1
|
Yanagisawa R, Kato M, Sakaguchi K, Nakagoshi R, Ogiso Y, Fukushima K, Sakashita K. A case of pure erythroid leukemia with MYB-GATA1 fusion that developed tumor lysis syndrome with dexamethasone. Pediatr Blood Cancer 2020; 67:e28285. [PMID: 32634254 DOI: 10.1002/pbc.28285] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 11/11/2022]
Affiliation(s)
- Ryu Yanagisawa
- Division of Blood Transfusion, Shinshu University Hospital, Matsumoto, Japan.,Center for Advanced Cell Therapy, Shinshu University Hospital, Matsumoto, Japan.,Life Science Research Center, Nagano Children's Hospital, Azumino, Japan
| | - Motohiro Kato
- Department of Pediatric Hematology and Oncology Research, Research Institute, National Center for Child Health and Development, Tokyo, Japan.,Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Kimiyoshi Sakaguchi
- Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Ritsuko Nakagoshi
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Yoshifumi Ogiso
- Department of Laboratory Medicine, Nagano Children's Hospital, Azumino, Japan
| | - Keitaro Fukushima
- Department of Pediatrics, Dokkyo Medical University, Shimotsuga, Japan
| | - Kazuo Sakashita
- Department of Hematology/Oncology, Nagano Children's Hospital, Azumino, Japan
| |
Collapse
|
2
|
Aldehyde dehydrogenases inhibition eradicates leukemia stem cells while sparing normal progenitors. Blood Cancer J 2016; 6:e469. [PMID: 27611922 PMCID: PMC5056970 DOI: 10.1038/bcj.2016.78] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 08/04/2016] [Indexed: 12/18/2022] Open
Abstract
The vast majority of patients with acute myeloid leukemia (AML) achieve complete remission (CR) after standard induction chemotherapy. However, the majority subsequently relapse and die of the disease. A leukemia stem cell (LSC) paradigm has been invoked to explain this failure of CR to reliably translate into cure. Indeed, LSCs are highly enriched in CD34+CD38− leukemic cells that exhibit positive aldehyde dehydrogenase activity (ALDH+) on flow cytometry, these LSCs are resistant to currently existing treatments in AML such as cytarabine and anthracycline that, at the cost of great toxicity on normal cells, are highly active against the leukemic bulk, but spare the LSCs responsible for relapse. To try to combat the LSC population selectively, a well-characterized ALDH inhibitor by the trivial name of dimethyl ampal thiolester (DIMATE) was assessed on sorted CD34+CD38− subpopulations from AML patients and healthy patients. ALDH activity and cell viability were monitored by flow cytometry. From enzyme kinetic studies DIMATE is an active enzyme-dependent, competitive, irreversible inhibitor of ALDH1. On cells in culture, DIMATE is a powerful inhibitor of ALDHs 1 and 3, has a major cytotoxic activity on human AML cell lines. Moreover, DIMATE is highly active against leukemic populations enriched in LSCs, but, unlike conventional chemotherapy, DIMATE is not toxic for healthy hematopoietic stem cells which retained, after treatment, their self-renewing and multi-lineage differentiation capacity in immunodeficient mice, xenografted with human leukemic cells. DIMATE eradicates specifically human AML cells and spares healthy mouse hematologic cells.
Collapse
|
3
|
Xie C, Drenberg C, Edwards H, Caldwell JT, Chen W, Inaba H, Xu X, Buck SA, Taub JW, Baker SD, Ge Y. Panobinostat enhances cytarabine and daunorubicin sensitivities in AML cells through suppressing the expression of BRCA1, CHK1, and Rad51. PLoS One 2013; 8:e79106. [PMID: 24244429 PMCID: PMC3823972 DOI: 10.1371/journal.pone.0079106] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 09/18/2013] [Indexed: 11/24/2022] Open
Abstract
Acute myeloid leukemia (AML) remains a challenging disease to treat and urgently requires new therapies to improve its treatment outcome. In this study, we investigated the molecular mechanisms underlying the cooperative antileukemic activities of panobinostat and cytarabine or daunorubicin (DNR) in AML cell lines and diagnostic blast samples in vitro and in vivo. Panobinostat suppressed expression of BRCA1, CHK1, and RAD51 in AML cells in a dose-dependent manner. Further, panobinostat significantly increased cytarabine- or DNR-induced DNA double-strand breaks and apoptosis, and abrogated S and/or G2/M cell cycle checkpoints. Analogous results were obtained by shRNA knockdown of BRCA1, CHK1, or RAD51. Cotreatment of NOD-SCID-IL2Rγnull mice bearing AML xenografts with panobinostat and cytarabine significantly increased survival compared to either cytarabine or panobinostat treatment alone. Additional studies revealed that panobinostat suppressed the expression of BRCA1, CHK1, and RAD51 through downregulation of E2F1 transcription factor. Our results establish a novel mechanism underlying the cooperative antileukemic activities of these drug combinations in which panobinostat suppresses expression of BRCA1, CHK1, and RAD51 to enhance cytarabine and daunorubicin sensitivities in AML cells.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/pharmacology
- BRCA1 Protein/biosynthesis
- Checkpoint Kinase 1
- Child
- Child, Preschool
- Cytarabine/pharmacology
- Daunorubicin/agonists
- Daunorubicin/pharmacology
- Drug Agonism
- Female
- G2 Phase Cell Cycle Checkpoints/drug effects
- Gene Expression Regulation, Leukemic/drug effects
- Heterografts
- Humans
- Hydroxamic Acids/pharmacology
- Indoles/pharmacology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- M Phase Cell Cycle Checkpoints/drug effects
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Neoplasm Transplantation
- Panobinostat
- Protein Kinases/biosynthesis
- Rad51 Recombinase/biosynthesis
- U937 Cells
Collapse
Affiliation(s)
- Chengzhi Xie
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- The State Engineering Laboratory of AIDS Vaccine, College of Life Science, Jilin University, Changchun, China
| | - Christina Drenberg
- Pharmaceutical Sciences Department, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - J. Timothy Caldwell
- MD/PhD Program, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Cancer Biology Program, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Wei Chen
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Hiroto Inaba
- Department of Oncology, Division of Leukemia/Lymphoma, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Xuelian Xu
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Steven A. Buck
- Division of Pediatric Hematology/Oncology, Children’s Hospital of Michigan, Detroit, Michigan, United States of America
| | - Jeffrey W. Taub
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Division of Pediatric Hematology/Oncology, Children’s Hospital of Michigan, Detroit, Michigan, United States of America
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Sharyn D. Baker
- Pharmaceutical Sciences Department, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Yubin Ge
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- The State Engineering Laboratory of AIDS Vaccine, College of Life Science, Jilin University, Changchun, China
- * E-mail:
| |
Collapse
|
4
|
Abstract
Pediatric acute myeloid leukemia (AML) is currently associated with survival rates as high as 70%. However, many events still occur, side effects are significant, and late effects occur and can even be life-threatening. Thus, the treatment of pediatric AML still needs further improvement. While most study groups agree on several principles of AML treatment, many unanswered questions and even controversies remain, which will be the topic of this review. Relapsed AML, the most frequent event in children, will also be discussed. The controversies justify future clinical studies. Fortunately, biotechnical developments provide novel treatment targets and targeted drugs, and will enable minimal residual disease-driven tailored therapy. Moreover, a wide range of new drugs is being developed. International collaboration is required to perform randomized, or even single-arm clinical studies, in this setting of subgroup-directed therapy, and fortunately is being accomplished. Therefore, optimism is justified and the treatment of pediatric AML will continue to improve.
Collapse
Affiliation(s)
- Gertjan J L Kaspers
- Department of Pediatric Oncology/Hematology of the VU University Medical Center, De Boelelaan 1117, NL-1081, HV Amsterdam, The Netherlands.
| |
Collapse
|
5
|
Grafone T, Palmisano M, Nicci C, Storti S. An overview on the role of FLT3-tyrosine kinase receptor in acute myeloid leukemia: biology and treatment. Oncol Rev 2012; 6:e8. [PMID: 25992210 PMCID: PMC4419636 DOI: 10.4081/oncol.2012.e8] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Revised: 03/06/2012] [Accepted: 04/13/2012] [Indexed: 01/10/2023] Open
Abstract
Hematopoiesis, the process by which the hematopoietic stem cells and progenitors differentiate into blood cells of various lineages, involves complex interactions of transcription factors that modulate the expression of downstream genes and mediate proliferation and differentiation signals. Despite the many controls that regulate hematopoiesis, mutations in the regulatory genes capable of promoting leukemogenesis may occur. The FLT3 gene encodes a tyrosine kinase receptor that plays a key role in controlling survival, proliferation and differentiation of hematopoietic cells. Mutations in this gene are critical in causing a deregulation of the delicate balance between cell proliferation and differentiation. In this review, we provide an update on the structure, synthesis and activation of the FLT3 receptor and the subsequent activation of multiple downstream signaling pathways. We also review activating FLT3 mutations that are frequently identified in acute myeloid leukemia, cause activation of more complex downstream signaling pathways and promote leukemogenesis. Finally, FLT3 has emerged as an important target for molecular therapy. We, therefore, report on some recent therapies directed against it.
Collapse
Affiliation(s)
- Tiziana Grafone
- Department of Onco-Hematology, Fondazione di Ricerca e Cura Giovanni Paolo II, Campobasso
| | - Michela Palmisano
- San Raffaele Vita-Salute University, School of Molecular Medicine, Milano, Italy
| | - Chiara Nicci
- Department of Onco-Hematology, Fondazione di Ricerca e Cura Giovanni Paolo II, Campobasso
| | - Sergio Storti
- Department of Onco-Hematology, Fondazione di Ricerca e Cura Giovanni Paolo II, Campobasso
| |
Collapse
|
6
|
Abraham A, Varatharajan S, Abbas S, Zhang W, Shaji RV, Ahmed R, Abraham A, George B, Srivastava A, Chandy M, Mathews V, Balasubramanian P. Cytidine deaminase genetic variants influence RNA expression and cytarabine cytotoxicity in acute myeloid leukemia. Pharmacogenomics 2012; 13:269-82. [DOI: 10.2217/pgs.11.149] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: Cytidine deaminase (CDA) irreversibly deaminates cytarabine (Ara-C), a key component of acute myeloid leukemia (AML) induction and consolidation therapy. CDA overexpression results in Ara-C resistance, while decreased expression is associated with toxicity. We evaluated factors influencing variation in CDA mRNA expression in adult AML patients and normal controls, and how they contributed to Ara-C cytotoxicity in AML cells. Materials & methods: CDA mRNA expression in 100 de novo AML patients and 36 normal controls were determined using quantitative reverse-transcriptase PCR. Genetic variants in the CDA gene were screened by direct sequencing. IC50 of Ara-C was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Results: CDA RNA expression as well as Ara-C IC50 showed wide variation in AML samples and normal controls. Fourteen sequence variants were identified, three of which (-33delC, intron 2 TCAT repeat and the 3´untranslated region 816delC variants) showed significant association with RNA expression and the nonsynonymous coding variant 79A>C was associated with Ara-C cytotoxicity. Conclusion: CDA genetic variants explain the variation in RNA expression and may be candidates for individualizing Ara-C therapy. Original submitted 8 July 2011; Revision submitted 10 October 2011
Collapse
Affiliation(s)
- Ajay Abraham
- Christian Medical College, Vellore 632004, India
| | | | - Salar Abbas
- Christian Medical College, Vellore 632004, India
| | - Wei Zhang
- Department of Pediatrics, Institute of Human Genetics, Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | - Rayaz Ahmed
- Christian Medical College, Vellore 632004, India
| | - Aby Abraham
- Christian Medical College, Vellore 632004, India
| | - Biju George
- Christian Medical College, Vellore 632004, India
| | | | | | | | | |
Collapse
|
7
|
Xu X, Xie C, Edwards H, Zhou H, Buck SA, Ge Y. Inhibition of histone deacetylases 1 and 6 enhances cytarabine-induced apoptosis in pediatric acute myeloid leukemia cells. PLoS One 2011; 6:e17138. [PMID: 21359182 PMCID: PMC3040224 DOI: 10.1371/journal.pone.0017138] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 01/21/2011] [Indexed: 01/31/2023] Open
Abstract
Background Pediatric acute myeloid leukemia (AML) remains a challenging disease to treat even with intensified cytarabine-based chemotherapy. Histone deacetylases (HDACs) have been reported to be promising therapeutic targets for treating AML. However, HDAC family members that are involved in chemotherapy sensitivities remain unknown. In this study, we sought to identify members of the HDAC family that are involved in cytarabine sensitivities, and to select the optimal HDACI that is most efficacious when combined with cytarabine for treating children with AML. Methodology Expression profiles of classes I, II, and IV HDACs in 4 pediatric AML cell lines were determined by Western blotting. Inhibition of class I HDACs by different HDACIs was measured post immnunoprecipitation. Individual down-regulation of HDACs in pediatric AML cells was performed with lentiviral shRNA. The effects of cytarabine and HDACIs on apoptosis were determined by flow cytometry analysis. Results Treatments with structurally diverse HDACIs and HDAC shRNA knockdown experiments revealed that down-regulation of both HDACs 1 and 6 is critical in enhancing cytarabine-induced apoptosis in pediatric AML, at least partly mediated by Bim. However, down-regulation of HDAC2 may negatively impact cytarabine sensitivities in the disease. At clinically achievable concentrations, HDACIs that simultaneously inhibited both HDACs 1 and 6 showed the best anti-leukemic activities and significantly enhanced cytarabine-induced apoptosis. Conclusion Our results further confirm that HDACs are bona fide therapeutic targets for treating pediatric AML and suggest that pan-HDACIs may be more beneficial than isoform-specific drugs.
Collapse
Affiliation(s)
- Xuelian Xu
- Developmental Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- The State Engineering Laboratory of AIDS Vaccine, College of Life Science, Jilin University, Changchun, People's Republic of China
| | - Chengzhi Xie
- Developmental Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- The State Engineering Laboratory of AIDS Vaccine, College of Life Science, Jilin University, Changchun, People's Republic of China
| | - Holly Edwards
- Developmental Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Hui Zhou
- The State Engineering Laboratory of AIDS Vaccine, College of Life Science, Jilin University, Changchun, People's Republic of China
| | - Steven A. Buck
- Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, Michigan, United States of America
| | - Yubin Ge
- Developmental Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- The State Engineering Laboratory of AIDS Vaccine, College of Life Science, Jilin University, Changchun, People's Republic of China
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
8
|
Xie C, Edwards H, Xu X, Zhou H, Buck SA, Stout ML, Yu Q, Rubnitz JE, Matherly LH, Taub JW, Ge Y. Mechanisms of synergistic antileukemic interactions between valproic acid and cytarabine in pediatric acute myeloid leukemia. Clin Cancer Res 2010; 16:5499-510. [PMID: 20889917 DOI: 10.1158/1078-0432.ccr-10-1707] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To determine the possibility of synergistic antileukemic activity and the underlying molecular mechanisms associated with cytarabine combined with valproic acid (VPA; a histone deacetylase inhibitor and a Food and Drug Administration-licensed drug for treating both children and adults with epilepsy) in pediatric acute myeloid leukemia (AML). EXPERIMENTAL DESIGN The type and extent of antileukemic interactions between cytarabine and VPA in clinically relevant pediatric AML cell lines and diagnostic blasts from children with AML were determined by MTT assays and standard isobologram analyses. The effects of cytarabine and VPA on apoptosis and cell cycle distributions were determined by flow cytometry analysis and caspase enzymatic assays. The effects of the two agents on DNA damage and Bcl-2 family proteins were determined by Western blotting. RESULTS We showed synergistic antileukemic activities between cytarabine and VPA in four pediatric AML cell lines and nine diagnostic AML blast samples. t(8;21) AML blasts were significantly more sensitive to VPA and showed far greater sensitivities to combined cytarabine and VPA than non-t(8;21) AML cases. Cytarabine and VPA cooperatively induced DNA double-strand breaks, reflected in induction of γH2AX and apoptosis, accompanied by activation of caspase-9 and caspase-3. Further, VPA induced Bim expression and short hairpin RNA knockdown of Bim resulted in significantly decreased apoptosis induced by cytarabine and by cytarabine plus VPA. CONCLUSIONS Our results establish global synergistic antileukemic activity of combined VPA and cytarabine in pediatric AML and provide compelling evidence to support the use of VPA in the treatment of children with this deadly disease.
Collapse
Affiliation(s)
- Chengzhi Xie
- Developmental Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, 110 E. Warren Avenue, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Gorman MF, Ji L, Ko RH, Barnette P, Bostrom B, Hutchinson R, Raetz E, Seibel NL, Twist CJ, Eckroth E, Sposto R, Gaynon PS, Loh ML. Outcome for children treated for relapsed or refractory acute myelogenous leukemia (rAML): a Therapeutic Advances in Childhood Leukemia (TACL) Consortium study. Pediatr Blood Cancer 2010; 55:421-9. [PMID: 20658611 DOI: 10.1002/pbc.22612] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Current event-free survival (EFS) rates for children with newly diagnosed acute myeloid leukemia (AML) approach 50-60%. We hypothesize that further improvements in survival are unlikely to be achieved with traditional approaches such as dose intensive chemotherapy or hematopoietic stem cell transplants, since these therapies have been rigorously explored in clinical trials. This report highlights efforts to assess the response rates and survival outcomes after first or greater relapse in children with AML. PROCEDURE We performed a retrospective cohort review of pediatric patients with relapsed and refractory AML (rAML) previously treated at TACL institutions between the years of 1995 and 2004. Data regarding disease characteristics at diagnosis and relapse, treatment response, and survival was collected on 99 patients and 164 medullary relapses or treatment failures. RESULTS The complete response (CR) rate following the second therapeutic attempt was 56 +/- 5%. CR rates following a third treatment attempt was 25 +/- 8% while 17 +/- 7% achieved CR following the fourth through sixth treatments. The 5-year disease-free survival in patients achieving CR following a second therapeutic attempt was 43 +/- 7%. The 5-year EFS and overall survival (OS) rates for all patients receiving a second treatment attempt was 24 +/- 5% and 29 +/- 5%, respectively. CONCLUSIONS This CR rate following a second therapeutic attempt and OS rate in patients with rAML is consistent with the literature. There are limited published data of CR rates for subsequent relapses. Our data can serve as a historical benchmark to compare outcomes of future therapeutic trials in rAML against traditional chemotherapy regimens.
Collapse
Affiliation(s)
- Matthew F Gorman
- Therapeutic Advances in Childhood Leukemia Consortium, USC-CHLA Institute for Pediatric Clinical Research, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Styczynski J, Wysocki M, Debski R, Czyzewski K, Kolodziej B, Rafinska B, Kubicka M, Koltan S, Koltan A, Pogorzala M, Kurylak A, Olszewska-Slonina D, Balwierz W, Juraszewska E, Wieczorek M, Olejnik I, Krawczuk-Rybak M, Kuzmicz M, Kowalczyk J, Stefaniak J, Badowska W, Sonta-Jakimczyk D, Szczepanski T, Matysiak M, Malinowska I, Stanczak E, Wachowiak J, Konatkowska B, Gil L, Balcerska A, Maciejka-Kapuscinska L. Predictive value of multidrug resistance proteins and cellular drug resistance in childhood relapsed acute lymphoblastic leukemia. J Cancer Res Clin Oncol 2007; 133:875-93. [PMID: 17671794 DOI: 10.1007/s00432-007-0274-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2006] [Accepted: 06/27/2007] [Indexed: 11/29/2022]
Abstract
PURPOSE Cellular resistance in childhood acute leukemias might be related to profile and function of multidrug resistance proteins and apoptosis regulating proteins. The aims of the study were: (1) analysis of expression of MRP1, PGP1, LRP, BCL-2 and p53 proteins; (2) correlation with ex vivo drug resistance, and (3) analysis of their prognostic impact on clinical outcome in childhood acute lymphoblastic (ALL) and acute myeloid (AML) leukemia. METHODS Total number of 787 children diagnosed for initial ALL (n = 527), relapsed ALL (n = 104), initial AML (n = 133) and relapsed AML (n = 23) were included into the study. Mean follow-up period was 3.5 years. Drug resistance for up to 30 anticancer agents was performed by the MTT assay. Expression of all proteins was tested by flow cytometry. RESULTS Both initial AML and relapsed ALL samples showed higher drug resistance than initial ALL samples. No significant differences were found in drug resistance between initial and relapsed AML samples. The presence of multidrug resistance and apoptosis proteins had no impact on pDFS in iALL and iAML, however strong trend towards adverse prognostic impact of MRP1, PGP and LRP on pDFS in rALL was observed. The same trend was observed for each of analyzed co-expressions of tested multidrug resistance proteins. CONCLUSIONS The phenomenon of cellular drug resistance in childhood acute leukemias is multifactorial and plays an important role in response to therapy. Expression of MRP1, PGP and LRP proteins, as well as their co-expression play possible role in childhood relapsed ALL.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Adolescent
- Adult
- Antineoplastic Agents/pharmacology
- Child
- Child, Preschool
- Drug Resistance, Multiple
- Drug Resistance, Neoplasm
- Female
- Flow Cytometry
- Gene Expression Regulation, Leukemic
- Humans
- Immunophenotyping
- Infant
- Infant, Newborn
- Male
- Multidrug Resistance-Associated Proteins/genetics
- Multidrug Resistance-Associated Proteins/metabolism
- Neoplasm Recurrence, Local/diagnosis
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Prognosis
- Vault Ribonucleoprotein Particles/genetics
- Vault Ribonucleoprotein Particles/metabolism
Collapse
Affiliation(s)
- Jan Styczynski
- Department of Pediatric Hematology and Oncology, Collegium Medicum, Nicolaus Copernicus University, ul. Curie-Sklodowskiej 9, 85-094, Bydgoszcz, Poland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Barnard DR, Alonzo TA, Gerbing RB, Lange B, Woods WG. Comparison of childhood myelodysplastic syndrome, AML FAB M6 or M7, CCG 2891: report from the Children's Oncology Group. Pediatr Blood Cancer 2007; 49:17-22. [PMID: 16856158 DOI: 10.1002/pbc.20951] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Myelodysplastic syndromes (MDS), acute erythroleukemia (FAB M6), and acute megakaryocytic leukemia (FAB M7) have overlapping features. PROCEDURE Children without Down syndrome or acute promyelocytic leukemia who were newly diagnosed with primary myelodysplastic syndrome or acute myeloid leukemia (AML) M6 or M7 were compared to children with de novo AML M0-M5. All children were entered on the Children's Cancer Group therapeutic research study CCG 2891. RESULTS The presentation and outcomes of the 132 children diagnosed with MDS (60 children), AML FAB M6 (19 children), or AML FAB M7 (53 children) were similar. Children with AML FAB M7 were diagnosed at a significantly younger age (P = 0.001). Children with MDS, M6, or M7 had significantly lower white blood cell (WBC) counts (P = 0.001), lower peripheral blast counts (P < 0.001), and an increased frequency of -7/7q- (P = 0.003) at presentation. All three groups had significantly inferior overall survival (OS) (P < 0.001) and event free survival (P < 0.001) compared with the 748 children diagnosed with AML FAB M0-M5 when assessed from entry on study. This poor survival was largely attributable to induction death and failure. However, when assessed from successful completion of induction therapy, the 5-year OS (P = 0.090)(49.1 vs. 56.9%) and disease-free survival (DFS) (P = 0.113)(38.0 vs. 46.3%) therapy were not significantly different from other children with AML. CONCLUSIONS Childhood AML FAB M6 and AML M7 resemble MDS in presentation, poor induction success rates, and outcomes.
Collapse
MESH Headings
- Acute Disease
- Child
- Child, Preschool
- Diagnosis, Differential
- Disease-Free Survival
- Female
- Humans
- Leukemia, Erythroblastic, Acute/diagnosis
- Leukemia, Erythroblastic, Acute/mortality
- Leukemia, Megakaryoblastic, Acute/diagnosis
- Leukemia, Megakaryoblastic, Acute/mortality
- Male
- Myelodysplastic Syndromes/diagnosis
- Myelodysplastic Syndromes/mortality
- Prognosis
- Remission Induction
- Survival Rate
- Treatment Outcome
Collapse
|
12
|
Sohn SK, Kim JG, Kim DH. Tailored strategy for AML patients receiving allogeneic peripheral blood stem cell transplantation. J Clin Apher 2007; 21:207-13. [PMID: 16425195 DOI: 10.1002/jca.20078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Considering the heterogeneity of acute myelogenous leukemia (AML), along with the pros and cons of allogeneic peripheral blood stem cell transplantation (PBSCT), a tailored strategy is needed to minimize the transplant-related mortality and maximize the transplant outcomes in AML patients exhibiting certain factors that have an impact on the post-transplant quality of life and outcomes. The factors that need to be considered when tailoring a strategy in an allogeneic PBSCT setting include the recipient's performance status and co-morbid disease include AML risk stratification, disease status, expected severity of graft-versus-host disease, and the necessity of a graft-versus-leukemia effect. Accordingly, this review article describes a possible tailoring strategy for AML patients receiving allogeneic PBSCT based on certain factors influencing the transplant outcome.
Collapse
Affiliation(s)
- Sang Kyun Sohn
- Department of Hematology/Oncology, Kyungpook National University Hospital, Daegu, Korea.
| | | | | |
Collapse
|
13
|
Gurbuxani S. CD36 - a marker for drug sensitive acute megakaryoblastic leukemia. Leuk Lymphoma 2006; 47:2004-5. [PMID: 17071466 DOI: 10.1080/10428190600763579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Sandeep Gurbuxani
- Department of Pathology, University of Chicago Hospitals, Chicago, IL 60637, USA.
| |
Collapse
|
14
|
Kaspers GJL, Reinhardt D, Fleischhack G, Armendariz H, Stark B, Zwaan CM, Zimmermann M, Creutzig U. Low efficacy of methotrexate in childhood acute myeloid leukemia (AML): single-agent therapeutic window study in relapsed AML. Pediatr Blood Cancer 2006; 47:539-42. [PMID: 16358301 DOI: 10.1002/pbc.20727] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND The efficacy in pediatric acute myeloid leukemia (AML) of single-agent methotrexate (MTX) at a higher dose than previously applied, 1,000 mg/m2, given as a theoretically beneficial 36-hr continuous infusion, is unknown, but may be beneficial based on preclinical data. PROCEDURE We performed a therapeutic window study in children with first relapsed AML treated in four different countries. RESULTS Based on a comparison between the percentage of leukemic blasts in the bone marrow shortly before and 7-10 days after the MTX infusion, none of the first cohort of nine patients showed a good response, defined as a reduction of blasts of at least 50%. Therefore, the study was closed, concluding that the probability of a good response in this patient-group was most likely to be less than 30%. By that time, another four patients had been enrolled, of which one patient with a late relapsed AML FAB type M7 showed a good response. Toxicity of MTX was limited and tolerable. CONCLUSIONS This study shows that single-agent MTX in the applied regimen in pediatric relapsed AML has limited efficacy. However, it also demonstrates the feasibility of an international and therapeutic window phase II study in pediatric relapsed AML.
Collapse
Affiliation(s)
- G J L Kaspers
- Department of Pediatric Hematology/Oncology, VU University Medical Center, Amsterdam, The Netherlands, and Department of Pediatric Hematology/Oncology, Children's Medical Hospital, University of Bonn, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
Childhood acute myeloid leukemia is rare, but accounts for a significant number of malignancy-related deaths in this age group. However, the prognosis has improved over past decades, and survival rates of 60% and above have been reported. Still, this implies that more than a third of children and adolescents die from this disease. Moreover, treatment is intensive, and quality of life and late effects are worrying issues. Therefore, there is a need for further improved treatment of pediatric acute myeloid leukemia. This review describes several important developments in this respect, such as improved diagnostics, prognostic factors, subgroup-directed and tailored treatment, and targeted therapy. In addition, background information is provided and current treatment strategies are described, as well as the late effects of treatment. Most groups now have risk-group adapted protocols, with allogeneic stem cell transplantation often being reserved for the higher risk group. Even in these cases, the benefit of stem cell transplantation has not been demonstrated beyond reasonable doubt with current high-intensive chemotherapy. Similarly, the use of cranial irradiation for CNS prophylaxis and maintenance treatment does not seem to be indicated in general. Subgroup-directed treatment has become a reality for acute myeloid leukemia in young children with Down's syndrome and in acute promyelocytic leukemia. In addition to tailoring therapy according to biologic features and especially monitoring treatment by measurements of minimal residual disease, targeted therapy for subgroups with activating mutations in receptor tyrosine kinases will further optimize the treatment of pediatric acute myeloid leukemia. Together with the development of many novel agents that have different mechanisms of action than the currently available anticancer agents, and improved supportive care, it is realistic that the prognosis of acute myeloid leukemia in children and adolescents will improve further in the next 5-10 years.
Collapse
Affiliation(s)
- Edske ter Bals
- Pediatric Oncology/Hematology, VU University Medical Center, De Boelelaan 1117, NL-1081 HV, Amsterdam, The Netherlands
| | | |
Collapse
|
16
|
Kaspers GJL, Creutzig U. Pediatric acute myeloid leukemia: international progress and future directions. Leukemia 2006; 19:2025-9. [PMID: 16304569 DOI: 10.1038/sj.leu.2403958] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
17
|
Jubert C, Geoerger B, Grill J, Hartmann O, Vassal G. [Targeted therapies in pediatric oncology: a new therapeutic approach?]. Arch Pediatr 2005; 13:189-94. [PMID: 16298518 DOI: 10.1016/j.arcped.2005.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2005] [Accepted: 10/01/2005] [Indexed: 10/25/2022]
Abstract
A multidisciplinary therapeutic approach has led to significant increase in survival of children with cancer, however often with a high rate of severe sequela. Better understanding in tumor cell biology and transformation process allowed to describe active tyrosine kinases (mainly growth factor receptors) as a new target for cancer treatment. This review presents 2 approaches to target receptor tyrosine kinase activity: on one hand, antibodies that target the extracellular domain, the natural ligand binding site, and on the other hand, small inhibiting molecules, such as imatinib, targeted against the activated intracellular receptor tyrosine kinase. We focus on their clinical development and current application in the treatment of childhood cancer. Targeted therapies are in full rise and new perspectives are explored, such as their association to other treatment modalities and the targeting of microenvironment. This new therapeutic approach necessitates well designed clinical trials that include relevant biomarkers to evaluate its real therapeutic potential.
Collapse
Affiliation(s)
- C Jubert
- UPRES EA 3535, Département de Pédiatrie, Pharmacologie et Nouveaux Traitements des Cancers, Institut Gustave-Roussy, 39, rue Camille-Desmoulins, 94805 Villejuif cedex, France
| | | | | | | | | |
Collapse
|
18
|
Current Awareness in Hematological Oncology. Hematol Oncol 2005. [DOI: 10.1002/hon.729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|