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022] Open
Abstract
Robust and applicable risk-stratifying genetic factors at diagnosis in pediatric T-cell acute lymphoblastic leukemia (T-ALL) are still lacking, and most protocols rely on measurable residual disease (MRD) assessment. In our study, we aimed to analyze the impact of NOTCH1, FBXW7, PTEN, and RAS mutations, the measurable residual disease (MRD) levels assessed by flow cytometry (FCM-MRD) and other reported risk factors in a Spanish cohort of pediatric T-ALL patients. We included 199 patients treated with SEHOP and PETHEMA consecutive protocols from 1998 to 2019. We observed a better outcome of patients included in the newest SEHOP-PETHEMA-2013 protocol compared to the previous SHOP-2005 cohort. FCM-MRD significantly predicted outcome in both protocols, but the impact at early and late time points differed between protocols. The impact of FCM-MRD at late time points was more evident in SEHOP-PETHEMA 2013, whereas in SHOP-2005 FCM-MRD was predictive of outcome at early time points. Genetics impact was different in SHOP-2005 and SEHOP-PETHEMA-2013 cohorts: NOTCH1 mutations impacted on overall survival only in the SEHOP-PETHEMA-2013 cohort, whereas homozygous deletions of CDKN2A/B had a significantly higher CIR in SHOP-2005 patients. We applied the clinical classification combining oncogenetics, WBC count and MRD levels at the end of induction as previously reported by the FRALLE group. Using this score, we identified different subgroups of patients with statistically different outcome in both Spanish cohorts. In SHOP-2005, the FRALLE classifier identified a subgroup of high-risk patients with poorer survival. In the newest protocol SEHOP-PETHEMA-2013, a very low-risk group of patients with excellent outcome and no relapses was detected, with borderline significance. Overall, FCM-MRD, WBC count and oncogenetics may refine the risk-stratification, helping to design tailored approaches for pediatric T-ALL patients.
Collapse
Affiliation(s)
- Nerea Vega-García
- Haematology Laboratory, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain.,Developmental Tumor Biology Group, Leukemia and Other Pediatric Hemopathies, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Sara Perez-Jaume
- Developmental Tumour Biology Laboratory, Institut de Recerca Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
| | - Elena Esperanza-Cebollada
- Haematology Laboratory, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain.,Developmental Tumor Biology Group, Leukemia and Other Pediatric Hemopathies, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Clara Vicente-Garcés
- Haematology Laboratory, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain.,Developmental Tumor Biology Group, Leukemia and Other Pediatric Hemopathies, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Montserrat Torrebadell
- Haematology Laboratory, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain.,Developmental Tumor Biology Group, Leukemia and Other Pediatric Hemopathies, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Margarita Ortega
- Cytogenetics Unit, Hematology Department, Hospital Vall d'Hebron, Barcelona, Spain
| | - Marta Llop
- Molecular Biology Unit, Clinical Analysis Service, La Fe University and Polytechnic Hospital, Valencia, Spain.,Centro de Investigación Biomédica en Red - Cáncer (CIBERONC CB16/12/00284), Madrid, Spain
| | - Lorea Abad
- Paediatric Hemato-Oncology Laboratory, Hospital Niño Jesús, Madrid, Spain
| | | | - Alfredo Minguela
- Immunology Service, Clinic University Hospital Virgen de la Arrixaca (HCUVA) and Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Marta Pratcorona
- Haematology Laboratory, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Clara B García-Calderón
- Instituto de Biomedicina de Sevilla (IBIS/Consejo Superior de Investigaciones Científicas (CSIC)/Centro de Investigación Biomédica en Red - Cáncer (CIBERONC)), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain
| | - María Teresa Gómez-Casares
- Biology and Molecular Haematology and Hemotherapy Service, Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas de Gran Canarias, Spain
| | - Estela Martín-Clavero
- Haematology-Cytology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Adela Escudero
- Translational Research in Pediatric Oncology Hematopoietic Transplantation and Cell Therapy, Institute of Medical and Molecular Genetics (INGEMM), Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | | | - Luz Muñoz
- Haematology Laboratory, Hospital Parc Taulí, Sabadell, Spain
| | | | - Marina García-Morin
- Paediatric Hematology Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Albert Català
- Developmental Tumor Biology Group, Leukemia and Other Pediatric Hemopathies, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,Paediatric Hematology and Oncology Departments, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | | | - Pablo Velasco
- Pediatric Hematology and Oncology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - José Mª Fernández
- Haematology and Oncology Department, Hospital de La Fe, Valencia, Spain
| | - Alvaro Lassaletta
- Haematology and Oncology Department, Hospital Niño Jesús, Madrid, Spain
| | - José Luis Fuster
- Paediatric Oncohematology Department, Clinic University Hospital Virgen de la Arrixaca (HCUVA) and Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, Spain
| | - Isabel Badell
- Paediatric Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Águeda Molinos-Quintana
- Instituto de Biomedicina de Sevilla (IBIS/Consejo Superior de Investigaciones Científicas (CSIC)/Centro de Investigación Biomédica en Red - Cáncer (CIBERONC)), Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Seville, Spain
| | - Antonio Molinés
- Unit of Hematology and Hemotherapy, H.U. Materno Infantil de Canarias, Canarias, Spain
| | - Pilar Guerra-García
- Paediatric Hemato-Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.,Department of Pediatric Hemato-Oncology and Stem Cell Transplantation, La Paz University Hospital, Madrid, Spain
| | - Antonio Pérez-Martínez
- Translational Research in Pediatric Oncology Hematopoietic Transplantation and Cell Therapy, Institute of Medical and Molecular Genetics (INGEMM), Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain.,Department of Pediatric Hemato-Oncology and Stem Cell Transplantation, La Paz University Hospital, Madrid, Spain
| | - Miriam García-Abós
- Pediatric Onco-Hematology Department, Hospital Universitario Donostia, Donostia, Spain
| | - Reyes Robles Ortiz
- Pediatric Onco-Hematology Department, Complejo Hospitalario de Navarra, Navarra, Spain
| | - Sandra Pisa
- Paediatric Hematology Department, Hospital Parc Taulí, Sabadell, Spain
| | - Rosa Adán
- Haematology and Oncology Department, Hospital de Cruces, Bilbao, Spain
| | - Cristina Díaz de Heredia
- Pediatric Hematology and Oncology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - José Luis Dapena
- Developmental Tumor Biology Group, Leukemia and Other Pediatric Hemopathies, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Paediatric Hematology and Oncology Departments, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | - Susana Rives
- Developmental Tumor Biology Group, Leukemia and Other Pediatric Hemopathies, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,Paediatric Hematology and Oncology Departments, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain
| | | | - Mireia Camós
- Haematology Laboratory, Hospital Sant Joan de Déu, University of Barcelona, Barcelona, Spain.,Developmental Tumor Biology Group, Leukemia and Other Pediatric Hemopathies, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Randomized post-induction and delayed intensification therapy in high-risk pediatric acute lymphoblastic leukemia: long-term results of the international AIEOP-BFM ALL 2000 trial. Leukemia 2019; 34:1694-1700. [DOI: 10.1038/s41375-019-0670-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/26/2019] [Accepted: 11/17/2019] [Indexed: 12/14/2022]
|
6
|
Yu Z, Liu L, Shu Q, Li D, Wang R. Leukemia stem cells promote chemoresistance by inducing downregulation of lumican in mesenchymal stem cells. Oncol Lett 2019; 18:4317-4327. [PMID: 31579426 DOI: 10.3892/ol.2019.10767] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 04/15/2019] [Indexed: 12/14/2022] Open
Abstract
Leukemia stem cells (LSCs) are responsible for therapeutic failure and relapse of acute lymphoblastic leukemia. As a result of the interplay between LSCs and bone marrow mesenchymal stem cells (BM-MSCs), cancer cells may escape from chemotherapy and immune surveillance, thereby promoting leukemia progress and relapse. The present study identified that the crosstalk between LSCs and BM-MSCs may contribute to changes of immune phenotypes and expression of hematopoietic factors in BM-MSCs. Furthermore, Illumina Genome Analyzer/Hiseq 2000 identified 7 differentially expressed genes between BM-MSCsLSC and BM-MSCs. The Illumina sequencing results were further validated by reverse transcription-quantitative polymerase chain reaction. Following LSC simulation, 2 genes were significantly upregulated, whereas the remaining 2 genes were significantly downregulated in MSCs. The most remarkable changes were identified in the expression levels of lumican (LUM) gene. These results were confirmed by western blot analysis. In addition, decreased LUM expression led to decreased apoptosis, and promoted chemoresistance to VP-16 in Nalm-6 cells. These results suggest that downregulation of LUM expression in BM-MSCs contribute to the anti-apoptotic properties and resistance to chemotherapy in LSCs.
Collapse
Affiliation(s)
- Zhen Yu
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lin Liu
- Department of Pediatrics, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qiang Shu
- Department of Immunology, Shenzhen Research Institute of Shandong University, Shenzhen, Guangdong 518057, P.R. China
| | - Dong Li
- Department of Immunology, Shenzhen Research Institute of Shandong University, Shenzhen, Guangdong 518057, P.R. China
| | - Ran Wang
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
7
|
Andjelković-Apostolović M, Ignjatović A, Stojanović M, Milošević Z, Apostolović B, Topalović M, Simić D. A JOINPOINT REGRESSION ANALYSIS OF LONG-TERM TRENDS IN LEUKEMIA INCIDENCE AND MORTALITY IN CENTRAL SERBIA AND NIŠAVA DISTRICT (1999-2014). ACTA MEDICA MEDIANAE 2019. [DOI: 10.5633/amm.2019.0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
8
|
Impact of an animal-assisted therapy programme on physiological and psychosocial variables of paediatric oncology patients. PLoS One 2018; 13:e0194731. [PMID: 29617398 PMCID: PMC5884536 DOI: 10.1371/journal.pone.0194731] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/08/2018] [Indexed: 11/19/2022] Open
Abstract
The objective of this study was to propose an intervention and safety protocol for performing animal-assisted therapy (AAT) and evaluating its efficacy in children under outpatient oncological treatment based on psychological, physiological, and quality of life indicators for the children and caregivers. The sample consisted of 24 children diagnosed with leukaemia and solid tumours (58% girls with a mean age of 8.0 years) who underwent an AAT programme consisting of three 30-min sessions in an open group. Two dogs (one Labrador retriever and one golden retriever) were used, and activities such as sensory stimulation, gait training, and socialization were conducted. The exclusion criteria were severe mental problems, inability to answer the questions included in the instruments used, allergy to animals, unavailability/lack of interest, isolation precaution, surgical wound, use of invasive devices, ostomy, no current blood count for evaluation, neutropaenia, infection, fever, diarrhoea, vomiting, respiratory symptoms at the beginning of the intervention or 1 week before the intervention, hospitalization or scheduled surgery, and non-completion of the AAT programme. The variables analysed using validated self or other evaluations were stress, pain, mood, anxiety, depression, quality of life, heart rate, and blood pressure. A quasi-experimental study design was used. We observed a decrease in pain (p = 0.046, d = –0.894), irritation (p = 0.041, d = –0.917), and stress (p = 0.005; d = –1.404) and a tendency towards improvement of depressive symptoms (p = 0.069; d = –0.801). Among the caregivers, an improvement was observed in anxiety (p = 0.007, d = –1.312), mental confusion (p = 0.006, d = –1.350), and tension (p = 0.006, d = –1.361). Therefore, the selection criteria and care protocols used for the AAT programme in the oncological context were adequate, and the programme was effective.
Collapse
|
9
|
Protocol II vs protocol III given twice during reinduction therapy in children with medium-risk ALL. Blood 2017; 130:2146-2149. [DOI: 10.1182/blood-2017-05-782086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
10
|
Lee JW, Kim SK, Jang PS, Jeong DC, Chung NG, Cho B, Kim HK. Treatment of children with acute lymphoblastic leukemia with risk group based intensification and omission of cranial irradiation: A Korean study of 295 patients. Pediatr Blood Cancer 2016; 63:1966-73. [PMID: 27463364 DOI: 10.1002/pbc.26136] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/03/2016] [Accepted: 06/15/2016] [Indexed: 11/05/2022]
Abstract
BACKGROUND Recent studies indicate 70-80% event-free survival (EFS) for pediatric acute lymphoblastic leukemia (ALL). In this study, we report the outcome of 295 children and adolescents treated at our institution, with stratification into four risk groups, and omission of cranial irradiation in all patients. PROCEDURE Patients were diagnosed from January 2005 to December 2011 and classified and treated as low, standard, high, and very high risk groups. A delayed intensification phase was given twice for high and very high risk groups. None of the patients received cranial irradiation for central nervous system (CNS) prophylaxis. RESULTS The 10-year EFS and overall survival (OS) were 78.5 ± 2.5% and 81.9 ± 2.7%, respectively. EFS according to risk group was as follows: low risk 91.2 ± 3.7%, standard risk 98.1 ± 1.9%, high risk 81.5 ± 4.3%, very high risk 59.4 ± 5.3%. In a multivariate analysis, high hyperdiploidy and infant ALL were significant predictors of EFS. Cumulative incidence of any relapse, isolated CNS relapse, and any CNS relapse were 17.1 ± 2.3%, 1.5 ± 0.7%, and 2.3 ± 0.9%, respectively. Other events included infection-related deaths during remission induction chemotherapy (3), primary refractory disease (2), and treatment-related deaths in first complete remission (8). CONCLUSIONS In this single-institution study of Korean pediatric ALL patients, risk group based intensification with omission of cranial irradiation resulted in EFS comparable to previous studies, excellent survival of low- and standard-risk patients, and a low rate of CNS relapse.
Collapse
Affiliation(s)
- Jae Wook Lee
- Division of Hematology and Oncology, Department of Pediatrics, The Catholic University of Korea, Seoul, Korea
| | - Seong-Koo Kim
- Division of Hematology and Oncology, Department of Pediatrics, The Catholic University of Korea, Seoul, Korea
| | - Pil-Sang Jang
- Division of Hematology and Oncology, Department of Pediatrics, The Catholic University of Korea, Seoul, Korea
| | - Dae-Chul Jeong
- Division of Hematology and Oncology, Department of Pediatrics, The Catholic University of Korea, Seoul, Korea
| | - Nack-Gyun Chung
- Division of Hematology and Oncology, Department of Pediatrics, The Catholic University of Korea, Seoul, Korea
| | - Bin Cho
- Division of Hematology and Oncology, Department of Pediatrics, The Catholic University of Korea, Seoul, Korea.
| | - Hack-Ki Kim
- Division of Hematology and Oncology, Department of Pediatrics, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
11
|
Matloub Y, Stork L, Asselin B, Hunger SP, Borowitz M, Jones T, Bostrom B, Gastier-Foster JM, Heerema NA, Carroll A, Winick N, Carroll WL, Camitta B, Devidas M, Gaynon PS. Outcome of Children with Standard-Risk T-Lineage Acute Lymphoblastic Leukemia--Comparison among Different Treatment Strategies. Pediatr Blood Cancer 2016; 63:255-61. [PMID: 26485054 PMCID: PMC4715507 DOI: 10.1002/pbc.25793] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/26/2015] [Accepted: 09/11/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND Children with T-lineage acute lymphoblastic leukemia ALL (T-ALL) historically have had inferior outcomes compared with the children with precursor-B ALL (B-ALL). After 1995, the Children's Cancer Group (CCG) treated patients with B- and T-ALL according to the National Cancer Institute (NCI) risk criteria, basing risk stratification on age and white blood cell (WBC) count regardless of immunophenotype. The Pediatric Oncology Group (POG) treated all the patients with T-ALL on separate, generally more intensive protocols than those used to treat the patients with B-ALL. PROCEDURE We compared the outcomes of children with T-ALL and NCI standard-risk (SR) criteria treated on CCG and POG trials between 1996 and 2005. CCG SR-ALL 1952 and 1991 enrolled 80 and 86 patients with T-ALL, respectively, utilizing a reduced intensity Berlin-Frankfurt-Münster backbone. Treatment was intensified for slow early responders and only patients with overt central nervous system leukemia received cranial irradiation. Eighty-four patients with T-ALL and SR features were enrolled on POG 9404 comprising more intensive therapy with all patients receiving cranial irradiation. RESULTS The 7-year event-free survival (EFS) for patients with SR T-ALL on CCG 1952, CCG 1991, and POG 9404 were 74.1 ± 5.8%, 81.8 ± 5.3%, and 84.2 ± 4.3%, respectively (P = 0.18). Overall 7-year survivals were 86.1 ± 4.6%, 88.3 ± 4.4%, 89.1 ± 3.6%, respectively (P = 0.84). CONCLUSIONS Comparable high rates of EFS and long-term survival were achieved with all three regimens, with the CCG regimens utilizing a less intensive chemotherapy backbone without prophylactic cranial irradiation for patients with SR T-ALL.
Collapse
Affiliation(s)
- Yousif Matloub
- Rainbow Babies & Children’s Hospital, Division of Hematology-Oncology, Case Western Reserve University, Cleveland, Ohio
| | - Linda Stork
- Doernbecher Children’s Hospital, Division of Hematology-Oncology, Oregon Health & Science University, Portland, Oregon
| | - Barbara Asselin
- Golisano Children’s Hospital, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | | | - Michael Borowitz
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Tamekia Jones
- Children’s Foundation Research Institute, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Bruce Bostrom
- Children’s Hospitals and Clinics of Minnesota, Minneapolis, Minnesota
| | - Julie M. Gastier-Foster
- Department of Pathology and Laboratory Medicine, Nationwide Children’s Hospital, Columbus, Ohio
- Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| | - Nyla A. Heerema
- Department of Pathology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Andrew Carroll
- Department of Genetics, University of Alabama at Birmingham, Alabama
| | - Naomi Winick
- Division of Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Bruce Camitta
- Midwest Center for Cancer and Blood Disorders, Pediatric Hematology-Oncology, Milwaukee, Wisconsin
| | - Meenakshi Devidas
- Department of Biostatistics, Colleges of Medicine, Public Health & Health Professions, Children’s University of Florida, Gainsville, Florida
| | - Paul S. Gaynon
- Division of Hematology-Oncology, Children’s Hospital Los Angeles, University of Southern California, Los Angeles, California
| |
Collapse
|
12
|
Childhood high-risk acute lymphoblastic leukemia in first remission: results after chemotherapy or transplant from the AIEOP ALL 2000 study. Blood 2014; 123:1470-8. [DOI: 10.1182/blood-2013-10-532598] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Key Points
Intensive BFM therapy is effective for HR childhood ALL if low MRD levels are achieved at the end of the induction/consolidation phase. Childhood ALL with high MRD levels at the end of induction/consolidation phase has a poor prognosis despite intensive BFM therapy or HSCT.
Collapse
|
13
|
Yokosuka T, Goto H, Fujii H, Naruto T, Takeuchi M, Tanoshima R, Kato H, Yanagimachi M, Kajiwara R, Yokota S. Flow cytometric chemosensitivity assay using JC‑1, a sensor of mitochondrial transmembrane potential, in acute leukemia. Cancer Chemother Pharmacol 2014; 72:1335-42. [PMID: 24121478 DOI: 10.1007/s00280-013-2303-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 09/25/2013] [Indexed: 10/26/2022]
Abstract
PURPOSE The purpose of the study is to establish a simple and relatively inexpensive flow cytometric chemosensitivity assay (FCCA) for leukemia to distinguish leukemic blasts from normal leukocytes in clinical samples. METHODS We first examined whether the FCCA with the mitochondrial membrane depolarization sensor, 5, 50, 6, 60-tetrachloro-1, 10, 3, 30 tetraethyl benzimidazolo carbocyanine iodide (JC-1), could detect drug-induced apoptosis as the conventional FCCA by annexin V/7-AAD detection did and whether it was applicable in the clinical samples. Second, we compared the results of the FCCA for prednisolone (PSL) with clinical PSL response in 18 acute lymphoblastic leukemia (ALL) patients to evaluate the reliability of the JC-1 FCCA. Finally, we performed the JC-1 FCCA for bortezomib (Bor) in 25 ALL or 11 acute myeloid leukemia (AML) samples as the example of the clinical application of the FCCA. RESULTS In ALL cells, the results of the JC-1 FCCA for nine anticancer drugs were well correlated with those of the conventional FCCA using anti-annexin V antibody (P < 0.001). In the clinical samples from 18 children with ALL, the results of the JC-1 FCCA for PSL were significantly correlated with the clinical PSL response (P = 0.005). In ALL samples, the sensitivity for Bor was found to be significantly correlated with the sensitivity for PSL (P = 0.005). In AML samples, the Bor sensitivity was strongly correlated with the cytarabine sensitivity (P = 0.0003). CONCLUSIONS This study showed the reliability of a relatively simple and the FCCA using JC-1, and the possibility for the further clinical application.
Collapse
|
14
|
Stary J, Zimmermann M, Campbell M, Castillo L, Dibar E, Donska S, Gonzalez A, Izraeli S, Janic D, Jazbec J, Konja J, Kaiserova E, Kowalczyk J, Kovacs G, Li CK, Magyarosy E, Popa A, Stark B, Jabali Y, Trka J, Hrusak O, Riehm H, Masera G, Schrappe M. Intensive chemotherapy for childhood acute lymphoblastic leukemia: results of the randomized intercontinental trial ALL IC-BFM 2002. J Clin Oncol 2013; 32:174-84. [PMID: 24344215 DOI: 10.1200/jco.2013.48.6522] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PURPOSE From 2002 to 2007, the International Berlin-Frankfurt-Münster Study Group conducted a prospective randomized clinical trial (ALL IC-BFM 2002) for the management of childhood acute lymphoblastic leukemia (ALL) in 15 countries on three continents. The aim of this trial was to explore the impact of differential delayed intensification (DI) on outcome in all risk groups. PATIENTS AND METHODS For this trial, 5,060 eligible patients were divided into three risk groups according to age, WBC, early treatment response, and unfavorable genetic aberrations. DI was randomized as follows: standard risk (SR), two 4-week intensive elements (protocol III) versus one 7-week protocol II; intermediate risk (IR), protocol III × 3 versus protocol II × 1; high risk (HR), protocol III × 3 versus either protocol II × 2 (Associazione Italiana Ematologia Oncologia Pediatrica [AIEOP] option), or 3 HR blocks plus single protocol II (Berlin-Frankfurt-Münster [BFM] option). RESULTS At 5 years, the probabilities of event-free survival and survival were 74% (± 1%) and 82% (± 1%) for all 5,060 eligible patients, 81% and 90% for the SR (n = 1,564), 75% and 83% for the IR (n = 2,650), and 55% and 62% for the HR (n = 846) groups, respectively. No improvement was accomplished by more intense and/or prolonged DI. CONCLUSION The ALL IC-BFM 2002 trial is a good example of international collaboration in pediatric oncology. A wide platform of countries able to run randomized studies in ALL has been established. Although the alternative DI did not improve outcome compared with standard treatment and the overall results are worse than those achieved by longer established leukemia groups, the national results have generally improved.
Collapse
Affiliation(s)
- Jan Stary
- Jan Stary, Jan Trka, and Ondrej Hrusak, Charles University and University Hospital Motol, Prague; Yahia Jabali, Regional Hospital, Ceske Budejovice, Czech Republic; Martin Zimmermann and Hansjörg Riehm, Medical School Hannover, Hannover; Martin Schrappe, University Hospital Schleswig-Holstein, Kiel, Germany; Myriam Campbell, Roberto del Rio Hospital, Universidad de Chile, Santiago, Chile; Luis Castillo, Hospital Pereira Rossell, Montevideo, Uruguay; Eduardo Dibar, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina; Svetlana Donska, Regional Oncologic Hospital, Kiev, Ukraine; Alejandro Gonzalez, Institute of Hematology and Immunology, La Habana, Cuba; Shai Izraeli, Sheba Medical Center of Israel, Sackler School of Medicine, Tel Aviv University, Tel Hashomer; Batia Stark, Schneider Children's Medical Center of Israel, Sackler School of Medicine, Tel Aviv University, Petah-Tikva, Israel; Dragana Janic, University Children's Hospital, University of Belgrade, Belgrade, Serbia; Janez Jazbec, University Children's Hospital, Ljubljana, Slovenia; Josip Konja, University Hospital Centre Rebro, Zagreb, Croatia; Emilia Kaiserova, University Children's Hospital, Bratislava, Slovakia; Jerzy Kowalczyk, University of Lublin, Lublin, Poland; Gabor Kovacs and Edina Magyarosy, Semmelweis University, Budapest, Hungary; Chi-Kong Li, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, Special Administrative Region, People's Republic of China; Alexander Popa, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia; and Giuseppe Masera, Ospedale S. Gerardo, University of Milano-Bicocca, Monza, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Kato M, Koh K, Manabe A, Saito T, Hasegawa D, Isoyama K, Kinoshita A, Maeda M, Okimoto Y, Kajiwara M, Kaneko T, Sugita K, Kikuchi A, Tsuchida M, Ohara A. No impact of high-dose cytarabine and asparaginase as early intensification with intermediate-risk paediatric acute lymphoblastic leukaemia: results of randomized trial TCCSG study L99-15. Br J Haematol 2013; 164:376-83. [DOI: 10.1111/bjh.12632] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/18/2013] [Indexed: 11/29/2022]
Affiliation(s)
- Motohiro Kato
- Department of Haematology/Oncology; Saitama Children's Medical Centre; Saitama Japan
- Department of Paediatrics; the University of Tokyo; Tokyo Japan
| | - Katsuyoshi Koh
- Department of Haematology/Oncology; Saitama Children's Medical Centre; Saitama Japan
| | - Atsushi Manabe
- Department of Paediatrics; St. Luke's International Hospital; Tokyo Japan
| | - Tomohiro Saito
- Department of Health Policy; National Research Institute for Child Health and Development; Tokyo Japan
| | - Daisuke Hasegawa
- Department of Paediatrics; St. Luke's International Hospital; Tokyo Japan
| | - Keiichi Isoyama
- Department of Paediatrics; Showa University Fujigaoka Hospital; Yokohama Japan
| | - Akitoshi Kinoshita
- Department of Paediatrics; St. Marianna University School of Medicine; Kawasaki Japan
| | - Miho Maeda
- Department of Paediatrics; Nippon Medical School; Tokyo Japan
| | - Yuri Okimoto
- Department of Haematology/Oncology; Chiba Children's Hospital; Chiba Japan
| | - Michiko Kajiwara
- Department of Paediatrics; Tokyo Medical and Dental University; Tokyo Japan
| | - Takashi Kaneko
- Department of Haematology/Oncology; Tokyo Metropolitan Children's Medical Centre; Tokyo Japan
| | - Kanji Sugita
- Department of Paediatrics; University of Yamanashi; Yamanashi Japan
| | - Akira Kikuchi
- Department of Paediatrics; Teikyo University; Tokyo Japan
| | - Masahiro Tsuchida
- Department of Paediatrics; Ibaraki Children's Hospital; Ibaraki Japan
| | - Akira Ohara
- Department of Paediatrics; Toho University; Tokyo Japan
| |
Collapse
|
16
|
Wang D, Zhu G, Wang N, Zhou X, Yang Y, Zhou S, Xiong J, He J, Jiang L, Li C, Xu D, Huang L, Zhou J. SIL-TAL1 rearrangement is related with poor outcome: a study from a Chinese institution. PLoS One 2013; 8:e73865. [PMID: 24040098 PMCID: PMC3767609 DOI: 10.1371/journal.pone.0073865] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 07/30/2013] [Indexed: 11/18/2022] Open
Abstract
SIL-TAL1 rearrangement is common in T-cell acute lymphoblastic leukemia (T-ALL), however its prognostic implication remains controversial. To investigate the clinical characteristics and outcome of this subtype in Chinese population, we systemically reviewed 62 patients with newly diagnosed T-ALL, including 15 patients with SIL-TAL1 rearrangement. We found that SIL-TAL1(+) T-ALL was characterized by higher white blood cell count (P = 0.029) at diagnosis, predominant cortical T-ALL immunophenotype (P = 0.028) of the leukemic blasts, and a higher prevalence of tumor lysis syndrome (TLS, P<0.001) and disseminated intravascular coagulation (DIC, P<0.001), which led to a higher early mortality (P = 0.011). Compared with SIL-TAL1(-) patients, SIL-TAL1(+) patients had shorter relapse free survival (P = 0.007) and overall survival (P = 0.002). Our NOD/SCID xenotransplantation model also demonstrated that SIL-TAL1(+) mice models had earlier disease onset, higher leukemia cell load in peripheral blood and shorter overall survival (P<0.001). Moreover, the SIL-TAL1(+) mice models exerted a tendency of TLS/DIC and seemed vulnerable towards chemotherapy, which further simulated our clinical settings. These data demonstrate that SIL-TAL1 rearrangement identifies a distinct subtype with inferior outcome which could allow for individual therapeutic stratification for T-ALL patients.
Collapse
Affiliation(s)
- Di Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| | - Guangrong Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
- Department of Hematology, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, Jiangsu, The People's Republic of China
| | - Na Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| | - Xiaoxi Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| | - Yunfan Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| | - Shiqiu Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| | - Jie Xiong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| | - Jing He
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| | - Lijun Jiang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| | - Chunrui Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| | - Danmei Xu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
- * E-mail:
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, The People's Republic of China
| |
Collapse
|
17
|
Fagioli F, Quarello P, Zecca M, Lanino E, Rognoni C, Balduzzi A, Messina C, Favre C, Foà R, Ripaldi M, Rutella S, Basso G, Prete A, Locatelli F. Hematopoietic stem cell transplantation for children with high-risk acute lymphoblastic leukemia in first complete remission: a report from the AIEOP registry. Haematologica 2013; 98:1273-81. [PMID: 23445874 DOI: 10.3324/haematol.2012.079707] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Children with high-risk acute lymphoblastic leukemia in first complete remission can benefit from allogeneic hematopoietic stem cell transplantation. We analyzed the outcome of 211 children with high-risk acute lymphoblastic leukemia in first complete remission who were given an allogeneic transplant between 1990 and 2008; the outcome of patients who, despite having an indication for transplantation and a suitable donor, did not receive the allograft for different reasons in the same time period was not analyzed. Sixty-nine patients (33%) were transplanted between 1990 and 1999, 58 (27%) between 2000 and 2005, and 84 (40%) between 2005 and 2008. A matched family donor was employed in 138 patients (65%) and an unrelated donor in 73 (35%). The 10-year probabilities of overall and disease-free survival were 63.4% and 61%, respectively. The 10-year cumulative incidences of transplantation-related mortality and relapse were 15% and 24%, respectively. After 1999, no differences in either disease-free survival or transplant-related mortality were observed in patients transplanted from unrelated or matched family donors. In multivariate analysis, grade IV acute graft-versus-host disease was an independent factor associated with worse disease-free survival. By contrast, grade I acute graft-versus-host disease and age at diagnosis between 1 and 9 years were favorable prognostic variables. Our study, not intended to evaluate whether transplantation is superior to chemotherapy for children with acute lymphoblastic leukemia in first complete remission and high-risk features, shows that the allograft cured more than 60% of these patients; in the most recent period, the outcome of recipients of grafts from matched family and unrelated donors was comparable.
Collapse
Affiliation(s)
- Franca Fagioli
- Pediatric Onco-Hematology, Stem Cell Transplantation and Cellular Therapy Division, Regina Margherita Children's Hospital, Torino.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Marshall GM, Dalla Pozza L, Sutton R, Ng A, de Groot-Kruseman HA, van der Velden VH, Venn NC, van den Berg H, de Bont ESJM, Maarten Egeler R, Hoogerbrugge PM, Kaspers GJL, Bierings MB, van der Schoot E, van Dongen J, Law T, Cross S, Mueller H, de Haas V, Haber M, Révész T, Alvaro F, Suppiah R, Norris MD, Pieters R. High-risk childhood acute lymphoblastic leukemia in first remission treated with novel intensive chemotherapy and allogeneic transplantation. Leukemia 2013; 27:1497-503. [PMID: 23407458 DOI: 10.1038/leu.2013.44] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 02/03/2013] [Accepted: 02/07/2013] [Indexed: 12/20/2022]
Abstract
Children with acute lymphoblastic leukemia (ALL) and high minimal residual disease (MRD) levels after initial chemotherapy have a poor clinical outcome. In this prospective, single arm, Phase 2 trial, 111 Dutch and Australian children aged 1-18 years with newly diagnosed, t(9;22)-negative ALL, were identified among 1041 consecutively enrolled patients as high risk (HR) based on clinical features or high MRD. The HR cohort received the AIEOP-BFM (Associazione Italiana di Ematologia ed Oncologia Pediatrica (Italy)-Berlin-Frankfurt-Münster ALL Study Group) 2000 ALL Protocol I, then three novel HR chemotherapy blocks, followed by allogeneic transplant or chemotherapy. Of the 111 HR patients, 91 began HR treatment blocks, while 79 completed the protocol. There were 3 remission failures, 12 relapses, 7 toxic deaths in remission and 10 patients who changed protocol due to toxicity or clinician/parent preference. For the 111 HR patients, 5-year event-free survival (EFS) was 66.8% (±5.5) and overall survival (OS) was 75.6% (±4.3). The 30 patients treated as HR solely on the basis of high MRD levels had a 5-year EFS of 63% (±9.4%). All patients experienced grade 3 or 4 toxicities during HR block therapy. Although cure rates were improved compared with previous studies, high treatment toxicity suggested that novel agents are needed to achieve further improvement.
Collapse
Affiliation(s)
- G M Marshall
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, UNSW, Sydney, New South Wales, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Schrappe M, Hunger SP, Pui CH, Saha V, Gaynon PS, Baruchel A, Conter V, Otten J, Ohara A, Versluys AB, Escherich G, Heyman M, Silverman LB, Horibe K, Mann G, Camitta BM, Harbott J, Riehm H, Richards S, Devidas M, Zimmermann M. Outcomes after induction failure in childhood acute lymphoblastic leukemia. N Engl J Med 2012; 366:1371-81. [PMID: 22494120 PMCID: PMC3374496 DOI: 10.1056/nejmoa1110169] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Failure of remission-induction therapy is a rare but highly adverse event in children and adolescents with acute lymphoblastic leukemia (ALL). METHODS We identified induction failure, defined by the persistence of leukemic blasts in blood, bone marrow, or any extramedullary site after 4 to 6 weeks of remission-induction therapy, in 1041 of 44,017 patients (2.4%) 0 to 18 years of age with newly diagnosed ALL who were treated by a total of 14 cooperative study groups between 1985 and 2000. We analyzed the relationships among disease characteristics, treatments administered, and outcomes in these patients. RESULTS Patients with induction failure frequently presented with high-risk features, including older age, high leukocyte count, leukemia with a T-cell phenotype, the Philadelphia chromosome, and 11q23 rearrangement. With a median follow-up period of 8.3 years (range, 1.5 to 22.1), the 10-year survival rate (±SE) was estimated at only 32±1%. An age of 10 years or older, T-cell leukemia, the presence of an 11q23 rearrangement, and 25% or more blasts in the bone marrow at the end of induction therapy were associated with a particularly poor outcome. High hyperdiploidy (a modal chromosome number >50) and an age of 1 to 5 years were associated with a favorable outcome in patients with precursor B-cell leukemia. Allogeneic stem-cell transplantation from matched, related donors was associated with improved outcomes in T-cell leukemia. Children younger than 6 years of age with precursor B-cell leukemia and no adverse genetic features had a 10-year survival rate of 72±5% when treated with chemotherapy only. CONCLUSIONS Pediatric ALL with induction failure is highly heterogeneous. Patients who have T-cell leukemia appear to have a better outcome with allogeneic stem-cell transplantation than with chemotherapy, whereas patients who have precursor B-cell leukemia without other adverse features appear to have a better outcome with chemotherapy. (Funded by Deutsche Krebshilfe and others.).
Collapse
Affiliation(s)
- Martin Schrappe
- Department of Pediatrics, University Medical Center Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Late MRD response determines relapse risk overall and in subsets of childhood T-cell ALL: results of the AIEOP-BFM-ALL 2000 study. Blood 2011; 118:2077-84. [DOI: 10.1182/blood-2011-03-338707] [Citation(s) in RCA: 296] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Abstract
The prognostic value of MRD in large series of childhood T-ALL has not yet been established. Trial AIEOP-BFM-ALL 2000 introduced standardized quantitative assessment of MRD for stratification, based on immunoglobulin and TCR gene rearrangements as polymerase chain reaction targets: Patients were considered MRD standard risk (MRD-SR) if MRD was negative at day 33 (time point 1 [TP1]) and day 78 (TP2), analyzed by at least 2 sensitive markers; MRD intermediate risk (MRD-IR) if positive either at day 33 or 78 and < 10−3 at day 78; and MRD high risk (MRD-HR) if ≥ 10−3 at day 78. A total of 464 patients with T-ALL were stratified by MRD: 16% of them were MRD-SR, 63% MRD-IR, and 21% MRD-HR. Their 7-year event-free-survival (SE) was 91.1% (3.5%), 80.6% (2.3%), and 49.8% (5.1%) (P < .001), respectively. Negativity of MRD at TP1 was the most favorable prognostic factor. An excellent outcome was also obtained in 32% of patients turning MRD negative only at TP2, indicating that early (TP1) MRD levels were irrelevant if MRD at TP2 was negative (48% of all patients). MRD ≥ 10−3 at TP2 constitutes the most important predictive factor for relapse in childhood T-ALL. The study is registered at http://www.clinicaltrials.gov; “Combination Chemotherapy Based on Risk of Relapse in Treating Young Patients With Acute Lymphoblastic Leukemia,” protocol identification #NCT00430118 for BFM and #NCT00613457 for AIEOP.
Collapse
|
21
|
Pulsipher MA, Peters C, Pui CH. High-risk pediatric acute lymphoblastic leukemia: to transplant or not to transplant? Biol Blood Marrow Transplant 2011; 17:S137-48. [PMID: 21195303 DOI: 10.1016/j.bbmt.2010.10.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Accepted: 10/06/2010] [Indexed: 11/27/2022]
Abstract
Because survival with both chemotherapy and allogeneic hematopoietic stem cell transplantation (HSCT) approaches to high-risk pediatric acute lymphoblastic leukemia (ALL) generally improves through the years, regular comparisons of outcomes with either approach for a given indication are needed to decide when HSCT is indicated. Improvements in risk classification are allowing clinicians to identify patients at high risk for relapse early in their course of therapy. Whether patients defined as high risk by new methods will benefit from HSCT requires careful testing. Standardization and improvement of transplant approaches has led to equivalent survival outcomes with matched sibling and well-matched unrelated donors; however, survival using mismatched and haploidentical donors is generally worse. Trials comparing chemotherapy and HSCT must obtain sufficient data about therapy and stratify the analysis to assess the outcomes of best-chemotherapy with best-HSCT approaches.
Collapse
Affiliation(s)
- Michael A Pulsipher
- Primary Children's Medical Center, Division of Hematology/Blood and Marrow Transplantation, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA.
| | | | | |
Collapse
|
22
|
Motta S, Decimi V, Pincelli AI, Fraschini D, Grimaldi M, Jankovic M, Masera N. Precocious puberty and empty sella syndrome in a girl cured of acute lymphoblastic leukemia. J Pediatr Endocrinol Metab 2011; 24:1067-9. [PMID: 22308868 DOI: 10.1515/jpem.2011.381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We describe a case of precocious puberty in a girl treated with chemoradiotherapy according to the Italian Association of Pediatric Hematology and Oncology ALL 9503 protocol for acute lymphoblastic leukemia (ALL) from the age of 15 months until the age of 3 years and 4 months. The patient was treated with chemotherapy and cranial irradiation (18 Gy in 12 fractions). At 7 years of age, during topical estrogenic treatment for congenital adhesions of the labia minora, she showed bilateral breast development that evolved into precocious puberty. A magnetic resonance imaging of the brain showed an "empty sella" (ES); the etiology of the ES, and the consequent precocious puberty, being presumably iatrogenic. Children treated with cranial radiotherapy should be carefully checked for signs of precocious puberty and the exogenous administration of estrogens should be avoided, as far as possible, because these could act as a trigger factor in a population at higher risk of precocious puberty.
Collapse
Affiliation(s)
- Serena Motta
- Department of Pediatrics, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | | | | | | | | | | | | |
Collapse
|
23
|
Pession A, Masetti R, Kleinschmidt K, Martoni A. Use of clofarabine for acute childhood leukemia. Biologics 2010; 4:111-8. [PMID: 20631817 PMCID: PMC2898101 DOI: 10.2147/btt.s10123] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Indexed: 12/16/2022]
Abstract
A second-generation of purine nucleoside analogs, starting with clofarabine, has been developed in the course of the search for new therapeutic agents for acute childhood leukemia, especially for refractory or relapsed disease. Clofarabine is a hybrid of fludarabine and cladribine, and has shown to have antileukemic activity in acute lymphoblastic leukemia as well as in myeloid disorders. As the only new antileukemic chemotherapeutic agent to enter clinical use in the last 10 years, clofarabine was approved as an orphan drug with the primary indication of use in pediatric patients. Toxicity has been tolerable in a heavily pretreated patient population, and clofarabine has been demonstrated to be safe, both as a single agent and in combination therapies. Liver dysfunction has been the most frequently observed adverse event, but this is generally reversible. Numerous Phase I and II trials have recently been conducted, and are still ongoing in an effort to find the optimal role for clofarabine in various treatment strategies. Concomitant use of clofarabine, cytarabine, and etoposide was confirmed to be safe and effective in two independent trials. Based on the promising results when used as a salvage regimen, clofarabine is now being investigated for its potential to become part of frontline protocols.
Collapse
Affiliation(s)
- A Pession
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, University of Bologna, Italy
| | - R Masetti
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, University of Bologna, Italy
| | - K Kleinschmidt
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, University of Bologna, Italy
| | - A Martoni
- Pediatric Oncology and Hematology “Lalla Seràgnoli”, University of Bologna, Italy
| |
Collapse
|
24
|
Molecular response to treatment redefines all prognostic factors in children and adolescents with B-cell precursor acute lymphoblastic leukemia: results in 3184 patients of the AIEOP-BFM ALL 2000 study. Blood 2010; 115:3206-14. [PMID: 20154213 DOI: 10.1182/blood-2009-10-248146] [Citation(s) in RCA: 572] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The Associazione Italiana di Ematologia Oncologia Pediatrica and the Berlin-Frankfurt-Münster Acute Lymphoblastic Leukemia (AIEOP-BFM ALL 2000) study has for the first time introduced standardized quantitative assessment of minimal residual disease (MRD) based on immunoglobulin and T-cell receptor gene rearrangements as polymerase chain reaction targets (PCR-MRD), at 2 time points (TPs), to stratify patients in a large prospective study. Patients with precursor B (pB) ALL (n = 3184) were considered MRD standard risk (MRD-SR) if MRD was already negative at day 33 (analyzed by 2 markers, with a sensitivity of at least 10(-4)); MRD high risk (MRD-HR) if 10(-3) or more at day 78 and MRD intermediate risk (MRD-IR): others. MRD-SR patients were 42% (1348): 5-year event-free survival (EFS, standard error) is 92.3% (0.9). Fifty-two percent (1647) were MRD-IR: EFS 77.6% (1.3). Six percent of patients (189) were MRD-HR: EFS 50.1% (4.1; P < .001). PCR-MRD discriminated prognosis even on top of white blood cell count, age, early response to prednisone, and genotype. MRD response detected by sensitive quantitative PCR at 2 predefined TPs is highly predictive for relapse in childhood pB-ALL. The study is registered at http://clinicaltrials.gov: NCT00430118 for BFM and NCT00613457 for AIEOP.
Collapse
|
25
|
Suzuki N, Yumura-Yagi K, Yoshida M, Hara J, Nishimura S, Kudoh T, Tawa A, Usami I, Tanizawa A, Hori H, Ito Y, Miyaji R, Oda M, Kato K, Hamamoto K, Osugi Y, Hashii Y, Nakahata T, Horibe K. Outcome of childhood acute lymphoblastic leukemia with induction failure treated by the Japan Association of Childhood Leukemia study (JACLS) ALL F-protocol. Pediatr Blood Cancer 2010; 54:71-8. [PMID: 19813250 DOI: 10.1002/pbc.22217] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Children with acute lymphoblastic leukemia (ALL) who fail to achieve complete remission (CR) after induction therapy (induction failure: IF) have a poor prognosis; however, there have been few prospective studies in patients with IF. PATIENTS AND METHODS Between April 1997 and March 2005, 27 of 1,237 leukemic patients (2.2%) failed to achieve CR after four- or five-drug induction therapy. Twenty-three of these patients entered the F-protocol study, which mainly consisted of acute-myeloid-leukemia-oriented chemotherapy followed by scheduled hematopoietic cell transplantation (HCT). RESULTS Seventeen (73.9%) of the 23 patients responded to re-induction chemotherapy with CR. Of note, 15 (93.8%) of 16 patients with Philadelphia-chromosome-negative (non-Ph(+)) ALL achieved CR; in contrast, only 2 (28.6%) of 7 Ph(+) patients achieved CR. Fourteen (82.4%) of 17 patients remained in CR (CCR) until their scheduled HCT, 12 of the 14 with CCR underwent HCT as scheduled, and 6 patients remain in first CR after a median of 78 months (range, 49-107 months). The 5-year overall survival (OS) rates of 16 patients with non-Ph(+) and 7 patients with Ph(+) were 43.8 +/- 12.4% and 14.3 +/- 13.2%, respectively (P = 0.012). The 5-year OS rate of the 17 patients who obtained CR by re-induction therapy and the 6 who did not were 47.1 +/- 12.1% and 0%, respectively (P < 0.001). CONCLUSION Acute-myeloid-leukemia-oriented chemotherapy followed by scheduled HCT is a promising treatment strategy for non-Ph(+) ALL patients with IF.
Collapse
Affiliation(s)
- Nobuhiro Suzuki
- Department of Pediatrics, Sapporo Medical University Hospital, Sapporo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Conter V, Aricò M, Basso G, Biondi A, Barisone E, Messina C, Parasole R, De Rossi G, Locatelli F, Pession A, Santoro N, Micalizzi C, Citterio M, Rizzari C, Silvestri D, Rondelli R, Lo Nigro L, Ziino O, Testi AM, Masera G, Valsecchi MG. Long-term results of the Italian Association of Pediatric Hematology and Oncology (AIEOP) Studies 82, 87, 88, 91 and 95 for childhood acute lymphoblastic leukemia. Leukemia 2009; 24:255-64. [PMID: 20016536 DOI: 10.1038/leu.2009.250] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We analyzed the long-term outcome of 4865 patients treated in Studies 82, 87, 88, 91 and 95 for childhood acute lymphoblastic leukemia (ALL) of the Italian Association of Pediatric Hematology and Oncology (AIEOP). Treatment was characterized by progressive intensification of systemic therapy and reduction of cranial radiotherapy. A progressive improvement of results with reduction of isolated central nervous system relapse rate was obtained. Ten-year event-free survival increased from 53% in Study 82 to 72% in Study 95, whereas survival improved from 64 to 82%. Since 1991, all patients were treated according to Berlin-Frankfurt-Muenster (BFM) ALL treatment strategy. In Study 91, reduced treatment intensity (25%) yielded inferior results, but intensification of maintenance with high-dose (HD)-L-asparaginase (randomized) allowed to compensate for this disadvantage; in high-risk patients (HR, 15%), substitution of intensive polychemotherapy blocks for conventional BFM backbone failed to improve results. A marked improvement of results was obtained in HR patients when conventional BFM therapy was intensified with three polychemotherapy blocks and double delayed intensification (Study 95). The introduction of minimal residual disease monitoring and evaluation of common randomized questions by AIEOP and BFM groups in the protocol AIEOP-BFM-ALL 2000 are expected to further ameliorate treatment of children with ALL.
Collapse
Affiliation(s)
- V Conter
- Department of Pediatrics, University of Milano-Bicocca, Ospedale S. Gerardo, Monza, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Long-term results of five consecutive trials in childhood acute lymphoblastic leukemia performed by the ALL-BFM study group from 1981 to 2000. Leukemia 2009; 24:265-84. [PMID: 20010625 DOI: 10.1038/leu.2009.257] [Citation(s) in RCA: 360] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Between 1981 and 2000, 6609 children (<18 years of age) were treated in five consecutive trials of the Berlin-Frankfurt-Münster (BFM) study group for childhood acute lymphoblastic leukemia (ALL). Patients were treated in up to 82 centers in Germany, Austria and Switzerland. Probability of 10-year event-free survival (EFS) (survival) improved from 65% (77%) in study ALL-BFM 81 to 78% (85%) in ALL-BFM 95. In parallel to relapse reduction, major efforts focused on reducing acute and late toxicity through advanced risk adaptation of treatment. The major findings derived from these ALL-BFM trials were as follows: (1) preventive cranial radiotherapy could be safely reduced to 12 Gy in T-ALL and high-risk (HR) ALL patients, and eliminated in non- HR non-T-ALL patients, if it was replaced by high-dose and intrathecal (IT) MTX; (2) omission of delayed re-intensification severely impaired outcome of low-risk patients; (3) 6-month-less maintenance therapy caused an increase in systemic relapses; (4) slow response to an initial 7-day prednisone window was identified as adverse prognostic factor; (5) condensed induction therapy resulted in significant improvement of outcome; (6) the daunorubicin dose in induction could be safely reduced in low-risk patients and (7) intensification of consolidation/re-intensification treatment led to considerable improvement of outcome in HR patients.
Collapse
|
28
|
Stark B, Avrahami G, Nirel R, Abramov A, Attias D, Ballin A, Bielorai B, Burstein Y, Gavriel H, Elhasid R, Kapelushnik J, Sthoeger D, Toren A, Wientraub M, Yaniv I, Izraeli S. Extended triple intrathecal therapy in children with T-cell acute lymphoblastic leukaemia: a report from the Israeli National ALL-Studies. Br J Haematol 2009; 147:113-24. [DOI: 10.1111/j.1365-2141.2009.07853.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
29
|
Luo XQ, Ke ZY, Huang LB, Guan XQ, Zhang YC, Zhang XL. High-risk childhood acute lymphoblastic leukemia in China: factors influencing the treatment and outcome. Pediatr Blood Cancer 2009; 52:191-5. [PMID: 18989886 DOI: 10.1002/pbc.21810] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Acute lymphoblastic leukemia (ALL) with high-risk features has an inferior outcome. Factors influencing the treatment and outcome of pediatric ALL with high-risk features in developing countries have not been well studied. METHODS High-risk features were defined as: age <1 year or >10 years, white blood cell (WBC) > 50 x 10(9)/L, CNS or testicular involvement at diagnosis, T-ALL, BCR-ABL/MLL-AF4, poor prednisone response, slow early response to induction chemotherapy which was defined as M3 status (>25% blasts) on day 15 bone marrow with age >6 years or presenting WBC > 20 x 10(9)/L at diagnosis and/or non-remission (NR) after 33 days of induction therapy. RESULTS Ninety-one children were analyzed. The total rate of treatment abandonment was 24.2% and treatment-related mortality was 3.3% (3/91). The event-free survival (EFS) was 52.3% (95% CI, 41.5-63.1%) at 4 years and 49.9% (95% CI, 38.9-60.9%) at 8 years, respectively. When the cases who abandoned treatment were excluded, the EFS of the remainder was 68.3% (95% CI, 56.5-80.1%) at 4 years and 65.2% (95% CI, 52.5-77.9%) at 8 years, respectively. NR on day 33 or BCR-ABL remained as an independent unfavorable prognostic factor in the Cox model even if more intense chemotherapy was administrated. CONCLUSION A decreased treatment-related death frequency was associated with an improved outcome of leukemia. This emphasizes the importance of improving supportive care in developing countries for children with high-risk ALL who receive very intensive chemotherapy. Treatment abandonment remains a prominent reason for treatment failure in China.
Collapse
Affiliation(s)
- Xue-Qun Luo
- Department of Pediatrics, The First Affiliated Hospital of Sun Yat-Sen University, Zhongshan Er Lu, Guangzhou, China.
| | | | | | | | | | | |
Collapse
|
30
|
Buendia MTA, Lozano JM, Suarez GE, Saavedra C, Guevara G. The impact of acute lymphoblastic leukemia treatment on central nervous system results in Bogota, Colombia. J Pediatr Hematol Oncol 2008; 30:643-50. [PMID: 18776755 DOI: 10.1097/mph.0b013e31817e4a7d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To improve the outcome of children with acute lymphoblastic leukemia (ALL) treated at the National Cancer Institute, Bogota, Colombia, a protocol based on the BFM-90 (Berlin, Frankfurt, Munster study) and the LSA2L2 regimens was implemented in the year 1993. The patients were classified as being standard risk (SR) or high risk (HR) according to clinical criteria, to which cytogenetic information and day-8 prednisone response were also added. A 123-patient cohort entered the study, 18 of them being considered SR and 105 HR. There was a 94% 10 years' event-free-survival rate for the SR group and 36% for the HR group. Decreased induction death rate (7% vs. 14%), increased complete remission (CR) rate (81% vs. 75%), and continuous CR (45% vs. 33%) were found in comparison with the previous study. A significant improvement was achieved in relapse rate, 44% to 28% (P=0.029), mainly due to reduced central nervous system relapse rate from 16% to 6% (P=0.037), whereas the number of patients receiving cranial radiation was reduced to 55%. A major problem concerned the increased CR mortality rate, 5% to 14% (P=0.06). Improved supportive care therapy and socioeconomic conditions will hopefully reduce the CR mortality rate in the future.
Collapse
|
31
|
Oudot C, Auclerc MF, Levy V, Porcher R, Piguet C, Perel Y, Gandemer V, Debre M, Vermylen C, Pautard B, Berger C, Schmitt C, Leblanc T, Cayuela JM, Socie G, Michel G, Leverger G, Baruchel A. Prognostic factors for leukemic induction failure in children with acute lymphoblastic leukemia and outcome after salvage therapy: the FRALLE 93 study. J Clin Oncol 2008; 26:1496-503. [PMID: 18349402 DOI: 10.1200/jco.2007.12.2820] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To identify prognostic factors and to evaluate the outcome of children with acute lymphoblastic leukemia (ALL) failure after induction therapy. PATIENTS AND METHODS Between June 1993 and December 1999, 1,395 leukemic children were included in the French Acute Lymphoblastic Leukemia 93 study. RESULTS Fifty-three patients (3.8%) had a leukemic induction failure (LIF) after three- or four-drug induction therapy. In univariate analysis, high WBC count (P = .001), mediastinal mass (P = .017), T-cell phenotype (T-ALL; P = .001), t(9;22) translocation (P = .001), and a slow early response (at day 8 and/or on day 21, P = .001) were predictive of LIF. The following three prognostic groups for LIF were identified by multivariate analysis: a low-risk group with B-cell progenitor (BCP) ALL without t(9;22) (odds ratio [OR] = 1), an intermediate-risk group with T-ALL and a mediastinal mass (OR = 7.4, P < .0001), and a high-risk group with BCP-ALL and t(9;22) or T-ALL without a mediastinal mass (OR = 28.4, P < .0001). Complete remission (CR) was subsequently obtained in 43 patients (81%). The 5-year overall survival (OS) rate of the 53 patients was 30% +/- 6%. The 5-year OS rate among allogeneic graft recipients, autologous graft recipients, and after chemotherapy were 30.4% +/- 9.6% (50% +/- 26% after genoidentical transplantation), 50% +/- 17.7%, and 41.7% +/- 14.2%, respectively (P = .18). Fourteen patients (26%) were still in first CR after a median of 83 months (range, 53 to 117 months). CONCLUSION Three risk categories for LIF in children with ALL were identified. Approximately one third of patients with LIF can be successfully treated with salvage therapy overall. Subsequent CR after LIF is mandatory for cure.
Collapse
Affiliation(s)
- Caroline Oudot
- Service d'Hématologie et Oncologie Pédiatrique, Hôpital Mère-Enfant, Limoges, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Aricò M, Valsecchi MG, Rizzari C, Barisone E, Biondi A, Casale F, Locatelli F, Lo Nigro L, Luciani M, Messina C, Micalizzi C, Parasole R, Pession A, Santoro N, Testi AM, Silvestri D, Basso G, Masera G, Conter V. Long-term results of the AIEOP-ALL-95 Trial for Childhood Acute Lymphoblastic Leukemia: insight on the prognostic value of DNA index in the framework of Berlin-Frankfurt-Muenster based chemotherapy. J Clin Oncol 2008; 26:283-9. [PMID: 18182669 DOI: 10.1200/jco.2007.12.3927] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Between May 1995 and August 2000 the Associazione Italiana di Ematologia Oncologia Pediatrica conducted the ALL-95 study for risk-directed, Berlin-Frankfurt-Muenster (BFM) -oriented therapy of childhood acute lymphoblastic leukemia, aimed at exploring treatment reduction in standard-risk patients (SR) and intensification during continuation therapy in intermediate-risk patients (IR) as randomized questions and treatment intensification in high-risk patients (HR). The prognostic value of DNA index was explored in this setting. PATIENTS AND METHODS A total of 1,744 patients were enrolled (115, SR; 1,385, IR; and 244, HR). SR patients (DNA index >/= 1.16 and < 1.60; age, 1 to 5 years; and WBC < 20,000, non-T-immunophenotype, with no high-risk features) received a reduced induction therapy (no anthracyclines); IR patients were randomly assigned to receive or not receive vincristine and dexamethasone pulses during maintenance; HR therapy was based on a conventional BFM schedule intensified with three chemotherapy blocks followed by a double reinduction phase. RESULTS The event-free survival and overall survival probabilities at 10 years for the entire group were 72.5% (SE, 1.3) and 83.6% (SE, 0.9); 85.0% (SE, 3.4) and 95.5% (SE, 2.0) in SR, 75.1% (SE, 1.5) and 87.5% (SE, 0.9) in IR, and 51.0% (SE, 3.2) and 57.2% (SE, 3.3) in HR patients, respectively. Patients with a favorable DNA index had superior EFS in both IR (83.8% [2.7%] v 73.9% [1.7%]) and in HR (67.8% [9.4%] and 49.6% [3.5%]). Of the six patients with DNA index less than 0.8, only one remained in remission. CONCLUSION Favorable DNA index was associated with a better prognosis in IR and HR patients defined by presenting clinical criteria and treatment with a BFM-oriented chemotherapy.
Collapse
Affiliation(s)
- Maurizio Aricò
- Pediatric Hematology Oncology, Ospedale dei Bambini G. Di Cristina, Palermo, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Risk-adjusted therapy of acute lymphoblastic leukemia can decrease treatment burden and improve survival: treatment results of 2169 unselected pediatric and adolescent patients enrolled in the trial ALL-BFM 95. Blood 2008; 111:4477-89. [PMID: 18285545 DOI: 10.1182/blood-2007-09-112920] [Citation(s) in RCA: 409] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The trial ALL-BFM 95 for treatment of childhood acute lymphoblastic leukemia was designed to reduce acute and long-term toxicity in selected patient groups with favorable prognosis and to improve outcome in poor-risk groups by treatment intensification. These aims were pursued through a stratification strategy using white blood cell count, age, immunophenotype, treatment response, and unfavorable genetic aberrations providing an excellent discrimination of risk groups. Estimated 6-year event-free survival (6y-pEFS) for all 2169 patients was 79.6% (+/- 0.9%). The large standard-risk (SR) group (35% of patients) achieved an excellent 6y-EFS of 89.5% (+/- 1.1%) despite significant reduction of anthracyclines. In the medium-risk (MR) group (53% of patients), 6y-pEFS was 79.7% (+/- 1.2%); no improvement was accomplished by the randomized use of additional intermediate-dose cytarabine after consolidation. Omission of preventive cranial irradiation in non-T-ALL MR patients was possible without significant reduction of EFS, although the incidence of central nervous system relapses increased. In the high-risk (HR) group (12% of patients), intensification of consolidation/reinduction treatment led to considerable improvement over the previous ALL-BFM trials yielding a 6y-pEFS of 49.2% (+/- 3.2%). Compared without previous trial ALL-BFM 90, consistently favorable results in non-HR patients were achieved with significant treatment reduction in the majority of these patients.
Collapse
|
34
|
Flohr T, Schrauder A, Cazzaniga G, Panzer-Grümayer R, van der Velden V, Fischer S, Stanulla M, Basso G, Niggli FK, Schäfer BW, Sutton R, Koehler R, Zimmermann M, Valsecchi MG, Gadner H, Masera G, Schrappe M, van Dongen JJM, Biondi A, Bartram CR. Minimal residual disease-directed risk stratification using real-time quantitative PCR analysis of immunoglobulin and T-cell receptor gene rearrangements in the international multicenter trial AIEOP-BFM ALL 2000 for childhood acute lymphoblastic leukemia. Leukemia 2008; 22:771-82. [PMID: 18239620 DOI: 10.1038/leu.2008.5] [Citation(s) in RCA: 291] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Detection of minimal residual disease (MRD) is the most sensitive method to evaluate treatment response and one of the strongest predictors of outcome in childhood acute lymphoblastic leukemia (ALL). The 10-year update on the I-BFM-SG MRD study 91 demonstrates stable results (event-free survival), that is, standard risk group (MRD-SR) 93%, intermediate risk group (MRD-IR) 74%, and high risk group (MRD-HR) 16%. In multicenter trial AIEOP-BFM ALL 2000, patients were stratified by MRD detection using quantitative PCR after induction (TP1) and consolidation treatment (TP2). From 1 July 2000 to 31 October 2004, PCR target identification was performed in 3341 patients: 2365 (71%) patients had two or more sensitive targets (< or =10(-4)), 671 (20%) patients revealed only one sensitive target, 217 (6%) patients had targets with lower sensitivity, and 88 (3%) patients had no targets. MRD-based risk group assignment was feasible in 2594 (78%) patients: 40% were classified as MRD-SR (two sensitive targets, MRD negativity at both time points), 8% as MRD-HR (MRD > or =10(-3) at TP2), and 52% as MRD-IR. The remaining 823 patients were stratified according to clinical risk features: HR (n=108) and IR (n=715). In conclusion, MRD-PCR-based stratification using stringent criteria is feasible in almost 80% of patients in an international multicenter trial.
Collapse
Affiliation(s)
- T Flohr
- Department of Pediatrics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
AbstractSurvival of children with acute lymphoblastic leukemia (ALL) is often described as the success story for oncology. The improvements in the treatment of ALL represent the work of cooperative groups at their best. Fifty years ago a pediatric oncologist would have never considered using the term “cure” in a discussion with a family whose child was diagnosed with ALL. Today the term is not only used in the initial discussion but referred to frequently thereafter. However, as we all know, cure is not assured and is not obtained without sequelae. This review will focus on the improvements in treatment for newly diagnosed ALL in children and adolescents according to risk group and some of the challenges that remain despite the improved outcome.
Collapse
|
36
|
Chowdhury T, Brady HJM. Insights from clinical studies into the role of the MLL gene in infant and childhood leukemia. Blood Cells Mol Dis 2007; 40:192-9. [PMID: 17905612 DOI: 10.1016/j.bcmd.2007.07.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Accepted: 07/19/2007] [Indexed: 11/16/2022]
Abstract
Translocations involving the Mixed Lineage Leukemia (MLL) gene at 11q23 are found in both acute lymphoblastic leukemia (ALL) and acute myeloblastic leukemia (AML), but have different prognostic implications depending on the phenotype of the leukemia in de novo pediatric cases. The majority of MLL gene rearrangements are associated with infant ALL, and their presence predicts a poor prognosis which worsens with earlier age of presentation. Rearrangements of the MLL gene are found in most cases of infant AML and regardless of age confer an intermediate risk. The treatment of MLL-rearranged ALL in children involves increased intensification of chemotherapy, and infants with ALL are treated with an intensive regimen of ALL- and AML-like chemotherapy, with the proportion of MLL-rearranged cases being responsible for the poor outcome in this age group. The use of DNA microarray analysis to distinguish a particular gene signature for MLL-rearranged leukemias is shedding light on the molecular mechanisms and potential therapeutic targets of these leukemias. It may also prove to have a useful role in both diagnosis and prognosis. This review considers recent advances in our understanding of the role of MLL gene rearrangements in pediatric clinical practice.
Collapse
Affiliation(s)
- Tanzina Chowdhury
- Molecular Haematology and Cancer Biology Unit, Institute of Child Health, University College London, London, UK
| | | |
Collapse
|
37
|
Bhojwani D, Moskowitz N, Raetz EA, Carroll WL. Potential of gene expression profiling in the management of childhood acute lymphoblastic leukemia. Paediatr Drugs 2007; 9:149-56. [PMID: 17523695 DOI: 10.2165/00148581-200709030-00003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Childhood acute lymphoblastic leukemia (ALL) is a heterogeneous disease. Current treatment approaches are tailored according to the clinical features of the host, genotypic features of the leukemic blast, and early response to therapy. Although these approaches have been successful in dramatically improving outcomes, approximately 20% of children with ALL still relapse and many of these children do not have an identifiable adverse risk factor at presentation. Further insights into the biologic basis of the disease may contribute to novel, rational treatment strategies. Childhood ALL has served as an example for demonstrating the feasibility and potential of high-throughput technologies such as global gene expression or transcript profiling. In the last decade or so, utilization of these techniques has grown exponentially. As the methodology undergoes refinement and validation, it is plausible that microarrays may be used in the routine management of childhood ALL in the next few years. This article discusses the numerous applications to date of gene expression profiling in childhood ALL. Multiple investigators have made it evident that microarrays can be used as a single platform for the accurate classification of ALL into the various cytogenetic subtypes. Additional promising utilities include prediction of early response to therapy, overall outcome, and adverse effects. Identification of patients who are predicted to have an unfavorable outcome may allow for early intervention such as intensification of therapy or avoidance of drugs that are associated with specific secondary effects such as therapy-related acute myelogenous leukemia. Knowledge has been gained into pathways contributing to leukemogenesis and chemoresistance. Therapeutic targets have been identified, some of which are entering clinical trials following validation in additional preclinical models. These newer methods of genome analyses complemented by studies involving the proteome as well as host polymorphisms will have a profound impact on the diagnosis and management of childhood ALL.
Collapse
Affiliation(s)
- Deepa Bhojwani
- NYU Cancer Institute, Division of Pediatric Hematology, New York University School of Medicine, New York, New York 10016, USA.
| | | | | | | |
Collapse
|
38
|
Balduzzi A, Conter V, Uderzo C, Valsecchi MG. Transplantation in childhood very high risk acute lymphoblastic leukemia in first complete remission: where are we now? J Clin Oncol 2007; 25:2625-6; author reply 2627-8. [PMID: 17577046 DOI: 10.1200/jco.2007.11.5014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
39
|
Ribera JM, Ortega JJ, Oriol A, Bastida P, Calvo C, Pérez-Hurtado JM, González-Valentín ME, Martín-Reina V, Molinés A, Ortega-Rivas F, Moreno MJ, Rivas C, Egurbide I, Heras I, Poderós C, Martínez-Revuelta E, Guinea JM, del Potro E, Deben G. Comparison of intensive chemotherapy, allogeneic, or autologous stem-cell transplantation as postremission treatment for children with very high risk acute lymphoblastic leukemia: PETHEMA ALL-93 Trial. J Clin Oncol 2007; 25:16-24. [PMID: 17194902 DOI: 10.1200/jco.2006.06.8312] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The optimal postremission therapy for children with very high-risk (VHR) acute lymphoblastic leukemia (ALL) is not well established. This randomized trial compared three options of postremission therapy: chemotherapy and allogeneic or autologous stem-cell transplantation (SCT). PATIENTS AND METHODS All 106 VHR-ALL patients received induction with five drugs followed by intensification with three cycles of chemotherapy. Patients in complete remission (CR) with an HLA-identical family donor were assigned to allogeneic SCT (n = 24) and the remaining were randomly assigned to autologous SCT (n = 38) or to delayed intensification followed by maintenance chemotherapy up to 2 years in CR (n = 38). RESULTS Overall, 100 patients achieved CR (94%). With a median follow-up of 6.5 years, 5-year disease-free survival (DFS) and overall survival (OS) probabilities were 45% (95% CI, 37% to 54%) and 48% (95% CI, 40% to 57%), respectively. The three groups were comparable in the main pretreatment ALL characteristics. Intention-to-treat analysis showed no differences for donor versus no donor in DFS (45%; 95% CI, 27% to 65% v 45%; 95% CI, 37% to 55%) and OS (48%; 95% CI, 30% to 67% v 51%; 95% CI, 43% to 61%), as well as for autologous SCT versus chemotherapy comparisons (DFS: 44%; 95% CI, 29% to 60% v 46%; 95% CI, 32% to 62%; OS: 45%; 95% CI, 31% to 62% v 57%; 95% CI, 43% to 73%). No differences were found within the different subgroups of ALL and neither were differences observed when the analysis was made by treatment actually performed. CONCLUSION This study failed to prove that, when a family donor is available, allogeneic SCT produces a better outcome than autologous SCT or chemotherapy in children with VHR-ALL.
Collapse
Affiliation(s)
- Jose-Maria Ribera
- Servicio de Hematología Clínica, Institut Català d'Oncologia-Hospital Universitari Germans Trias i Pujol, Badalona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Luciani M, Rana I, Pansini V, Caniglia M, Coletti V, Maraschini A, Lombardi A, De Rossi G. Infant leukaemia: clinical, biological and therapeutic advances. Acta Paediatr 2006; 95:47-51. [PMID: 16801167 DOI: 10.1080/08035320600649580] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
UNLABELLED Infant acute lymphoid leukaemia (IALL) represents a distinct subset with an extremely poor response to therapy, despite major progress in the treatment of childhood leukaemia. However, several studies have shown that, even in this generally considered homogeneous group, a distinction could be made with regard to prognosis. The outcome of IALL patients with ALL-1/MLL rearrangements at the 11q23 cytogenetic band, early pre-B immunophenotype, high WBC count and age below 6 mo is significantly worse than in patients without these characteristics, and current therapies appear inadequate in a significant number of cases. Therefore, an international protocol (Interfant 99) was recently started, using a more aggressive approach, which included lymphoid- and myeloid-specific drugs, and indications for stem-cell transplantation. We reviewed the clinical characteristics of the disease, the results of several recent international clinical trials, and our experience with 16 infants with acute lymphoid leukaemia diagnosed and treated at our institution. CONCLUSION It is extremely important to stratify patients for prognosis, taking into account clinical and biological variables with independent prognostic value. The aim is to select more adequate, risk-adapted, therapeutic strategies which also consider related or unrelated bone marrow transplant consolidation for patients with very poor prognosis.
Collapse
Affiliation(s)
- Matteo Luciani
- Haematology Division, Ospedale Pediatrico Bambino Gesù, Research Institute, IRCCS, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Svoboda J, Kotloff R, Tsai DE. Management of patients with post-transplant lymphoproliferative disorder: the role of rituximab. Transpl Int 2006; 19:259-69. [PMID: 16573540 DOI: 10.1111/j.1432-2277.2006.00284.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Post-transplant lymphoproliferative disorder (PTLD) is a serious complication of solid organ and bone marrow transplantations. Rituximab (Rituxan, Mabthera), a chimeric monoclonal antibody to the CD20 antigen on the surface of B-cell lymphocytes, has been used increasingly in the treatment of PTLD. Rituximab was initially approved for the treatment of low-grade non-Hodgkin lymphomas, but multiple case studies, retrospective analyses, and phase II trials demonstrate the benefit of rituximab in PTLD. This paper reviews the current data on rituximab and its promising role in the management of PTLD.
Collapse
Affiliation(s)
- Jakub Svoboda
- University of Pennsylvania Cancer Center, Bone Marrow and Stem Cell Transplant Program, Philadelphia, 19104, USA
| | | | | |
Collapse
|
42
|
Ziino O, D'Urbano LE, De Benedetti F, Conter V, Barisone E, De Rossi G, Basso G, Aricò M. The MIF-173G/C polymorphism does not contribute to prednisone poor response in vivo in childhood acute lymphoblastic leukemia. Leukemia 2006; 19:2346-7. [PMID: 16208413 DOI: 10.1038/sj.leu.2403973] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
43
|
Tzortzatou-Stathopoulou F, Moschovi MA, Papadopoulou AL, Barbounaki IG, Lambrou GI, Balafouta M, Syriopoulou V. Could intensified treatment in childhood acute lymphoblastic leukemia improve outcome independently of risk factors? Eur J Haematol 2005; 75:361-9. [PMID: 16191084 DOI: 10.1111/j.1600-0609.2005.00527.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE Many risk-directed therapeutic protocols have been proposed in acute lymphoblastic leukemia (ALL). However, the relapse rates remain high. The effectiveness of each protocol depends on how quickly the clearance of blast cells is achieved. In an attempt to improve survival, by minimizing treatment toxicity and relapse rate, different therapeutic protocols were used every 3 yr in our Unit. PATIENTS AND METHODS During 1991-2000, 132 children with ALL were diagnosed in our Unit. Modified and intensified NY II and BFM protocols, in three consecutive periods [(Hematology/Oncology Pediatric Department of the University of Athens) HOPDA-91, HOPDA-94, HOPDA-97] were used. RESULTS At a median follow-up time of 96 months, the 8-year overall survival (OS) was 88% +/- 3%, whereas the event-free survival (EFS) was 85% +/- 3%. There was a significant increase of the 5-year EFS of the high-risk (HR) group through time (65% in HOPDA-91 vs. 80% in HOPDA-97), whereas EFS of the low risk (LR) group in HOPDA-97 was 96%. Five cases relapsed (3.8%), four of which underwent successful bone marrow transplantation. Fifteen children died (13 diagnosed by 1996, two in the last 4 yr). CONCLUSION Modification of the protocols significantly improved survival in both HR and LR groups. The intensified regimen in the LR group did not increase the adverse toxic events, but on the contrary was extremely effective.
Collapse
Affiliation(s)
- Fotini Tzortzatou-Stathopoulou
- Hematology/Oncology Unit, First Department of Pediatrics, University of Athens, 'Aghia Sophia' Children's Hospital, Athens, Greece.
| | | | | | | | | | | | | |
Collapse
|
44
|
Roy A, Bradburn M, Moorman AV, Burrett J, Love S, Kinsey SE, Mitchell C, Vora A, Eden T, Lilleyman JS, Hann I, Saha V. Early response to induction is predictive of survival in childhood Philadelphia chromosome positive acute lymphoblastic leukaemia: results of the Medical Research Council ALL 97 trial. Br J Haematol 2005; 129:35-44. [PMID: 15801953 DOI: 10.1111/j.1365-2141.2005.05425.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We report on the outcome of children with Philadelphia positive acute lymphoblastic leukaemia (Ph+ ALL) treated on the UK Medical Research Council (MRC) trial for childhood ALL, MRC ALL 97, between January 1997 and June 2002. Forty-two (2.3%) patients were Ph+. Nineteen (45%) had <25% blasts in bone marrow (BM) within the first 2 weeks of treatment and were defined as a good response group (GRG), the others as a poor response group (PRG). Thirty-six (86%) achieved first complete remission (CR1) at the end of induction, of which 28 underwent BM transplantation (BMT). The median follow-up was 42 months (range, 21-84). The 3-year event-free survival (EFS; 52%, 95% CI, 36-66%) was a considerable improvement on the previous MRC UKALL XI trial (27%). EFS for the GRG and PRG were 68% (43-84%) and 39% (18-59%), respectively (P = 0.03); presenting white cell count <50 x 10(9)/l (P = 0.02) was predictive for overall survival. Changes in the MRC ALL97 trial within the study period resulted in some Ph+ ALL receiving daunorubicin and either prednisolone or dexamethasone during induction. Though the use of daunorubicin during induction was not a prospective study question, EFS was significantly better for those whose induction included this drug (P = 0.02). Steroid randomization was not stratified for Ph+ ALL patients and was not predictive for EFS. BMT in CR1 appeared to reduce the risk of a subsequent BM relapse. These results show significant improvement on previous MRC trials; future therapeutic strategies should include early intensive therapy and BMT in CR1.
Collapse
Affiliation(s)
- Anindita Roy
- Cancer Research UK Children's Cancer Group, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Balduzzi A, Valsecchi MG, Uderzo C, De Lorenzo P, Klingebiel T, Peters C, Stary J, Felice MS, Magyarosy E, Conter V, Reiter A, Messina C, Gadner H, Schrappe M. Chemotherapy versus allogeneic transplantation for very-high-risk childhood acute lymphoblastic leukaemia in first complete remission: comparison by genetic randomisation in an international prospective study. Lancet 2005; 366:635-42. [PMID: 16112299 DOI: 10.1016/s0140-6736(05)66998-x] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND The dismal prognosis of very-high-risk childhood acute lymphoblastic leukaemia could be improved by allogeneic haemopoietic cell transplantation. We compared this strategy with intensified chemotherapy protocols, with the aim to improve the outcome of children with very-high-risk acute lymphoblastic leukaemia in first complete remission. METHODS A cooperative prospective study was set up in seven countries. Very-high-risk acute lymphoblastic leukaemia in first complete remission was defined by the presence of at least one of the following criteria: (1) failure to achieve complete remission after the first four-drug induction phase; (2) t(9;22) or t(4;11) clonal abnormalities; and (3) poor response to prednisone associated with T immunophenotype, white-blood-cell count of 100x10(9)/L or greater, or both. Children were allocated treatment by genetic chance, according to the availability of a compatible related donor, and assigned chemotherapy or haemopoietic-cell transplantation. The primary outcome was disease-free survival and analysis was by intention to treat. FINDINGS Between April, 1995, and December, 2000, 357 children entered the study, of whom 280 were assigned chemotherapy and 77 related-donor haemopoietic-cell transplantation. 5-year disease-free survival was 40.6% (SE 3.1) in children allocated chemotherapy and 56.7% (5.7) in those assigned transplantation (hazard ratio 0.67 [95% CI 0.46-0.99]; p=0.02); 5-year survival was 50.1% (3.1) and 56.4% (5.9), respectively (0.73 [0.49-1.09]; p=0.12). INTERPRETATION Children with very-high-risk acute lymphoblastic leukaemia benefit from related-donor haemopoietic-cell transplantation compared with chemotherapy. The gap between the two strategies increases as the risk profile of the patient worsens.
Collapse
Affiliation(s)
- Adriana Balduzzi
- Clinica Pediatrica dell'Università degli Studi di Milano Bicocca, Ospedale San Gerardo Via Pergolesi 33, 20052 Monza, Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mayer J, Krejcí M, Doubek M, Pospísil Z, Brychtová Y, Tomíska M, Rácil Z. Pulse cyclophosphamide for corticosteroid-refractory graft-versus-host disease. Bone Marrow Transplant 2005; 35:699-705. [PMID: 15696180 DOI: 10.1038/sj.bmt.1704829] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Corticosteroid-resistant GVHD is difficult to manage and is associated with high morbidity and mortality. Cyclophosphamide (Cy) is an established immunosuppressive and cytotoxic drug widely used as part of pretransplant conditioning regimens. In a retrospective study of 15 patients who had not responded to corticosteroids (nine with acute GVHD, three with GVHD after donor leukocyte infusion, and three progressive chronic GVHD), pulse Cy at a median dose of 1 g/m(2) was very effective in the treatment of skin (100% response), liver (70% response), and the oral cavity (100% response). Severe intestinal GVHD responded poorly. The toxicity profile was acceptable, with manageable, short-term myelosuppression in some patients. The risk of opportunistic infections, mixed chimerism, relapses, or post-transplant lymphoproliferative disease was not increased. Overall survival was 57%, with median and maximum follow-up of 9 and 37 months, respectively. The cost of the drug was negligible, especially when compared to monoclonal antibodies. Pulse Cy requires further investigation in corticosteroid-resistant GVHD.
Collapse
Affiliation(s)
- J Mayer
- Department of Internal Medicine--Hemato-oncology, University Hospital Brno, Brno, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
On current chemotherapeutic regimens, children with Philadelphia positive acute lymphoblastic leukaemia show a heterogeneous response to treatment. A few respond quickly to treatment and achieve long-term remission. Some fail to achieve remission after induction and the majority respond slowly to treatment. Relapse on treatment is common and remission is sustained in a proportion of cases only after allogeneic stem cell transplantation (allo-SCT). The use of imatinib along with conventional cytoreductive therapy, prior to allo-SCT appears to be the most promising strategy. The future lies in the molecular evaluation of response to treatment and combination targeted chemotherapy.
Collapse
Affiliation(s)
- Louise K Jones
- Cancer Research UK Children's Cancer Group, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW Graft-versus-host disease is one of the commonest complications of allogeneic bone marrow or peripheral blood stem cell transplantation. This review will cover advances in the pathophysiology of graft-versus-host disease and new agents under investigation for the treatment of this disorder. Patients developing graft-versus-host disease who fail to respond to steroids have a poor prognosis. In this group of people, morbidity and mortality are very high. RECENT FINDINGS Novel agents are currently under investigation for the treatment of such devastating disorders. Pentostatin, denileukin diftitox, mycophenolate mofetil, extracorporeal photopheresis, and several monoclonal antibodies have been used, some of them with encouraging results. SUMMARY As supportive care improves and new agents are added to the armamentarium against steroid-refractory acute graft-versus-host disease, the prognosis of this entity may start to change. Patients with this complication after transplantation should be enrolled, whenever possible, in clinical trials to find effective therapies.
Collapse
Affiliation(s)
- Javier Bolaños-Meade
- Department of Medicine, University of Maryland Greenebaum Cancer Center and University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
49
|
Morimoto A, Kuriyama K, Hibi S, Todo S, Yoshihara T, Kuroda H, Imashuku S. Prognostic Value of Early Response to Treatment Combined with Conventional Risk Factors in Pediatric Acute Lymphoblastic Leukemia. Int J Hematol 2005; 81:228-34. [PMID: 15902780 DOI: 10.1532/ijh97.04114] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To determine useful prognostic factors in treating childhood acute lymphoblastic leukemia (ALL), we correlated conventional risk factors and bone marrow response 14 days after induction chemotherapy. Our study included 116 precursor B-cell (n = 104) and T-cell (n = 12) ALL patients treated with our protocol between 1988 and 1999. The patients were classified into 3 initial risk groups on the basis of conventional risk factors (56 in the low-risk, 33 in the high-risk, and 27 in the very high-risk groups). All patients received similar systemic chemotherapy regimens before the evaluation of their bone marrow on day 14. We evaluated the marrow of 69 patients as M1 (less than 5% blasts), 25 as M2 (5%-25% blasts), and 22 as M3 (more than 25% blasts). Although all patients attained an initial complete remission (CR), relapse was noted in 33 of the 116 patients, and 15 patients died. All of the M1 marrow patients, irrespective of the initial risk group, showed the best event-free survival rate (85.1% +/- 3 4.4%), the lowest relapse rate (14.5%), and the highest attainment of a second CR (100%); they were defined as the new R1 prognostic group. The low-risk patients with M2 or M3 marrow (R2 group) had a relatively high relapse rate, but all of these relapsed patients were treated successfully with subsequent therapy. High- or very high-risk patients with M2 or M3 marrow (R3 group) had the worst prognosis. Our new prognostic definition (R1, R2, R3) incorporating day 14 marrow findings is useful to tailor early-phase treatments for better therapeutic results in childhood ALL.
Collapse
Affiliation(s)
- Akira Morimoto
- Department of Pediatrics, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Japan.
| | | | | | | | | | | | | |
Collapse
|
50
|
Fioredda F, Plebani A, Hanau G, Haupt R, Giacchino M, Barisone E, Balbo L, Castagnola E. Re-immunisation schedule in leukaemic children after intensive chemotherapy: a possible strategy. Eur J Haematol 2005; 74:20-3. [PMID: 15613102 DOI: 10.1111/j.1600-0609.2004.00340.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The aim of this retrospective study was to test the residual humoral immunity to compulsory vaccines after the end of chemotherapy for acute lymphoblastic leukaemia in a cohort of 70 Italian children. All the patients, who had been immunised according to the Italian schedule prior to the disease, were tested for antibody levels against tetanus and hepatitis B at a median of 10 months after the end of therapy. Median age at diagnosis of leukaemia was 48 months, and median age at vaccine titration was 84 months. The protective level of antibodies for tetanus and hepatitis B was shown in 83% and 81% of patients, respectively; the remaining 17% and 19% were not protected against the two pathogens. Double negativity was observed in only four of 62 (6%) patients in the cohort. These data were comparable with published data regarding healthy children of the same age and from the same geographical areas. Therefore, given the direct and indirect costs of performing laboratory tests, as well as the cost of revaccination, our proposal is to continue the vaccination schedule according to the child's age without any titration screening 6 months after the end of therapy. Larger studies are needed to confirm these observations.
Collapse
Affiliation(s)
- Francesca Fioredda
- Department of Pediatric Hematology, Oncology and Bone Marrow Transplantation, Scientific Directorate, G. Gaslini Children's Hospital, Genova, Italy.
| | | | | | | | | | | | | | | |
Collapse
|