951
|
In vitro secretion of matrix metalloprotease 9 is a prognostic marker in childhood acute lymphoblastic leukemia. Leuk Res 2010; 34:24-31. [DOI: 10.1016/j.leukres.2009.07.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2009] [Revised: 07/25/2009] [Accepted: 07/26/2009] [Indexed: 11/18/2022]
|
952
|
Tangen JM, Fløisand Y, Haukås E, Næss I, Skjelbakken T, Stapnes C, Tjønnfjord G. Overlevelse hos voksne med akutt lymfoblastisk leukemi. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2010; 130:1710-3. [DOI: 10.4045/tidsskr.09.1293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
|
953
|
Stocco G, Crews KR, Evans WE. Genetic polymorphism of inosine-triphosphate-pyrophosphatase influences mercaptopurine metabolism and toxicity during treatment of acute lymphoblastic leukemia individualized for thiopurine-S-methyl-transferase status. Expert Opin Drug Saf 2010; 9:23-37. [PMID: 20021291 DOI: 10.1517/14740330903426151] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
IMPORTANCE OF THE FIELD Although genetic polymorphisms in the gene encoding human thiopurine methyltransferase (TPMT) are known to have a marked effect on mercaptopurine metabolism and toxicity, there are many patients with wild-type TPMT who develop toxicity. Furthermore, when mercaptopurine dosages are adjusted in patients who are heterozygous at the TPMT locus, there are still some patients who develop toxicity for reasons that are not fully understood. Therefore, we recently studied the effects of a common polymorphism in another gene encoding an enzyme involved in mercaptopurine metabolism (SNP rs1127354 in inosine-triphospate-pyrophosphatase, ITPA), showing that genetic polymorphism of ITPA is a significant determinant of mercaptopurine metabolism and of febrile neutropenia following combination chemotherapy of acute lymphoblastic leukemia (ALL) in which mercaptopurine doses are individualized based on TPMT genotype. AREA COVERED IN THIS REVIEW In this review, we summarize the knowledge available about the effect and clinical relevance of TPMT and ITPA on mercaptopurine pharmacogenomics, with a particular focus on the use of this medication in pediatric patients with ALL. WHAT THE READER WILL GAIN Reader will gain insights into: i) the effects of pharmacogenomic traits on mercaptopurine toxicity and efficacy for the treatment of ALL and ii) individualization strategies that can be used to mitigate toxicity without compromising efficacy in pediatric patients with ALL. TAKE HOME MESSAGE Mercaptopurine dose can be adjusted on the basis of TPMT genotype to mitigate toxicity in pediatric patients with ALL. As treatment is individualized in this way for the most relevant genetic determinant of drug response (i.e., for mercaptopurine, TPMT), the importance of other genetic polymorphisms emerges (e.g., ITPA).
Collapse
Affiliation(s)
- Gabriele Stocco
- St. Jude Children's Research Hospital, Department of Pharmaceutical Sciences, 262 Danny Thomas Place MS 272, Memphis, TN 38105, USA
| | | | | |
Collapse
|
954
|
Immunoglobulin heavy chain locus chromosomal translocations in B-cell precursor acute lymphoblastic leukemia: rare clinical curios or potent genetic drivers? Blood 2009; 115:1490-9. [PMID: 20042721 DOI: 10.1182/blood-2009-09-235986] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chromosomal translocations involving the immunoglobulin heavy chain (IGH) locus define common subgroups of B-cell lymphoma but are rare in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Recent fluorescent in situ hybridization and molecular cloning studies have identified several novel IGH translocations involving genes that play important roles in normal hemopoiesis, including the cytokine receptor genes CRLF2 and EPOR, all members of the CCAAT enhancer-binding protein gene family, as well as genes not normally expressed in hemopoietic cells including inhibitor of DNA binding 4. IGH translocation results in deregulated target gene expression because of juxtaposition with IGH transcriptional enhancers. However, many genes targeted by IGH translocations are also more commonly deregulated in BCP-ALL as a consequence of other genetic or epigenetic mechanisms. For example, interstitial genomic deletions also result in deregulated CRLF2 expression, whereas EPOR expression is deregulated as a consequence of the ETV6-RUNX1 fusion. The possible clinical importance of many of the various IGH translocations in BCP-ALL remains to be determined from prospective studies, but CRLF2 expression is associated with a poor prognosis. Despite their rarity, IGH chromosomal translocations in BCP-ALL therefore define not only new mechanisms of B-cell transformation but also clinically important subgroups of disease and suggest new targeted therapeutic approaches.
Collapse
|
955
|
Menendez P, Catalina P, Rodríguez R, Melen GJ, Bueno C, Arriero M, García-Sánchez F, Lassaletta A, García-Sanz R, García-Castro J. Bone marrow mesenchymal stem cells from infants with MLL-AF4+ acute leukemia harbor and express the MLL-AF4 fusion gene. J Exp Med 2009; 206:3131-41. [PMID: 19995953 PMCID: PMC2806455 DOI: 10.1084/jem.20091050] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Accepted: 11/09/2009] [Indexed: 01/15/2023] Open
Abstract
MLL-AF4 fusion is a hallmark genetic abnormality in infant B-acute lymphoblastic leukemia (B-ALL) known to arise in utero. The cellular origin of leukemic fusion genes during human development is difficult to ascertain. The bone marrow (BM) microenvironment plays an important role in the pathogenesis of several hematological malignances. BM mesenchymal stem cells (BM-MSC) from 38 children diagnosed with cytogenetically different acute leukemias were screened for leukemic fusion genes. Fusion genes were absent in BM-MSCs of childhood leukemias carrying TEL-AML1, BCR-ABL, AML1-ETO, MLL-AF9, MLL-AF10, MLL-ENL or hyperdiploidy. However, MLL-AF4 was detected and expressed in BM-MSCs from all cases of MLL-AF4(+) B-ALL. Unlike leukemic blasts, MLL-AF4(+) BM-MSCs did not display monoclonal Ig gene rearrangements. Endogenous or ectopic expression of MLL-AF4 exerted no effect on MSC culture homeostasis. These findings suggest that MSCs may be in part tumor-related, highlighting an unrecognized role of the BM milieu on the pathogenesis of MLL-AF4(+) B-ALL. MLL-AF4 itself is not sufficient for MSC transformation and the expression of MLL-AF4 in MSCs is compatible with a mesenchymal phenotype, suggesting a differential impact in the hematopoietic system and mesenchyme. The absence of monoclonal rearrangements in MLL-AF4(+) BM-MSCs precludes the possibility of cellular plasticity or de-differentiation of B-ALL blasts and suggests that MLL-AF4 might arise in a population of prehematopoietic precursors.
Collapse
Affiliation(s)
- Pablo Menendez
- Andalusian Stem Cell Bank, Centro de Investigación Biomédica, Consejería de Salud-Universidad de Granada, 18100 Granada, Spain
| | - Purificación Catalina
- Andalusian Stem Cell Bank, Centro de Investigación Biomédica, Consejería de Salud-Universidad de Granada, 18100 Granada, Spain
| | - René Rodríguez
- Andalusian Stem Cell Bank, Centro de Investigación Biomédica, Consejería de Salud-Universidad de Granada, 18100 Granada, Spain
| | - Gustavo J. Melen
- Andalusian Stem Cell Bank, Centro de Investigación Biomédica, Consejería de Salud-Universidad de Granada, 18100 Granada, Spain
| | - Clara Bueno
- Andalusian Stem Cell Bank, Centro de Investigación Biomédica, Consejería de Salud-Universidad de Granada, 18100 Granada, Spain
| | | | | | | | - Ramón García-Sanz
- University Hospital of Salamanca, Department of Haematology, Molecular Biology and HLA Typing Unit, 37007 Salamanca, Spain
| | - Javier García-Castro
- Andalusian Stem Cell Bank, Centro de Investigación Biomédica, Consejería de Salud-Universidad de Granada, 18100 Granada, Spain
- Instituto de Salud Carlos III, Centro Nacional de Microbiología, Biología Celular y del Desarrollo, 28220 Madrid, Spain
| |
Collapse
|
956
|
Stark B, Nirel R, Avrahami G, 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. Long-term results of the Israeli National Studies in childhood acute lymphoblastic leukemia: INS 84, 89 and 98. Leukemia 2009; 24:419-24. [PMID: 20016534 DOI: 10.1038/leu.2009.254] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
957
|
Kong Y, Ma W, Liu X, Zu Y, Fu Y, Wu N, Liang L, Yao L, Efferth T. Cytotoxic activity of curcumin towards CCRF-CEM leukemia cells and its effect on DNA damage. Molecules 2009; 14:5328-38. [PMID: 20032896 PMCID: PMC6255027 DOI: 10.3390/molecules14125328] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 11/20/2009] [Accepted: 12/02/2009] [Indexed: 11/16/2022] Open
Abstract
The cytotoxic activity of curcumin towards CCRF-CEM human T-cell leukemia cells was measured by the MTT assay. Tumor cells were more sensitive to the cytotoxic activity of curcumin or curcumin-Cu (II) compared to normal cells, and the IC50 of curcumin towards CCRF-CEM cells was 8.68 µM, and that of curcumin-Cu (II) was 8.14 µM. The cell cycle distribution of curcumin-treated CCRF-CEM cells was analyzed by flow cytometry. DNA damage induced by oxidants such as curcumin-Cu (II) ions is considered as one of the main causes of cell inactivation. Therefore, we analyzed the effect of curcumin on DNA damage by agarose gel electrophoresis and atomic force microscopy (AFM). Gel electrophoresis analyses showed that curcumin or Cu (II) alone failed to cause DNA damage in pBR322 plasmid DNA as compared with the normal plasmid. However, DNA plasmids were mostly damaged after treatment with curcumin of different concentrations in the presence of Cu (II). Two forms were observed by means of AFM: closed circular plasmids and linear plasmids. DNA damage induced by a combination of curcumin and Cu (II) was also found by agarose gel electrophoresis, which was applied as control method to verify the results obtained by AFM.
Collapse
Affiliation(s)
- Yu Kong
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; E-Mails: (Y.K.); (Y.Z.); (X.L.); (N.W.); (L.L.); (L.Y.)
- Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Wei Ma
- Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China; E-Mail: (W.M.)
| | - Xia Liu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; E-Mails: (Y.K.); (Y.Z.); (X.L.); (N.W.); (L.L.); (L.Y.)
- Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Yuangang Zu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; E-Mails: (Y.K.); (Y.Z.); (X.L.); (N.W.); (L.L.); (L.Y.)
- Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Yujie Fu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; E-Mails: (Y.K.); (Y.Z.); (X.L.); (N.W.); (L.L.); (L.Y.)
- Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
- Author to whom correspondence should be addressed; E-Mail: ; Fax: +86-451-82190535
| | - Nan Wu
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; E-Mails: (Y.K.); (Y.Z.); (X.L.); (N.W.); (L.L.); (L.Y.)
- Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Lu Liang
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; E-Mails: (Y.K.); (Y.Z.); (X.L.); (N.W.); (L.L.); (L.Y.)
- Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Liping Yao
- Key Laboratory of Forest Plant Ecology, Ministry of Education, Northeast Forestry University, Harbin 150040, China; E-Mails: (Y.K.); (Y.Z.); (X.L.); (N.W.); (L.L.); (L.Y.)
- Engineering Research Center of Forest Bio-preparation, Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy, University of Mainz, 55099 Mainz, Germany; E-Mail: (T.E.)
| |
Collapse
|
958
|
Long-term results of NOPHO ALL-92 and ALL-2000 studies of childhood acute lymphoblastic leukemia. Leukemia 2009; 24:345-54. [PMID: 20010622 DOI: 10.1038/leu.2009.251] [Citation(s) in RCA: 282] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Analysis of 2668 children with acute lymphoblastic leukemia (ALL) treated in two successive Nordic clinical trials (Nordic Society of Paediatric Haematology and Oncology (NOPHO) ALL-92 and ALL-2000) showed that 75% of all patients are cured by first-line therapy, and 83% are long-term survivors. Improvements in systemic and intrathecal chemotherapy have reduced the use of central nervous system (CNS) irradiation to <10% of the patients and provided a 5-year risk of isolated CNS relapse of 2.6%. Improved risk stratification and chemotherapy have eliminated the previous independent prognostic significance of gender, CNS leukemia and translocation t(1;19)(q23;p13), whereas the post-induction level of minimal residual disease (MRD) has emerged as a new risk grouping feature. Infant leukemia, high leukocyte count, T-lineage immunophenotype, translocation t(4;11)(q21;q23) and hypodiploidy persist to be associated with lower cure rates. To reduce the overall toxicity of the treatment, including the risk of therapy-related second malignant neoplasms, the current NOPHO ALL-2008 protocol does not include CNS irradiation in first remission, the dose of 6-mercaptopurine is reduced for patients with low thiopurine methyltransferase activity, and the protocol restricts the use of hematopoietic stem cell transplantation in first remission to patients without morphological remission after induction therapy or with high levels of MRD after 3 months of therapy.
Collapse
|
959
|
Angiolillo AL, Yu AL, Reaman G, Ingle AM, Secola R, Adamson PC. A phase II study of Campath-1H in children with relapsed or refractory acute lymphoblastic leukemia: a Children's Oncology Group report. Pediatr Blood Cancer 2009; 53:978-83. [PMID: 19637330 PMCID: PMC3120889 DOI: 10.1002/pbc.22209] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Despite the increasing cure rates for children with acute lymphoblastic leukemia (ALL), patients who relapse continue to have poor prognosis. The Children's Oncology Group (COG) conducted a limited institution Phase II trial of Campath-1H, a monoclonal antibody that targets CD52 on leukemic cells, in children with relapsed or refractory ALL. METHODS From October 2005 to December 2006, 13 eligible patients were enrolled on the COG phase II study of Campath-1H (ADVL0222). Campath-1H was initially administered as an intravenous infusion over 2 hr, five times per week for 1 week, then three times per week for three additional weeks. Patients with stable disease or better on day 29 could continue on to combination therapy with Campath-1H, methotrexate, and 6-mercaptopurine for two additional cycles. RESULTS One of 13 patients enrolled had a complete response to Campath-1H and 4 had stable disease. Dose limiting toxicity occurred in two out of nine fully evaluable patients (Grade IV pain and Grade III allergic reaction/hypersensitivity). No patients received combination therapy. Serum Campath-1H concentrations appeared to be somewhat lower in children with ALL compared with adult patients with chronic lymphocytic leukemia. CONCLUSION Although a single complete response was observed, activity of single agent Campath-1H appears limited. Our study does not support future single agent evaluation of Campath-1H in children with relapsed ALL.
Collapse
Affiliation(s)
- Anne L. Angiolillo
- Division of Oncology, Children’s National Medical Center, Washington, DC
| | - Alice L. Yu
- Department of Pediatrics, Rady Children’s Hospital San Diego, San Diego, CA
| | - Gregory Reaman
- Division of Oncology, Children’s National Medical Center, Washington, DC
,Children’s Oncology Group-Chair’s Office, Bethesda, MD
| | | | - Rita Secola
- Children’s Hospital of Los Angeles, Los Angeles, CA
| | - Peter C. Adamson
- Abramson Research Center, Children’s Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
960
|
Marino S, Verzegnassi F, Tamaro P, Stocco G, Bartoli F, Decorti G, Rabusin M. Response to glucocorticoids and toxicity in childhood acute lymphoblastic leukemia: role of polymorphisms of genes involved in glucocorticoid response. Pediatr Blood Cancer 2009; 53:984-91. [PMID: 19621425 DOI: 10.1002/pbc.22163] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Glucocorticoids (GCs) play a fundamental role in the treatment of pediatric acute lymphoblastic leukemia (ALL), but therapy with these agents often results in a number of severe side effects. The aim of our study was to evaluate the association between polymorphisms of genes encoding for proteins involved in the pharmacokinetics/pharmacodynamics of these drugs and the occurrence of side effects, in particular infections, in a small population of ALL children. PROCEDURE Common polymorphisms of NR3C1, ABCB1, glutathione-S-transferase (GST)-M1, GST-P1, GST-T1, and IL-10 genes were analyzed in 36 pediatric patients with ALL, treated according to the AIEOP-BMF ALL 2000 study protocol. Toxicities occurring during the induction and reinduction periods were assessed and their association with genotypes was evaluated. RESULTS In univariate analysis, the risk of severe infections was increased in subjects with the GST-M1 null genotype, while patients with the GST-M1 normal genotype had significantly more moderate degree infections. The results were confirmed by multivariate analysis. Selection from the reference models of independent variables based on Akaike Information Criteria (AIC) scores maintained the GST-M1 genotype variable in the model to predict severe infections, and the ABCB1 C3435T and GST-M1 genotype variables in the model for moderate infections. CONCLUSIONS GST-M1 genotype may influence the severity of infections in ALL children during GC administration.
Collapse
Affiliation(s)
- Sara Marino
- I.R.C.C.S Burlo Garofolo, UO Pediatric Hemato-Oncology, Trieste, Italy
| | | | | | | | | | | | | |
Collapse
|
961
|
Early lineage switch in an infant acute lymphoblastic leukemia. Int J Hematol 2009; 90:653-655. [PMID: 19936875 DOI: 10.1007/s12185-009-0446-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 10/20/2009] [Accepted: 10/21/2009] [Indexed: 10/20/2022]
|
962
|
Goldsby RE, Liu Q, Nathan PC, Bowers DC, Yeaton-Massey A, Raber SH, Hill D, Armstrong GT, Yasui Y, Zeltzer L, Robison LL, Packer RJ. Late-occurring neurologic sequelae in adult survivors of childhood acute lymphoblastic leukemia: a report from the Childhood Cancer Survivor Study. J Clin Oncol 2009; 28:324-31. [PMID: 19917844 DOI: 10.1200/jco.2009.22.5060] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Children with acute lymphoblastic leukemia (ALL) are often cured, but the therapies they receive may be neurotoxic. Little is known about the incidence and severity of late-occurring neurologic sequelae in ALL survivors. Data were analyzed to determine the incidence of adverse long-term neurologic outcomes and treatment-related risk factors. PATIENTS AND METHODS We analyzed adverse neurologic outcomes that occurred after diagnosis in 4,151 adult survivors of childhood ALL who participated in the Childhood Cancer Survivor Study (CCSS), a retrospective cohort of 5-year survivors of childhood cancer diagnosed between 1970 and 1986. A randomly selected cohort of the survivors' siblings served as a comparison group. Self-reported auditory-vestibular-visual sensory deficits, focal neurologic dysfunction, seizures, and serious headaches were assessed. RESULTS The median age at outcome assessment was 20.2 years for survivors. The median follow-up time to death or last survey since ALL diagnosis was 14.1 years. Of the survivors, 64.5% received cranial radiation and 94% received intrathecal chemotherapy. Compared with the sibling cohort, survivors were at elevated risk for late-onset auditory-vestibular-visual sensory deficits (rate ratio [RR], 1.8; 95% CI, 1.5 to 2.2), coordination problems (RR, 4.1; 95% CI, 3.1 to 5.3), motor problems (RR, 5.0; 95% CI, 3.8 to 6.7), seizures (RR, 4.6; 95% CI, 3.4 to 6.2), and headaches (RR, 1.6; 95% CI, 1.4 to 1.7). In multivariable analysis, relapse was the most influential factor that increased risk of late neurologic complications. CONCLUSION Children treated with regimens that include cranial radiation for ALL and those who suffer a relapse are at increased risk for late-onset neurologic sequelae.
Collapse
Affiliation(s)
- Robert E Goldsby
- UCSF Pediatric Oncology, 505 Parnassus Ave, Box 0106, San Francisco, CA 94143-0106, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
963
|
Kim M, Yim SH, Cho NS, Kang SH, Ko DH, Oh B, Kim TY, Min HJ, She CJ, Kang HJ, Shin HY, Ahn HS, Yoon SS, Kim BK, Shin HR, Han KS, Cho HI, Lee DS. Homozygous deletion of CDKN2A (p16, p14) and CDKN2B (p15) genes is a poor prognostic factor in adult but not in childhood B-lineage acute lymphoblastic leukemia: a comparative deletion and hypermethylation study. ACTA ACUST UNITED AC 2009; 195:59-65. [PMID: 19837270 DOI: 10.1016/j.cancergencyto.2009.06.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 06/11/2009] [Indexed: 12/01/2022]
Abstract
The biological behavior of childhood B-lineage acute lymphoblastic leukemia (B-ALL) is different from that of adults. We performed a comprehensive analysis of the deletion and the methylation profile of CDKN2A (hereafter identified separately as p16 and p14, for the different proteins encoded) and CDKN2B (hereafter p15) in 91 newly diagnosed B-ALL patients (61 children, 30 adults). The prognostic significance of the profiles of these genes and the association between alterations in these genes and known cytogenetic prognostic factors (BCR/ABL; ETV6/RUNX1, formerly TEL/AML1; MLL rearrangement; and ploidy changes of chromosomes) were also assessed. The prevalence of homozygous deletion, hemizygous deletion, and no deletion of the 9p21 region was 11.5%, 16.4%, and 72.1%, respectively, in children and 30.0%, 20.0%, and 50.0%, respectively, in adults; the higher incidence of homozygous deletion in adults was significant (P=0.029). Homozygous deletion was associated with poor overall survival in adults (P=0.019), but not in children. The incidence of promoter methylation of p16, p14, and p15 was 34.4%, 14.8%, and 34.4%, respectively, in children and 26.7%, 10.0%, and 40.0%, respectively, in adults, with no significant difference between the two groups. No significant association was observed between deletion and methylation or with known cytogenetic prognostic factors. The difference in incidence, distribution, and prognostic effect of homozygous deletion in children and adults may explain the prognostic disparity.
Collapse
Affiliation(s)
- Miyoung Kim
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Chongro-gu, Seoul 110-744, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
964
|
Xavier AC, Ge Y, Taub JW. Down syndrome and malignancies: a unique clinical relationship: a paper from the 2008 william beaumont hospital symposium on molecular pathology. J Mol Diagn 2009; 11:371-80. [PMID: 19710397 DOI: 10.2353/jmoldx.2009.080132] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The patterns of malignancies in Down syndrome (DS) are unique and highlight the relationship between chromosome 21 and cancer. DS children have a approximately 10- to 20-fold higher risk for developing acute lymphoblastic leukemia and acute myeloid leukemia (AML), as compared with non-DS children, although they do not have a uniformly increased risk of developing solid tumors. DS children with acute lymphoblastic leukemia frequently experience higher levels of treatment-related toxicity and inferior event-free survival rates, as compared with non-DS children. DS children also develop AML with unique features and have a 500-fold increased risk of developing the AML subtype, acute megakaryocytic leukemia (AMkL; M7). Nearly 10% of DS newborns are diagnosed with a variant of AMkL, the transient myeloproliferative disorder, which can resolve spontaneously without treatment; event-free survival rates for DS patients with AMkL ranges from 80% to 100%, in comparison with <30% for non-DS children with AMkL. In addition, somatic mutations of the GATA1 gene have been detected in nearly all DS TMD and AMkL cases and not in leukemia cases in non-DS children. GATA1 mutations are key factors linked to both leukemogenesis and the high cure rates of DS AMkL patients. Identifying the mechanisms that account for the high event-free survival rates of DS AMkL patients may ultimately improve AML treatment as well. Examining leukemogenesis in DS children may identify factors linked to the general development of childhood leukemia and lead to potential new therapeutic strategies to fight this disease.
Collapse
Affiliation(s)
- Ana C Xavier
- Division of Hematology/Oncology, Children's Hospital of Michigan, Department of Pediatrics, Wayne State University School of Medicine, 3901 Beaubien Blvd., Detroit, Michigan 48201, USA
| | | | | |
Collapse
|
965
|
Gene expression classifiers for relapse-free survival and minimal residual disease improve risk classification and outcome prediction in pediatric B-precursor acute lymphoblastic leukemia. Blood 2009; 115:1394-405. [PMID: 19880498 DOI: 10.1182/blood-2009-05-218560] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
To determine whether gene expression profiling could improve outcome prediction in children with acute lymphoblastic leukemia (ALL) at high risk for relapse, we profiled pretreatment leukemic cells in 207 uniformly treated children with high-risk B-precursor ALL. A 38-gene expression classifier predictive of relapse-free survival (RFS) could distinguish 2 groups with differing relapse risks: low (4-year RFS, 81%, n = 109) versus high (4-year RFS, 50%, n = 98; P < .001). In multivariate analysis, the gene expression classifier (P = .001) and flow cytometric measures of minimal residual disease (MRD; P = .001) each provided independent prognostic information. Together, they could be used to classify children with high-risk ALL into low- (87% RFS), intermediate- (62% RFS), or high- (29% RFS) risk groups (P < .001). A 21-gene expression classifier predictive of end-induction MRD effectively substituted for flow MRD, yielding a combined classifier that could distinguish these 3 risk groups at diagnosis (P < .001). These classifiers were further validated on an independent high-risk ALL cohort (P = .006) and retainedindependent prognostic significance (P < .001) in the presence of other recently described poor prognostic factors (IKAROS/IKZF1 deletions, JAK mutations, and kinase expression signatures). Thus, gene expression classifiers improve ALL risk classification and allow prospective identification of children who respond or fail current treatment regimens. These trials were registered at http://clinicaltrials.gov under NCT00005603.
Collapse
|
966
|
Specific promoter methylation identifies different subgroups of MLL-rearranged infant acute lymphoblastic leukemia, influences clinical outcome, and provides therapeutic options. Blood 2009; 114:5490-8. [PMID: 19855078 DOI: 10.1182/blood-2009-06-227660] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MLL-rearranged infant acute lymphoblastic leukemia (ALL) remains the most aggressive type of childhood leukemia, displaying a unique gene expression profile. Here we hypothesized that this characteristic gene expression signature may have been established by potentially reversible epigenetic modifications. To test this hypothesis, we used differential methylation hybridization to explore the DNA methylation patterns underlying MLL-rearranged ALL in infants. The obtained results were correlated with gene expression data to confirm gene silencing as a result of promoter hypermethylation. Distinct promoter CpG island methylation patterns separated different genetic subtypes of MLL-rearranged ALL in infants. MLL translocations t(4;11) and t(11;19) characterized extensively hypermethylated leukemias, whereas t(9;11)-positive infant ALL and infant ALL carrying wild-type MLL genes epigenetically resembled normal bone marrow. Furthermore, the degree of promoter hypermethylation among infant ALL patients carrying t(4;11) or t(11;19) appeared to influence relapse-free survival, with patients displaying accentuated methylation being at high relapse risk. Finally, we show that the demethylating agent zebularine reverses aberrant DNA methylation and effectively induces apoptosis in MLL-rearranged ALL cells. Collectively these data suggest that aberrant DNA methylation occurs in the majority of MLL-rearranged infant ALL cases and guides clinical outcome. Therefore, inhibition of aberrant DNA methylation may be an important novel therapeutic strategy for MLL-rearranged ALL in infants.
Collapse
|
967
|
Rearrangement of CRLF2 in B-progenitor- and Down syndrome-associated acute lymphoblastic leukemia. Nat Genet 2009; 41:1243-6. [PMID: 19838194 DOI: 10.1038/ng.469] [Citation(s) in RCA: 434] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 09/18/2009] [Indexed: 12/16/2022]
Abstract
Aneuploidy and translocations are hallmarks of B-progenitor acute lymphoblastic leukemia (ALL), but many individuals with this cancer lack recurring chromosomal alterations. Here we report a recurring interstitial deletion of the pseudoautosomal region 1 of chromosomes X and Y in B-progenitor ALL that juxtaposes the first, noncoding exon of P2RY8 with the coding region of CRLF2. We identified the P2RY8-CRLF2 fusion in 7% of individuals with B-progenitor ALL and 53% of individuals with ALL associated with Down syndrome. CRLF2 alteration was associated with activating JAK mutations, and expression of human P2RY8-CRLF2 together with mutated mouse Jak2 resulted in constitutive Jak-Stat activation and cytokine-independent growth of Ba/F3 cells overexpressing interleukin-7 receptor alpha. Our findings indicate that these two genetic lesions together contribute to leukemogenesis in B-progenitor ALL.
Collapse
|
968
|
Carlos Jaime-Pérez J, Gómez-Almaguer D, Sandoval-González A, Chapa-Rodríguez A, Gonzàlez-Llano O. Random serum methotrexate determinations for assessing compliance with maintenance therapy for childhood acute lymphoblastic leukemia. Leuk Lymphoma 2009; 50:1843-7. [DOI: 10.3109/10428190903216812] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
969
|
|
970
|
Litzow MR. Therapy of Philadelphia chromosome-negative acute lymphoblastic leukemia in adults: new paradigms. Future Oncol 2009; 5:1039-50. [PMID: 19792972 DOI: 10.2217/fon.09.74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although the outcomes for adults with acute lymphoblastic leukemia (ALL) lag behind the stunningly successful results seen in children, new paradigms and new discoveries bring hope that this disparity will steadily lessen. The adoption of the use of pediatric intensity-type regimens in adolescents and young adults show promise in improving outcomes in this population. Recent donor-versus-no-donor comparisons in the allogeneic transplant setting highlight a potent graft-versus-leukemia effect in ALL, and the application of reduced intensity conditioning transplants may exploit this effect while reducing nonrelapse mortality. New therapeutic targets, such as CD22 in precusor B-cell ALL and mutations in NOTCH1 in T-cell ALL, are being exploited in clinical trials. Finally, use of molecular techniques and flow cytometry to quantitate minimal residual disease will allow further stratifications of patients by risk, identification of new therapeutic targets and will lessen drug toxicity through the use of pharmacogenomics.
Collapse
Affiliation(s)
- Mark R Litzow
- Division of Hematology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
971
|
Looking Toward the Future: Novel Strategies Based on Molecular Pathogenesis of Acute Lymphoblastic Leukemia. Hematol Oncol Clin North Am 2009; 23:1099-119, vii. [DOI: 10.1016/j.hoc.2009.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
972
|
Nathan PC, Wasilewski-Masker K, Janzen LA. Long-term Outcomes in Survivors of Childhood Acute Lymphoblastic Leukemia. Hematol Oncol Clin North Am 2009; 23:1065-82, vi-vii. [DOI: 10.1016/j.hoc.2009.07.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
973
|
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]
|
974
|
Abujamra AL, Dos Santos MP, Roesler R, Schwartsmann G, Brunetto AL. Histone deacetylase inhibitors: a new perspective for the treatment of leukemia. Leuk Res 2009; 34:687-95. [PMID: 19762081 DOI: 10.1016/j.leukres.2009.08.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 07/15/2009] [Accepted: 08/17/2009] [Indexed: 11/16/2022]
Abstract
Histone deacetylase inhibitors (HDIs) promote or enhance several different anticancer mechanisms and therefore are in evidence as potential antileukemia agents. Studies on leukemia have provided examples for their functional implications in cancer development and progression, as well as their relevance for therapeutic targeting. A number of HDIs have been tested in clinical trials and have been proven safe with significant clinical activity. The use of HDIs in association with other molecules, such as classical chemotherapeutic drugs and DNA demethylating agents, has been implied as a promising treatment alternative for leukemia patients in the future. Here we describe the histone deacetylase inhibitors that have been tested in clinical trials for the treatment of leukemia and lymphoma. We conclude that further clinical trials involving a broader number of HDIs used either alone or in combination with other agents are needed to consolidate the use of these epigenetic modulators on leukemia therapy.
Collapse
Affiliation(s)
- Ana Lucia Abujamra
- Children's Cancer Institute and Pediatric Oncology Unit, Cancer Research Laboratory, Academic Hospital, Federal University of Rio Grande do Sul, and National Institute for Translational Medicine (INCT Program), Porto Alegre, Brazil.
| | | | | | | | | |
Collapse
|
975
|
Abstract
Frequent hallmarks of T-cell acute lymphoblastic leukemia (T-ALL) include aberrant NOTCH signaling and deletion of the CDKN2A locus, which contains 2 closely linked tumor suppressor genes (INK4A and ARF). When bone marrow cells or thymocytes transduced with a vector encoding the constitutively activated intracellular domain of Notch1 (ICN1) are expanded ex vivo under conditions that support T-cell development, cultured progenitors rapidly induce CD4+/CD8+ T-ALLs after infusion into healthy syngeneic mice. Under these conditions, enforced ICN1 expression also drives formation of T-ALLs in unconditioned CD-1 nude mice, bypassing any requirements for thymic maturation. Retention of Arf had relatively modest activity in suppressing the formation of T-ALLs arising from bone marrow-derived ICN1+ progenitors in which the locus is epigenetically silenced, and all resulting Arf (+/+) tumors failed to express the p19(Arf) protein. In striking contrast, retention of Arf in thymocyte-derived ICN1+ donor cells significantly delayed disease onset and suppressed the penetrance of T-ALL. Use of cultured thymocyte-derived donor cells expressing a functionally null Arf-GFP knock-in allele confirmed that ICN1 signaling can induce Arf expression in vivo. Arf activation by ICN1 in T cells thereby provides stage-specific tumor suppression but also a strong selective pressure for deletion of the locus in T-ALL.
Collapse
|
976
|
Zhang H, Yang JH, Zheng YS, Zhang P, Chen X, Wu J, Xu L, Luo XQ, Ke ZY, Zhou H, Qu LH, Chen YQ. Genome-wide analysis of small RNA and novel MicroRNA discovery in human acute lymphoblastic leukemia based on extensive sequencing approach. PLoS One 2009; 4:e6849. [PMID: 19724645 PMCID: PMC2731166 DOI: 10.1371/journal.pone.0006849] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2009] [Accepted: 08/03/2009] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) have been proved to play an important role in various cellular processes and function as tumor suppressors or oncogenes in cancers including leukemia. The identification of a large number of novel miRNAs and other small regulatory RNAs will provide valuable insights into the roles they play in tumorgenesis. METHODOLOGY/PRINCIPAL FINDINGS To gain further understanding of the role of miRNAs relevant to acute lymphoblastic leukemia (ALL), we employed the sequencing-by-synthesis (SBS) strategy to sequence small RNA libraries prepared from ALL patients and normal donors. In total we identified 159 novel miRNAs and 116 novel miRNA*s from both libraries. Among the 159 novel miRNAs, 42 were identified with high stringency in our data set. Furthermore, we demonstrated the different expression patterns of 20 newly identified and several known miRNAs between ALL patients and normal donors, suggesting these miRNAs may be associated with ALL and could constitute an ALL-specific miRNA signature. Interestingly, GO "biological process" classifications revealed that a set of significantly abnormally expressed miRNAs are associated with disease relapse, which implies that these dysregulated miRNAs might promote the progression of ALL by regulating genes involved in the pathway of the disease development. CONCLUSION/SIGNIFICANCE The study presents a comprehensive picture of the expression of small RNAs in human acute lymphoblastic leukemia and highlights novel and known miRNAs differentially expressed between ALL patients and normal donors. To our knowledge, this is the first study to look at genome-wide known and novel miRNA expression patterns in in human acute lymphoblastic leukemia. Our data revealed that these deregulated miRNAs may be associated with ALL or the onset of relapse.
Collapse
Affiliation(s)
- Hua Zhang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jian-Hua Yang
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yu-Sheng Zheng
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Peng Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Xiao Chen
- The Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jun Wu
- The Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Ling Xu
- The Second Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xue-Qun Luo
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhi-Yong Ke
- The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hui Zhou
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Liang-Hu Qu
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yue-Qin Chen
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
977
|
Abstract
Cancer remains an outstanding cause of global morbidity and mortality, despite intensive research and unprecedented insights into the basic mechanisms of cancer development. A plethora of clinical and experimental evidence suggests that cancers from individual patients are likely to be molecularly heterogeneous in their use of distinct oncogenic pathways and biological programs. Efforts to significantly impact cancer patient outcomes will almost certainly require the development of robust strategies to subdivide such heterogeneous panels of cancers into biologically and clinically homogenous subgroups, for the purposes of personalizing treatment protocols and identifying optimal drug targets. In this review, I describe recent progress in the development of both targeted and genome-wide approaches for the molecular stratification of cancers, drawing examples from both the haematopoietic and solid tumor malignancies.
Collapse
Affiliation(s)
- Patrick Tan
- Duke-NUS Graduate Medical School, Genome Institute of Singapore, Singapore; Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
978
|
Schindler JW, Van Buren D, Foudi A, Krejci O, Qin J, Orkin SH, Hock H. TEL-AML1 corrupts hematopoietic stem cells to persist in the bone marrow and initiate leukemia. Cell Stem Cell 2009; 5:43-53. [PMID: 19570513 DOI: 10.1016/j.stem.2009.04.019] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 03/16/2009] [Accepted: 04/29/2009] [Indexed: 10/20/2022]
Abstract
The initial steps in the pathogenesis of acute leukemia remain incompletely understood. The TEL-AML1 gene fusion, the hallmark translocation in Childhood Acute Lymphoblastic Leukemia and the first hit, occurs years before the clinical disease, most often in utero. We have generated mice in which TEL-AML1 expression is driven from the endogenous promoter and can be targeted to specific populations. TEL-AML1 renders mice prone to malignancy after chemical mutagenesis when expressed in hematopoietic stem cells (HSCs), but not in early lymphoid progenitors. We reveal that TEL-AML1 markedly increases the number of HSCs and predominantly maintains them in the quiescent (G(0)) stage of the cell cycle. TEL-AML1(+) HSCs retain self-renewal properties and contribute to hematopoiesis, but fail to out-compete normal HSCs. Our work shows that stem cells are susceptible to subversion by weak oncogenes that can subtly alter their molecular program to provide a latent reservoir for the accumulation of further mutations.
Collapse
|
979
|
Semsei AF, Antal P, Szalai C. Strengths and weaknesses of gene association studies in childhood acute lymphoblastic leukemia. Leuk Res 2009; 34:269-71. [PMID: 19716175 DOI: 10.1016/j.leukres.2009.07.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 07/22/2009] [Accepted: 07/23/2009] [Indexed: 11/27/2022]
|
980
|
Elevated mRNA level of hST6Gal I and hST3Gal V positively correlates with the high risk of pediatric acute leukemia. Leuk Res 2009; 34:463-70. [PMID: 19709745 DOI: 10.1016/j.leukres.2009.07.042] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2009] [Revised: 07/25/2009] [Accepted: 07/27/2009] [Indexed: 11/22/2022]
Abstract
Altered sialylation occurs in essentially all types of human and experimental cancers. Although, aberrant sialylation is believed to mainly due to altered sialyltransferase (ST) level, so far, expression pattern of different STs in acute lymphoblastic leukemia has never been investigated. Accordingly, the aim of our study was to monitor the changes in mRNA expression of ST6Gal I, ST3Gal V and ST8Sia I in patients by real-time PCR, which may provide prognostic information useful in defining appropriate therapeutic options. Our data demonstrated that ST6Gal I and ST3Gal V mRNA were up-regulated in lymphoblasts whereas its presence was negligible in non-malignant donors. In contrast, ST8SiaI was downregulated in patients. The extents of linkage-specific sialylation of glycoconjugates were found to be associated with disease establishment. Additionally, ST6Gal I and ST3Gal V were positively correlated with the high risk of the disease (P=0.0032 and 0.0016). This differential ST level can be used as biomarker with the molecular method of quantitative PCR and may be useful to discriminate normal and cancer patients.
Collapse
|
981
|
Chowdhury S, Mandal C. O-acetylated sialic acids: multifaceted role in childhood acute lymphoblastic leukaemia. Biotechnol J 2009; 4:361-74. [PMID: 19296441 DOI: 10.1002/biot.200800253] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Childhood acute lymphoblastic leukaemia (ALL), a malignant transformation of the lymphoblasts, is highly responsive to chemotherapy. However, due to certain inadequacy in detection of minimal residual disease (MRD), relapse is a common phenomenon. To address this question, the present review deals with the induction of an unique O-acetyl derivative of sialic acid on a few disease-associated glycoproteins and glycolipids at the onset of childhood ALL, a finding of our group in the last decade. This information has been successfully utilized for diagnosis and prognosis of the disease. Existing literature is included for comparison. Additionally, cell surface overexpression of 9-O-acetylated sialoglycoproteins and antibodies against them present in patients' sera aid the survival of the malignant lymphoblasts and suggest a multifaceted role played by these molecules. Taken together, monitoring these molecules helps not only in unravelling the biology of this paediatric malignancy but also in personalizing the treatment strategies for the betterment of the patient population.
Collapse
Affiliation(s)
- Suchandra Chowdhury
- Infectious Diseases and Immunology Division, Indian Institute of Chemical Biology, Kolkata, India
| | | |
Collapse
|
982
|
|
983
|
Adams BD, Baker R, Lopez JA, Spencer S. Myeloproliferative Disorders and the Hyperviscosity Syndrome. Emerg Med Clin North Am 2009; 27:459-76. [DOI: 10.1016/j.emc.2009.04.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
984
|
Copland M. Ikaros deletions are associated with poor prognosis in acute lymphoblastic leukemia. Future Oncol 2009; 5:455-8. [PMID: 19450174 DOI: 10.2217/fon.09.16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Evaluation of: Mullighan CG, Su X, Zhang J et al.: Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N. Engl. J. Med. 360, 470–480 (2009). Acute lymphoblastic leukemia (ALL) is the most common malignancy of childhood and despite impressive remission rates, approximately 20% of children with ALL relapse with disease that is often difficult to treat. Recently, genome-wide analysis has identified recurring genetic alterations in B-cell ALL, including mutation of PAX5 and deletions of IKZF1, IKZF3, E2A, EBF and LEF1. This large clinical study evaluates the prognostic impact of these genetic abnormalities in two independent cohorts of children with B-cell progenitor ALL and identifies deletion of IKZF1 (which encodes the lymphoid transcription factor Ikaros) as a predictor of poor outcome. This finding demonstrates the importance of IKZF1 status in pediatric ALL, and provides strong supporting evidence for its inclusion in pediatric ALL risk stratification for treatment protocols.
Collapse
Affiliation(s)
- Mhairi Copland
- Paul O'Gorman Leukaemia Research Centre, University of Glasgow, Gartnavel General Hospital, 1053 Great Western Road, Glasgow, G12 0YN, UK.
| |
Collapse
|
985
|
RUNX1 regulates phosphoinositide 3-kinase/AKT pathway: role in chemotherapy sensitivity in acute megakaryocytic leukemia. Blood 2009; 114:2744-52. [PMID: 19638627 DOI: 10.1182/blood-2008-09-179812] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
RUNX1 (AML1) encodes the core binding factor alpha subunit of a heterodimeric transcription factor complex which plays critical roles in normal hematopoiesis. Translocations or down-regulation of RUNX1 have been linked to favorable clinical outcomes in acute leukemias, suggesting that RUNX1 may also play critical roles in chemotherapy responses in acute leukemias; however, the molecular mechanisms remain unclear. The median level of RUNX1b transcripts in Down syndrome (DS) children with acute megakaryocytic leukemia (AMkL) were 4.4-fold (P < .001) lower than that in non-DS AMkL cases. Short hairpin RNA knockdown of RUNX1 in a non-DS AMkL cell line, Meg-01, resulted in significantly increased sensitivity to cytosine arabinoside, accompanied by significantly decreased expression of PIK3CD, which encodes the delta catalytic subunit of the survival kinase, phosphoinositide 3 (PI3)-kinase. Transcriptional regulation of PIK3CD by RUNX1 was further confirmed by chromatin immunoprecipitation and promoter reporter gene assays. Further, a PI3-kinase inhibitor, LY294002, and cytosine arabinoside synergized in antileukemia effects on Meg-01 and primary pediatric AMkL cells. Our results suggest that RUNX1 may play a critical role in chemotherapy response in AMkL by regulating the PI3-kinase/Akt pathway. Thus, the treatment of AMkL may be improved by integrating PI3-kinase or Akt inhibitors into the chemotherapy of this disease.
Collapse
|
986
|
Patel N, Krishnan S, Offman MN, Krol M, Moss CX, Leighton C, van Delft FW, Holland M, Liu J, Alexander S, Dempsey C, Ariffin H, Essink M, Eden TO, Watts C, Bates PA, Saha V. A dyad of lymphoblastic lysosomal cysteine proteases degrades the antileukemic drug L-asparaginase. J Clin Invest 2009; 119:1964-73. [PMID: 19509471 PMCID: PMC2701869 DOI: 10.1172/jci37977] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Accepted: 04/08/2009] [Indexed: 01/23/2023] Open
Abstract
l-Asparaginase is a key therapeutic agent for treatment of childhood acute lymphoblastic leukemia (ALL). There is wide individual variation in pharmacokinetics, and little is known about its metabolism. The mechanisms of therapeutic failure with l-asparaginase remain speculative. Here, we now report that 2 lysosomal cysteine proteases present in lymphoblasts are able to degrade l-asparaginase. Cathepsin B (CTSB), which is produced constitutively by normal and leukemic cells, degraded asparaginase produced by Escherichia coli (ASNase) and Erwinia chrysanthemi. Asparaginyl endopeptidase (AEP), which is overexpressed predominantly in high-risk subsets of ALL, specifically degraded ASNase. AEP thereby destroys ASNase activity and may also potentiate antigen processing, leading to allergic reactions. Using AEP-mediated cleavage sequences, we modeled the effects of the protease on ASNase and created a number of recombinant ASNase products. The N24 residue on the flexible active loop was identified as the primary AEP cleavage site. Sole modification at this site rendered ASNase resistant to AEP cleavage and suggested a key role for the flexible active loop in determining ASNase activity. We therefore propose what we believe to be a novel mechanism of drug resistance to ASNase. Our results may help to identify alternative therapeutic strategies with the potential of further improving outcome in childhood ALL.
Collapse
Affiliation(s)
- Naina Patel
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Shekhar Krishnan
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Marc N. Offman
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Marcin Krol
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Catherine X. Moss
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Carly Leighton
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Frederik W. van Delft
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Mark Holland
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - JiZhong Liu
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Seema Alexander
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Clare Dempsey
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Hany Ariffin
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Monika Essink
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Tim O.B. Eden
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Colin Watts
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Paul A. Bates
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| | - Vaskar Saha
- Cancer Research UK Children’s Cancer Group, Paterson Institute for Cancer Research, University of Manchester, Manchester, United Kingdom.
Biomolecular Modelling Laboratory, Cancer Research UK London Research Institute, Lincoln’s Inn Fields Laboratories, London, United Kingdom.
Division of Cell Biology and Immunology, School of Life Sciences Research Biocentre, University of Dundee, Dundee, United Kingdom.
Department of Paediatrics, University of Malaya, Kuala Lumpur, Malaysia.
Medac GmbH, Wedel, Germany.
Paediatric and Adolescent Oncology Unit, Christie Hospital, Manchester, United Kingdom
| |
Collapse
|
987
|
Advanced vertebral fracture among newly diagnosed children with acute lymphoblastic leukemia: results of the Canadian Steroid-Associated Osteoporosis in the Pediatric Population (STOPP) research program. J Bone Miner Res 2009; 24:1326-34. [PMID: 19210218 PMCID: PMC3890351 DOI: 10.1359/jbmr.090202] [Citation(s) in RCA: 154] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Vertebral compression is a serious complication of childhood acute lymphoblastic leukemia (ALL). The prevalence and pattern of vertebral fractures, as well as their relationship to BMD and other clinical indices, have not been systematically studied. We evaluated spine health in 186 newly diagnosed children (median age, 5.3 yr; 108 boys) with ALL (precursor B cell: N = 167; T cell: N = 19) who were enrolled in a national bone health research program. Patients were assessed within 30 days of diagnosis by lateral thoraco-lumbar spine radiograph, bone age (also used for metacarpal morphometry), and BMD. Vertebral morphometry was carried out by the Genant semiquantitative method. Twenty-nine patients (16%) had a total of 75 grade 1 or higher prevalent vertebral compression fractures (53 thoracic, 71%; 22 lumbar). Grade 1 fractures as the worst grade were present in 14 children (48%), 9 patients (31%) had grade 2 fractures, and 6 children (21%) had grade 3 fractures. The distribution of spine fracture was bimodal, with most occurring in the midthoracic and thoraco-lumbar regions. Children with grade 1 or higher vertebral compression had reduced lumbar spine (LS) areal BMD Z-scores compared with those without (mean +/- SD, -2.1 +/- 1.5 versus -1.1 +/- 1.2; p < 0.001). LS BMD Z-score, second metacarpal percent cortical area Z-score, and back pain were associated with increased odds for fracture. For every 1 SD reduction in LS BMD Z-score, the odds for fracture increased by 80% (95% CI: 10-193%); the presence of back pain had an OR of 4.7 (95% CI: 1.5-14.5). These results show that vertebral compression is an under-recognized complication of newly diagnosed ALL. Whether the fractures will resolve through bone growth during or after leukemia chemotherapy remains to be determined.
Collapse
|
988
|
Abstract
The molecular mechanisms involved in disease progression and relapse in T-cell acute lymphoblastic leukemia (T-ALL) are poorly understood. We used single nucleotide polymorphism array analysis to analyze paired diagnostic and relapsed T-ALL samples to identify recurrent genetic alterations in T-ALL. This analysis showed that diagnosis and relapsed cases have common genetic alterations, but also that relapsed samples frequently lose chromosomal markers present at diagnosis, suggesting that relapsed T-ALL emerges from an ancestral clone different from the major leukemic population at diagnosis. In addition, we identified deletions and associated mutations in the WT1 tumor suppressor gene in 2 of 9 samples. Subsequent analysis showed WT1 mutations in 28 of 211 (13.2%) of pediatric and 10 of 85 (11.7%) of adult T-ALL cases. WT1 mutations present in T-ALL are predominantly heterozygous frameshift mutations resulting in truncation of the C-terminal zinc finger domains of this transcription factor. WT1 mutations are most prominently found in T-ALL cases with aberrant rearrangements of the oncogenic TLX1, TLX3, and HOXA transcription factor oncogenes. Survival analysis demonstrated that WT1 mutations do not confer adverse prognosis in pediatric and adult T-ALL. Overall, these results identify the presence of WT1 mutations as a recurrent genetic alteration in T-ALL.
Collapse
|
989
|
Abstract
Pediatric acute lymphoblastic leukemia (ALL) is a heterogeneous disease consisting of distinct clinical and biological subtypes that are characterized by specific chromosomal abnormalities or gene mutations. Mutation of genes encoding tyrosine kinases is uncommon in ALL, with the exception of Philadelphia chromosome-positive ALL, where the t(9,22)(q34;q11) translocation encodes the constitutively active BCR-ABL1 tyrosine kinase. We recently identified a poor prognostic subgroup of pediatric BCR-ABL1-negative ALL patients characterized by deletion of IKZF1 (encoding the lymphoid transcription factor IKAROS) and a gene expression signature similar to BCR-ABL1-positive ALL, raising the possibility of activated tyrosine kinase signaling within this leukemia subtype. Here, we report activating mutations in the Janus kinases JAK1 (n = 3), JAK2 (n = 16), and JAK3 (n = 1) in 20 (10.7%) of 187 BCR-ABL1-negative, high-risk pediatric ALL cases. The JAK1 and JAK2 mutations involved highly conserved residues in the kinase and pseudokinase domains and resulted in constitutive JAK-STAT activation and growth factor independence of Ba/F3-EpoR cells. The presence of JAK mutations was significantly associated with alteration of IKZF1 (70% of all JAK-mutated cases and 87.5% of cases with JAK2 mutations; P = 0.001) and deletion of CDKN2A/B (70% of all JAK-mutated cases and 68.9% of JAK2-mutated cases). The JAK-mutated cases had a gene expression signature similar to BCR-ABL1 pediatric ALL, and they had a poor outcome. These results suggest that inhibition of JAK signaling is a logical target for therapeutic intervention in JAK mutated ALL.
Collapse
|
990
|
Le Y, Xu L, Lu J, Fang J, Nardi V, Chai L, Silberstein LE. FAK silencing inhibits leukemogenesis in BCR/ABL-transformed hematopoietic cells. Am J Hematol 2009; 84:273-8. [PMID: 19358301 DOI: 10.1002/ajh.21381] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Focal adhesion kinase (FAK) is constitutively activated and tyrosine phosphorylated in BCR/ABL-transformed hematopoietic cells, but the role it plays during leukemogenesis remains unclear. Here, we examined the effects of RNA interference-mediated FAK silencing on leukemogenesis induced by a BCR/ABL-transformed cell line. Transduction of BCR/ABL-BaF3 cells with FAK shRNA inhibited FAK expression and reduced STAT5 phosphorylation, but induced caspase-3 activation. In vitro studies showed that treatment with FAK shRNA resulted in impaired cell proliferation and colony formation, while increasing cell apoptosis. Mice that received transplants of BCR/ABL-BaF3 cells with FAK shRNA displayed significantly prolonged survival time and diminished leukemia progression. In addition, FAK silencing enhanced in vitro and in vivo efficacy of ABL tyrosine kinase inhibitor imatinib in BCR/ABL-BaF3 cells. Our results suggest that FAK is critical for leukemogenesis and might be a potential target for leukemia therapy.
Collapse
Affiliation(s)
- Yi Le
- Department of Pathology, Joint Program in Transfusion Medicine, Children's Hospital Boston, Karp Research Building, Room 10217, One Blackfan Circle, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
991
|
Verstovsek S. Preclinical and clinical experience with dasatinib in Philadelphia chromosome-negative leukemias and myeloid disorders. Leuk Res 2009; 33:617-23. [PMID: 19013641 PMCID: PMC4428150 DOI: 10.1016/j.leukres.2008.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Revised: 10/02/2008] [Accepted: 10/02/2008] [Indexed: 12/29/2022]
Abstract
Recent advances in the molecular characterization of Philadelphia chromosome-negative (Ph-) leukemias and related myeloid disorders have provided a clear rationale for investigating novel targeted therapies. Dasatinib is a tyrosine kinase inhibitor with activity against BCR-ABL, platelet-derived growth factor receptors (PDGFRs), c- KIT, fibroblast growth factor receptors (FGFRs), SRC family kinases (SFKs), and EPHA receptors, all of which have been implicated in the pathogenesis of Ph- leukemias and myeloid disorders. This review presents emerging data on the preclinical and clinical activity of dasatinib in these diseases, which suggest that larger clinical studies are warranted.
Collapse
Affiliation(s)
- Srdan Verstovsek
- Leukemia Department, M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 428, Houston, TX 77030, USA.
| |
Collapse
|
992
|
Weerkamp F, Dekking E, Ng YY, van der Velden VHJ, Wai H, Böttcher S, Brüggemann M, van der Sluijs AJ, Koning A, Boeckx N, Van Poecke N, Lucio P, Mendonça A, Sedek L, Szczepański T, Kalina T, Kovac M, Hoogeveen PG, Flores-Montero J, Orfao A, Macintyre E, Lhermitte L, Chen R, Brouwer-De Cock KAJ, van der Linden A, Noordijk AL, Comans-Bitter WM, Staal FJT, van Dongen JJM. Flow cytometric immunobead assay for the detection of BCR-ABL fusion proteins in leukemia patients. Leukemia 2009; 23:1106-17. [PMID: 19387467 DOI: 10.1038/leu.2009.93] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BCR-ABL fusion proteins show increased signaling through their ABL tyrosine kinase domain, which can be blocked by specific inhibitors, thereby providing effective treatment. This makes detection of BCR-ABL aberrations of utmost importance for diagnosis, classification and treatment of leukemia patients. BCR-ABL aberrations are currently detected by karyotyping, fluorescence in situ hybridization (FISH) or PCR techniques, which are time consuming and require specialized facilities. We developed a simple flow cytometric immunobead assay for detection of BCR-ABL fusion proteins in cell lysates, using a bead-bound anti-BCR catching antibody and a fluorochrome-conjugated anti-ABL detection antibody. We noticed protein stability problems in lysates caused by proteases from mature myeloid cells. This problem could largely be solved by adding protease inhibitors in several steps of the immunobead assay. Testing of 145 patient samples showed fully concordant results between the BCR-ABL immunobead assay and reverse transcriptase PCR of fusion gene transcripts. Dilution experiments with BCR-ABL positive cell lines revealed sensitivities of at least 1%. We conclude that the BCR-ABL immunobead assay detects all types of BCR-ABL proteins in leukemic cells with high specificity and sensitivity. The assay does not need specialized laboratory facilities other than a flow cytometer, provides results within approximately 4 h, and can be run in parallel to routine immunophenotyping.
Collapse
Affiliation(s)
- F Weerkamp
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
993
|
Chiarini F, Falà F, Tazzari PL, Ricci F, Astolfi A, Pession A, Pagliaro P, McCubrey JA, Martelli AM. Dual inhibition of class IA phosphatidylinositol 3-kinase and mammalian target of rapamycin as a new therapeutic option for T-cell acute lymphoblastic leukemia. Cancer Res 2009; 69:3520-8. [PMID: 19351820 PMCID: PMC3836286 DOI: 10.1158/0008-5472.can-08-4884] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Recent investigations have documented that constitutively activated phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling is a common feature of T-cell acute lymphoblastic leukemia (T-ALL), where it strongly influences growth and survival. These findings lend compelling weight for the application of PI3K/Akt/mTOR inhibitors in T-ALL. However, our knowledge of PI3K/Akt/mTOR signaling in T-ALL is limited and it is not clear whether it could be an effective target for innovative therapeutic strategies. Here, we have analyzed the therapeutic potential of the dual PI3K/mTOR inhibitor PI-103, a small synthetic molecule of the pyridofuropyrimidine class, on both T-ALL cell lines and patient samples, which displayed constitutive activation of PI3K/Akt/mTOR signaling. PI-103 inhibited the growth of T-ALL cells, including 170-kDa P-glycoprotein overexpressing cells. PI-103 cytotoxicity was independent of p53 gene status. PI-103 was more potent than inhibitors that are selective only for PI3K (Wortmannin, LY294002) or for mTOR (rapamycin). PI-103 induced G(0)-G(1) phase cell cycle arrest and apoptosis, which was characterized by activation of caspase-3 and caspase-9. PI-103 caused Akt dephosphorylation, accompanied by dephosphorylation of the Akt downstream target, glycogen synthase kinase-3beta. Also, mTOR downstream targets were dephosphorylated in response to PI-103, including p70S6 kinase, ribosomal S6 protein, and 4E-BP1. PI-103 strongly synergized with vincristine. These findings indicate that multitargeted therapy toward PI3K and mTOR alone or with existing drugs may serve as an efficient treatment toward T-ALL cells, which require up-regulation of PI3K/Akt/mTOR signaling for their survival and growth.
Collapse
Affiliation(s)
- Francesca Chiarini
- Department of Human Anatomical Sciences University of Bologna, Bologna, Italy
| | - Federica Falà
- Department of Human Anatomical Sciences University of Bologna, Bologna, Italy
| | - Pier Luigi Tazzari
- Immunohaematology and Transfusion Center, Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Francesca Ricci
- Immunohaematology and Transfusion Center, Policlinico S.Orsola-Malpighi, Bologna, Italy
| | - Annalisa Astolfi
- Pediatric Oncology and Haematology Unit, University of Bologna, Bologna, Italy
| | - Andrea Pession
- Pediatric Oncology and Haematology Unit, University of Bologna, Bologna, Italy
| | | | - James A. McCubrey
- Department of Microbiology & Immunology, School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Alberto M. Martelli
- Department of Human Anatomical Sciences University of Bologna, Bologna, Italy
- IGM-CNR, Sezione di Bologna c/o I.O.R., Bologna, Italy
| |
Collapse
|
994
|
The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 2009; 114:937-51. [PMID: 19357394 DOI: 10.1182/blood-2009-03-209262] [Citation(s) in RCA: 3101] [Impact Index Per Article: 206.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recently the World Health Organization (WHO), in collaboration with the European Association for Haematopathology and the Society for Hematopathology, published a revised and updated edition of the WHO Classification of Tumors of the Hematopoietic and Lymphoid Tissues. The 4th edition of the WHO classification incorporates new information that has emerged from scientific and clinical studies in the interval since the publication of the 3rd edition in 2001, and includes new criteria for the recognition of some previously described neoplasms as well as clarification and refinement of the defining criteria for others. It also adds entities-some defined principally by genetic features-that have only recently been characterized. In this paper, the classification of myeloid neoplasms and acute leukemia is highlighted with the aim of familiarizing hematologists, clinical scientists, and hematopathologists not only with the major changes in the classification but also with the rationale for those changes.
Collapse
|
995
|
Lehrnbecher T, Creutzig U. Myeloid growth factors as anti-infective measures in children with leukemia and lymphoma. Expert Rev Hematol 2009; 2:159-72. [DOI: 10.1586/ehm.09.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
996
|
Lee JA, Kim MS, Kim DH, Lim JS, Park KD, Song WS, Cho WH, Lee SY, Jeon DG. Risk stratification based on the clinical factors at diagnosis is closely related to the survival of localized osteosarcoma. Pediatr Blood Cancer 2009; 52:340-5. [PMID: 19021220 DOI: 10.1002/pbc.21843] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Survival of osteosarcoma patients has reached a plateau with the addition of chemotherapy and in part predicted based on histologic response. Risk-adapted therapy might be an alternative approach. We aimed to identify risk groups using clinical variables available at time of diagnosis in order to better predict outcomes and form the basis for risk-adapted therapy. METHODS This retrospective study analyzed 288 patients with high-grade osteosarcoma of their extremities. Clinicopathologic variables were analyzed to identify factors relevant to risk stratification. RESULTS A risk assessment system was developed using age, maximal tumor length (MTL) and tumor location. Index scores were based on the sum total of factor scores, for each of the following: Age (years); "1" for >or=40 or 12-14 (female) or 13-15 (male), "0" for other ages; MTL (cm); "2" for >or=8, "1" for 6-8, "0" for <or=6; Tumor location; "1" for humerus, "0" for elsewhere. Index score 0 or 1 was assigned as low-risk, 2 as intermediate-risk, and 3 or 4 as high-risk. Of the 288 patients, 98 (34.0%) were designated as low-risk, 128 (44.4%) as intermediate-risk and 62 (21.6%) as high-risk. Risk group was related to histologic response and survival. While 63.3% of low-risk patients were good responders, only 43.0% and 33.9% of intermediate- and high-risk patients were good responders. Ten-year event-free survival (EFS) was 81.6 +/- 3.9% for low-risk group, but 31.6 +/- 6.0% for high-risk group. CONCLUSION We defined three risk groups that could be used as basis of risk-adapted therapy for osteosarcoma.
Collapse
Affiliation(s)
- Jun Ah Lee
- Department of Pediatrics, Korea Cancer Center Hospital, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
997
|
Genome-wide profiling of genetic alterations in acute lymphoblastic leukemia: recent insights and future directions. Leukemia 2009; 23:1209-18. [DOI: 10.1038/leu.2009.18] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
998
|
Affiliation(s)
- Ching-Hon Pui
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
999
|
Abstract
A key goal in cancer research is to identify the total complement of genetic and epigenetic alterations that contribute to tumorigenesis. We are currently witnessing the rapid evolution and convergence of multiple genome-wide platforms that are making this goal a reality. Leading this effort are studies of the molecular lesions that underlie pediatric acute lymphoblastic leukemia (ALL). The recent application of microarray-based analyses of DNA copy number abnormalities (CNAs) in pediatric ALL, complemented by transcriptional profiling, resequencing and epigenetic approaches, has identified a high frequency of common genetic alterations in both B-progenitor and T-lineage ALL. These approaches have identified abnormalities in key pathways, including lymphoid differentiation, cell cycle regulation, tumor suppression, and drug responsiveness. Moreover, the nature and frequency of CNAs differ markedly among ALL genetic subtypes. In this article, we review the key findings from the published data on genome-wide analyses of ALL and highlight some of the technical aspects of data generation and analysis that must be carefully controlled to obtain optimal results.
Collapse
|
1000
|
Reilly KM, Kisor DF. Profile of nelarabine: use in the treatment of T-cell acute lymphoblastic leukemia. Onco Targets Ther 2009; 2:219-28. [PMID: 20616909 PMCID: PMC2886323 DOI: 10.2147/ott.s4770] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Indexed: 11/23/2022] Open
Abstract
Nelarabine is the prodrug of 9-beta-arabinofuranosylguanine (ara-G) and is therapeutically classified as a purine nucleoside analog. Nelarabine is converted to ara-G by adenosine deaminase and transported into cells by a nucleoside transporter. Ara-G is subsequently phosphorylated to ara-G triphosphate (ara-GTP), thereby initiating the therapeutic effect by inhibiting DNA synthesis. Nelarabine has been extensively studied in regards to its pharmacokinetics, and the data have demonstrated that ara-GTP preferentially accumulates in malignant T-cells. Clinical responses to nelarabine have been demonstrated in various T-cell malignancies and appear to correlate with a relatively high intracellular concentration of ara-GTP compared to nonresponders. Therefore, this unique drug feature of nelarabine accounts for clinical utilization in treating adult and pediatric patients with relapsed or refractory T-cell acute lymphoblastic leukemia or T-cell lymphoblastic lymphoma. Neuropathy is the most predominant adverse effect associated with nelarabine and the incidence correlates with the dose administered. Myelosuppression has been observed, with thrombocytopenia and neutropenia as the most common hematologic complications. This article reviews the pharmacology, mechanism of action, and pharmacokinetic properties of nelarabine, as well as nelarabine's clinical efficacy in T-ALL, T-LBL, and other hematologic malignancies. The toxicity profile, dosage, and administration, and areas of ongoing and future research, are also presented.
Collapse
Affiliation(s)
| | - David F Kisor
- Department of Pharmaceutical and Biomedical Sciences, Raabe College of Pharmacy, Ohio Northern University, Ada, Ohio, USA
| |
Collapse
|