Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-05-16T17:54:34.568Z Has data issue: false hasContentIssue false

71 - Acute lymphoblastic leukemia (ALL)

from Part 3.6 - Molecular pathology: lymphoma and leukemia

Published online by Cambridge University Press:  05 February 2015

Ido Paz-Priel
Affiliation:
Departments of Oncology and Pediatrics, Division of Pediatric Oncology, Johns Hopkins University, Baltimore, MD, USA
Alan D. Friedman
Affiliation:
Departments of Oncology and Pediatrics, Division of Pediatric Oncology, Johns Hopkins University, Baltimore, MD, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

It is estimated that 5760 new cases of acute lymphoblastic leukemia (ALL) will be diagnosed in 2013 (including 3509 children younger than 20 years), and 1400 patients will die from this disease(1). Genetically, ALL is one of the better-characterized malignancies, and common recurrent abnormalities are recognized and integrated into the risk stratification.

BCR–ABL1

Approximately 20–30% of adult cases of ALL are associated with the reciprocal translocation t(9;22)(q34,q11), or Philadelphia chromosome (Ph; 2,3) This translocation results in a head-to-tail fusion of the ABL1 proto-oncogene on chromosome 9 to the 5ʹ half of the breakpoint cluster region (BCR) from chromosome 22. The resulting chimeric BCR–ABL1 gene is expressed as a 210 or 190 kDa protein. The p190 product is common in ALL, while the p210 is characteristic of chronic myeloid leukemia (4). The N-terminus of the ABL1 kinase is the “Cap” region, which binds to the kinase domain and keeps the Src-homology-2 (SH2) and SH3 autoinhibitory structure in place, thus displacing catalytic residues from the active site and diminishing ATP accessibility (i.e. “off state”; 5,6). In contrast, the chimeric BCR–ABL1 protein is a constitutive kinase that activates the RAS, PI3K/AKT, MEK/ERK, and c-Jun/N-terminal kinase (JNK) pathways, and up-regulates the anti-apoptotic Bcl-2 and Bcl-X, overall resulting in enhanced cell proliferation and survival (7–10).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 777 - 785
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

American Cancer Society. Cancer Facts and Figures 2009. Atlanta, GA: American Cancer Society, 2009. .Google Scholar
Pui, C-H, Relling, MV, Downing, JR. Acute lymphoblastic leukemia. New England Journal of Medicine 2004;350:1535–48.CrossRefGoogle ScholarPubMed
Rowley, JD. Letter: a new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature 1973;243:290–3.CrossRefGoogle ScholarPubMed
Clark, SS, McLaughlin, J, Crist, WM, Champlin, R, Witte, ON. Unique forms of the abl tyrosine kinase distinguish Ph1-positive CML from Ph1-positive ALL. Science 1987;235:85–8.CrossRefGoogle ScholarPubMed
Nagar, B, Hantschel, O, Seeliger, M, et al. Organization of the SH3-SH2 unit in active and inactive forms of the c-Abl tyrosine kinase. Molecular Cell 2006;21:787–98.CrossRefGoogle ScholarPubMed
Nagar, B, Hantschel, O, Young, MA, et al. Structural basis for the autoinhibition of c-Abl tyrosine kinase. Cell 2003;112:859–71.CrossRefGoogle ScholarPubMed
Gesbert, F, Griffin, JD. Bcr/Abl activates transcription of the Bcl-X gene through STAT5. Blood 2000;96:2269–76.Google ScholarPubMed
Notari, M, Neviani, P, Santhanam, R, et al. A MAPK/HNRPK pathway controls BCR/ABL oncogenic potential by regulating MYC mRNA translation. Blood 2006;107:2507–16.CrossRefGoogle ScholarPubMed
Ren, R.Mechanisms of BCR-ABL in the pathogenesis of chronic myelogenous leukaemia. Nature Reviews Cancer 2005;5:172–83.CrossRefGoogle ScholarPubMed
Skorski, T, Kanakaraj, P, Nieborowska-Skorska, M, et al. Phosphatidylinositol-3 kinase activity is regulated by BCR/ABL and is required for the growth of Philadelphia chromosome-positive cells. Blood 1995;86:726–36.Google ScholarPubMed
Mullighan, CG, Miller, CB, Radtke, I, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature 2008;453:110–14.CrossRefGoogle ScholarPubMed
Martinelli, G, Iacobucci, I, Storlazzi, CT, et al. IKZF1 (Ikaros) deletions in BCR-ABL1-positive acute lymphoblastic leukemia are associated with short disease-free survival and high rate of cumulative incidence of relapse: a GIMEMA AL WP Report. Journal of Clinical Oncology 2009;27:5202–7.CrossRefGoogle ScholarPubMed
Gleiner, B, Gokbuget, N, Bartram, CR, et al. Leading prognostic relevance of the BCR-ABL translocation in adult acute B-lineage lymphoblastic leukemia: a prospective study of the German Multicenter Trial Group and confirmed polymerase chain reaction analysis. Blood 2002;99:1536–43.Google Scholar
Farhad, R, Partow, K. Philadelphia chromosome-positive acute lymphoblastic leukemia. Hematology/Oncology Clinics of North America 2009;23:1043–63.Google Scholar
Schultz, KR, Bowman, WP, Aledo, A, et al. Improved early event-free survival with imatinib in philadelphia chromosome-positive acute lymphoblastic leukemia: a Children's Oncology Group Study. Journal of Clinical Oncology 2009;27:5175–81.CrossRefGoogle ScholarPubMed
Pier Paolo, P, Stefania, P, Giovanni, M. Tyrosine kinase inhibitors for the treatment of Philadelphia chromosome-positive adult acute lymphoblastic leukemia. Cancer 2007;110:1178–86.Google Scholar
O’Hare, T, Shakespeare, WC, Zhu, X, et al. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell 2009;16:401–12.CrossRefGoogle ScholarPubMed
Yokoyama, A, Kitabayashi, I, Ayton, PM, Cleary, ML, Ohki, M. Leukemia proto-oncoprotein MLL is proteolytically processed into 2 fragments with opposite transcriptional properties. Blood 2002;100:3710–18.CrossRefGoogle ScholarPubMed
Yokoyama, A, Somervaille, TC, Smith, KS, et al. The menin tumor suppressor protein is an essential oncogenic cofactor for MLL-associated leukemogenesis. Cell 2005;123:207–18.CrossRefGoogle ScholarPubMed
Nie, Z, Yan, Z, Chen, EH, et al. Novel SWI/SNF chromatin-remodeling complexes contain a mixed-lineage leukemia chromosomal translocation partner. Molecular and Cellular Biology 2003;23:2942–52.CrossRefGoogle ScholarPubMed
Milne, TA, Briggs, SD, Brock, HW, et al. MLL targets SET domain methyltransferase activity to Hox gene promoters. Molecular Cell 2002;10:1107–17.CrossRefGoogle ScholarPubMed
Yu, BD, Hess, JL, Horning, SE, et al. Altered Hox expression and segmental identity in Mll-mutant mice. Nature 1995;378:505–8.CrossRefGoogle ScholarPubMed
McMahon, KA, Hiew, SY, Hadjur, S, et al. Mll has a critical role in fetal and adult hematopoietic stem cell self-renewal. Cell Stem Cell 2007;1:338–45.CrossRefGoogle Scholar
Lavau, C, Szilvassy, SJ, Slany, R, Cleary, ML. Immortalization and leukemic transformation of a myelomonocytic precursor by retrovirally transduced HRX-ENL. EMBO Journal 1997;16:4226–37.CrossRefGoogle ScholarPubMed
Chen, J, Santillan, DA, Koonce, M, et al. Loss of MLL PHD finger 3 is necessary for MLL-ENL-induced hematopoietic stem cell immortalization. Cancer Research 2008;68:6199–207.CrossRefGoogle ScholarPubMed
Horton, SJ, Grier, DG, McGonigle, GJ, et al. Continuous MLL-ENL expression is necessary to establish a “Hox Code” and maintain immortalization of hematopoietic progenitor cells. Cancer Research 2005;65:9245–52.CrossRefGoogle ScholarPubMed
Wong, P, Iwasaki, M, Somervaille, TC, So, CW, Cleary, ML. Meis1 is an essential and rate-limiting regulator of MLL leukemia stem cell potential. Genes and Development 2007;21:2762–74.CrossRefGoogle ScholarPubMed
Liedtke, M, Cleary, ML. Therapeutic targeting of MLL. Blood 2009;113:6061–8.CrossRefGoogle ScholarPubMed
Cauwelier, B, Cave, H, Gervais, C, et al. Clinical, cytogenetic and molecular characteristics of 14 T-ALL patients carrying the TCR[beta]-HOXA rearrangement: a study of the Groupe Francophone de Cytogenetique Hematologique. Leukemia 2006;21:121–8.CrossRefGoogle Scholar
Argiropoulos, B, Humphries, RK. Hox genes in hematopoiesis and leukemogenesis. Oncogene 2007;26:6766–76.CrossRefGoogle ScholarPubMed
Slany, RK. The molecular biology of mixed lineage leukemia. Haematologica 2009;94:984–93.CrossRefGoogle ScholarPubMed
So, CW, Lin, M, Ayton, PM, Chen, EH, Cleary, ML. Dimerization contributes to oncogenic activation of MLL chimeras in acute leukemias. Cancer Cell 2003;4:99–110.CrossRefGoogle ScholarPubMed
Armstrong, SA, Staunton, JE, Silverman, LB, et al. MLL translocations specify a distinct gene expression profile that distinguishes a unique leukemia. Nature Genetics 2002;30:41–7.CrossRefGoogle ScholarPubMed
Ferrando, AA, Armstrong, SA, Neuberg, DS, et al. Gene expression signatures in MLL-rearranged T-lineage and B-precursor acute leukemias: dominance of HOX dysregulation. Blood 2003;102:262–8.CrossRefGoogle ScholarPubMed
Yeoh, EJ, Ross, ME, Shurtleff, SA, et al. Classification, subtype discovery, and prediction of outcome in pediatric acute lymphoblastic leukemia by gene expression profiling. Cancer Cell 2002;1:133–43.CrossRefGoogle ScholarPubMed
Shi, A, Murai, MJ, He, S, et al. Structural insights into inhibition of the bivalent menin-MLL interaction by small molecules in leukemia. Blood 2012;120:4461–9.CrossRefGoogle ScholarPubMed
Filippakopoulos, P, Qi, J, Picaud, S, et al. Selective inhibition of BET bromodomains. Nature 2010;468:1067–73.CrossRefGoogle ScholarPubMed
Armstrong, SA, Kung, AL, Mabon, ME, et al. Inhibition of FLT3 in MLL. Validation of a therapeutic target identified by gene expression based classification. Cancer Cell 2003;3:173–83.CrossRefGoogle ScholarPubMed
Brown, P, Levis, M, McIntyre, E, Griesemer, M, Small, D. Combinations of the FLT3 inhibitor CEP-701 and chemotherapy synergistically kill infant and childhood MLL-rearranged ALL cells in a sequence-dependent manner. Leukemia 2006;20:1368–76.CrossRefGoogle Scholar
Brown, P, Hilden, J, Dreyer, Z, et al. Report on excessive induction toxicity in infants with ALL enrolled on COG Protocol AALL0631: a Children's Oncology Group Study. Blood 2009;114: Abstract #3091.Google Scholar
Friedman, AD. Leukemogenesis by CBF oncoproteins. Leukemia 1999;13:1932–42.CrossRefGoogle ScholarPubMed
Friedman, AD. Cell cycle and developmental control of hematopoiesis by Runx1. Journal of Cell Physiology 2009;219:520–4.CrossRefGoogle ScholarPubMed
Golub, TR., Barker, GF, Bohlander, SK, et al. Fusion of the TEL gene on 12p13 to the AML1 gene on 21q22 in acute lymphoblastic leukemia. Proceedings of the National Academy of Sciences USA 1995;92:4917–21.CrossRefGoogle ScholarPubMed
Raynaud, S, Cave, H, Baens, M, et al. The 12;21 translocation involving TEL and deletion of the other TEL allele: two frequently associated alterations found in childhood acute lymphoblastic leukemia. Blood 1996;87:2891–9.Google ScholarPubMed
Romana, SP, Mauchauffe, M, Le Coniat, M, et al. The t(12;21) of acute lymphoblastic leukemia results in a tel-AML1 gene fusion. Blood 1995;85:3662–70.Google Scholar
Loh, ML, Goldwasser, MA, Silverman, LB, et al. Prospective analysis of TEL/AML1-positive patients treated on Dana–Farber Cancer Institute Consortium Protocol 95–01. Blood 2006;107:4508–13.CrossRefGoogle ScholarPubMed
Bernardin, F, Yang, Y, Cleaves, R, et al. TEL-AML1, expressed from t(12;21) in human acute lymphocytic leukemia, induces acute leukemia in mice. Cancer Research 2002;62:3904–8.Google Scholar
Schindler, JW, Van Buren, D, Foudi, A, et al. TEL-AML1 corrupts hematopoietic stem cells to persist in the bone marrow and initiate leukemia. Cell Stem Cell 2009;5:43–53.CrossRefGoogle ScholarPubMed
Fenrick, R, Amann, JM, Lutterbach, B, et al. Both TEL and AML-1 contribute repression domains to the t(12;21) fusion protein. Molecular and Cellular Biology 1999;19:6566–74.CrossRefGoogle Scholar
Hong, D, Gupta, R, Ancliff, P, et al. Initiating and cancer-propagating cells in TEL-AML1-associated childhood leukemia. Science 2008;319:336–9.CrossRefGoogle ScholarPubMed
Ford, AM, Palmi, C, Bueno, C, et al. The TEL-AML1 leukemia fusion gene dysregulates the TGF-beta pathway in early B lineage progenitor cells. Journal of Clinical Investigation 2009;119:826–36.Google ScholarPubMed
Moorman, AV, Richards, SM, Robinson, HM, et al. Prognosis of children with acute lymphoblastic leukemia (ALL) and intrachromosomal amplification of chromosome 21 (iAMP21). Blood 2007;109:2327–30.CrossRefGoogle Scholar
Heerema, NA, Carroll, AJ, Borowitz, MJ, et al. Amplification of AML1 does not impact early outcome of children with acute lymphoblastic leukemia (ALL) treated with risk-directed chemotherapy: a report from the Children's Oncology Group (COG). Blood 2009;114:Abstract #2598.Google Scholar
Moosavi, SA, Sanchez, J, Adeyinka, A. Marker chromosomes are a significant mechanism of high-level RUNX1 gene amplification in hematologic malignancies. Cancer Genetics and Cytogenetics 2009;189:24–8.CrossRefGoogle ScholarPubMed
Monica, K, LeBrun, DP, Dedera, DA, Brown, R, Cleary, ML. Transformation properties of the E2a-Pbx1 chimeric oncoprotein: fusion with E2a is essential, but the Pbx1 homeodomain is dispensable. Molecular and Cellular Biology 1994;14:8304–14.CrossRefGoogle ScholarPubMed
Green, NC, Rambaldi, I, Teakles, J, Featherstone, MS. A conserved C-terminal domain in PBX increases DNA binding by the PBX homeodomain and is not a primary site of contact for the YPWM motif of HOXA1. Journal of Biological Chemistry 1998;273:13 273–9.CrossRefGoogle Scholar
Aulisa, L, Forraz, N, McGuckin, C, Hartgerink, JD. Inhibition of cancer cell proliferation by designed peptide amphiphiles. Acta Biomaterialia 2009;5:842–53.CrossRefGoogle ScholarPubMed
Morgan, R, Pirard, PM, Shears, L, et al. Antagonism of HOX/PBX dimer formation blocks the in vivo proliferation of melanoma. Cancer Research 2007;67:5806–13.CrossRefGoogle ScholarPubMed
Palomero, T, Sulis, ML, Cortina, M, et al. Mutational loss of PTEN induces resistance to NOTCH1 inhibition in T-cell leukemia. Nature Medicine 2007;13:1203–10.CrossRefGoogle ScholarPubMed
Kumano, K, Chiba, S, Kunisato, A, et al. Notch1 but not Notch2 is essential for generating hematopoietic stem cells from endothelial cells. Immunity 2003;18:699–711.CrossRefGoogle Scholar
Pui, JC, Allman, D, Xu, L, et al. Notch1 expression in early lymphopoiesis influences B versus T lineage determination. Immunity 1999;11:299–308.CrossRefGoogle Scholar
Radtke, F, Wilson, A, Stark, G, et al. Deficient T cell fate specification in mice with an induced inactivation of Notch1. Immunity 1999;10:547–58.CrossRefGoogle ScholarPubMed
Weng, AP, Ferrando, AA, Lee, W, et al. Activating mutations of NOTCH1 in human T cell acute lymphoblastic leukemia. Science 2004;306:269–71.CrossRefGoogle ScholarPubMed
Breit, S, Stanulla, M, Flohr, T, et al. Activating NOTCH1 mutations predict favorable early treatment response and long-term outcome in childhood precursor T-cell lymphoblastic leukemia. Blood 2006;108:1151–7.CrossRefGoogle ScholarPubMed
Real, PJ, Tosello, V, Palomero, T, et al. [gamma]-secretase inhibitors reverse glucocorticoid resistance in T cell acute lymphoblastic leukemia. Nature Medicine 2009;15:50–8.CrossRefGoogle ScholarPubMed
Tammam, J, Ware, C, Efferson, C, et al. Down-regulation of the Notch pathway mediated by a gamma-secretase inhibitor induces anti-tumour effects in mouse models of T-cell leukaemia. British Journal of Pharmacology 2009;158:1183–95.CrossRefGoogle ScholarPubMed
Wadman, IA, Osada, H, Grutz, GG, et al. The LIM-only protein Lmo2 is a bridging molecule assembling an erythroid, DNA-binding complex which includes the TAL1, E47, GATA-1 and Ldb1/NLI proteins. EMBO Journal 1997;16:3145–57.CrossRefGoogle ScholarPubMed
Lancrin, C, Sroczynska, P, Stephenson, C, et al. The haemangioblast generates haematopoietic cells through a haemogenic endothelium stage. Nature 2009;457:892–5.CrossRefGoogle ScholarPubMed
Porcher, C, Swat, W, Rockwell, K, et al. The T cell leukemia oncoprotein SCL/tal-1 is essential for development of all hematopoietic lineages. Cell 1996;86:47–57.CrossRefGoogle Scholar
O’Neil, J, Shank, J, Cusson, N, Murre, C, Kelliher, M. TAL1/SCL induces leukemia by inhibiting the transcriptional activity of E47/HEB. Cancer Cell 2004;5:587–96.CrossRefGoogle ScholarPubMed
Boehm, T, Foroni, L, Kaneko, Y, Perutz, MF, Rabbitts, TH. The rhombotin family of cysteine-rich LIM-domain oncogenes: distinct members are involved in T-cell translocations to human chromosomes 11p15 and 11p13. Proceedings of the National Academy of Sciences USA 1991;88:4367–71.CrossRefGoogle ScholarPubMed
Gothert, JR, Brake, RL, Smeets, M, et al. NOTCH1 pathway activation is an early hallmark of SCL T leukemogenesis. Blood 2007;110:3753–62.CrossRefGoogle ScholarPubMed
Serrano, M, Lee, H, Chin, L, et al. Role of the INK4a locus in tumor suppression and cell mortality. Cell 1996;85:27–37.CrossRefGoogle ScholarPubMed
Wong, IHN, Ng, MHL, Huang, DP, Lee, JCK. Aberrant p15 promoter methylation in adult and childhood acute leukemias of nearly all morphologic subtypes: potential prognostic implications. Blood 2000;95:1942–9.Google ScholarPubMed
Miyoung, K, Seon-Hee, Y, Nam-Sun, C, et al. 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. Cancer Genetics and Cytogenetics 2009;195:59–65.Google Scholar
Novara, F, Beri, S, Bernardo, ME, et al. Different molecular mechanisms causing 9p21 deletions in acute lymphoblastic leukemia of childhood. Human Genetics 2009;126:511–20.CrossRefGoogle ScholarPubMed
Drexler, HG. Review of alterations of the cyclin-dependent kinase inhibitor INK4 family genes p15, p16, p18 and p19 in human leukemia-lymphoma cells. Leukemia 1998;12:845–59.CrossRefGoogle ScholarPubMed
Williams, RT, Sherr, CJ. The INK4-ARF (CDKN2A/B) locus in hematopoiesis and BCR-ABL-induced leukemias. Cold Spring Harbor Symposia on Quantitative Biology 2008;73:461–7.CrossRefGoogle ScholarPubMed
Dalle, JH, Fournier, M, Nelken, B, et al. p16(INK4a) immunocytochemical analysis is an independent prognostic factor in childhood acute lymphoblastic leukemia. Blood 2002;99:2620–3.CrossRefGoogle ScholarPubMed
Tutor, O, Diaz, MA, Ramirez, M, et al. Loss of heterozygosity of p16 correlates with minimal residual disease at the end of the induction therapy in non-high risk childhood B-cell precursor acute lymphoblastic leukemia. Leukemia Research 2002;26:817–20.CrossRefGoogle ScholarPubMed
Sulong, S, Moorman, AV, Irving, JAE, et al. A comprehensive analysis of the CDKN2A gene in childhood acute lymphoblastic leukemia reveals genomic deletion, copy number neutral loss of heterozygosity, and association with specific cytogenetic subgroups. Blood 2009;113:100–7.CrossRefGoogle ScholarPubMed
Scott, LM, Civin, CI, Rorth, P, Friedman, AD. A novel temporal expression pattern of three C/EBP family members in differentiating myelomonocytic cells. Blood 1992;80:1725–35.Google ScholarPubMed
Xie, H, Ye, M, Feng, R, Graf, T. Stepwise reprogramming of B cells into macrophages. Cell 2004;117:663–76.CrossRefGoogle ScholarPubMed
Chapiro, E, Russell, L, Radford-Weiss, I, et al. Overexpression of CEBPA resulting from the translocation t(14;19)(q32;q13) of human precursor B acute lymphoblastic leukemia. Blood 2006;108:3560–3.CrossRefGoogle Scholar
Pabst, T, Mueller, BU, Zhang, P, et al. Dominant-negative mutations of CEBPA, encoding CCAAT/enhancer binding protein-alpha (C/EBPalpha), in acute myeloid leukemia. Nature Genetics 2001;27:263–70.CrossRefGoogle Scholar
Akasaka, T, Balasas, T, Russell, LJ, et al. Five members of the CEBP transcription factor family are targeted by recurrent IGH translocations in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Blood 2007;109:3451–61.CrossRefGoogle Scholar
Paz-Priel, I, Cai, DH, Wang, D, et al. CCAAT/enhancer binding protein alpha (C/EBPalpha) and C/EBPalpha myeloid oncoproteins induce bcl-2 via interaction of their basic regions with nuclear factor-kappaB p50. Molecular Cancer Research 2005;3:585–96.CrossRefGoogle Scholar
Paz-Priel, I, Ghosal, AK, Kowalski, J, Friedman, AD. C/EBPalpha or C/EBPalpha oncoproteins regulate the intrinsic and extrinsic apoptotic pathways by direct interaction with NF-kappaB p50 bound to the bcl-2 and FLIP gene promoters. Leukemia 2009;23:365–74.CrossRefGoogle ScholarPubMed
Porse, BT, Pedersen, TA, Xu, X, et al. E2F repression by C/EBPalpha is required for adipogenesis and granulopoiesis in vivo. Cell 2001;107:247–58.CrossRefGoogle Scholar
Dooher, JE, Paz-Priel, I, Houng, S, Baldwin, AS Jr., Friedman AD. C/EBPalpha, C/EBPalpha oncoproteins, or C/EBPbeta preferentially bind NF-kappaB p50 compared with p65, focusing therapeutic targeting on the C/EBP:p50 interaction. Molecular Cancer Research 2011;9:1395–405.CrossRefGoogle ScholarPubMed
Paz-Priel, I, Houng, S, Dooher, J, Friedman, AD. C/EBPalpha and C/EBPalpha oncoproteins regulate nfkb1 and displace histone deacetylases from NF-kappaB p50 homodimers to induce NF-kappaB target genes. Blood 2011;117:4085–94.CrossRefGoogle Scholar
Karin, M, Ben-Neriah, Y. Phosphorylation meets ubiquitination: the control of NF-[kappa]B activity. Annual Review of Immunology 2000;18:621–63.CrossRefGoogle ScholarPubMed
Zabel, U, Baeuerle, PA. Purified human I kappa B can rapidly dissociate the complex of the NF-kappa B transcription factor with its cognate DNA. Cell 1990;61:255–65.CrossRefGoogle Scholar
Kordes, U, Krappmann, D, Heissmeyer, V, Ludwig, WD, Scheidereit, C.Transcription factor NF-kappaB is constitutively activated in acute lymphoblastic leukemia cells. Leukemia 2000;14:399–402.CrossRefGoogle ScholarPubMed
Munzert, G, Kirchner, D, Ottmann, O, Bergmann, L, Schmid, RM. Constitutive NF-kappab/Rel activation in philadelphia chromosome positive (Ph+) acute lymphoblastic leukemia (ALL). Leukemia and Lymphoma 2004;45:1181–4.CrossRefGoogle Scholar
Mori, N, Fujii, M, Ikeda, S, et al. Constitutive activation of NF-kappaB in primary adult T-cell leukemia cells. Blood 1999;93:2360–8.Google ScholarPubMed
Kirchner, D, Duyster, J, Ottmann, O, et al. Mechanisms of Bcr-Abl-mediated NF-kappaB/Rel activation. Experimental Hematology 2003;31:504–11.CrossRefGoogle ScholarPubMed
Weston, VJ, Austen, B, Wei, W, et al. Apoptotic resistance to ionizing radiation in pediatric B-precursor acute lymphoblastic leukemia frequently involves increased NF-kappaB survival pathway signaling. Blood 2004;104:1465–73.CrossRefGoogle ScholarPubMed
Mayo, MW, Wang, CY, Cogswell, PC, et al. Requirement of NF-kappaB activation to suppress p53-independent apoptosis induced by oncogenic Ras. Science 1997;278:1812–15.CrossRefGoogle ScholarPubMed
Wang, CY, Mayo, MW, Korneluk, RG, Goeddel, DV, Baldwin, AS Jr. NF-kappaB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science 1998;281:1680–3.CrossRefGoogle ScholarPubMed
Wang, CY, Mayo, MW, Baldwin, AS Jr. TNF- and cancer therapy-induced apoptosis: potentiation by inhibition of NF-kappaB. Science 1996;274:784–7.CrossRefGoogle ScholarPubMed
Wang, CY, Cusack, JC Jr., Liu, R, Baldwin, AS Jr. Control of inducible chemoresistance: enhanced anti-tumor therapy through increased apoptosis by inhibition of NF-kappaB. Nature Medicine 1999;5:412–17.CrossRefGoogle ScholarPubMed
Guthridge, MA, Barry, EF, Felquer, FA, et al. The phosphoserine-585-dependent pathway of the GM-CSF/IL-3/IL-5 receptors mediates hematopoietic cell survival through activation of NF-kappaB and induction of bcl-2. Blood 2004;103:820–7.CrossRefGoogle ScholarPubMed
Catz, SD, Johnson, JL. Transcriptional regulation of bcl-2 by nuclear factor kappa B and its significance in prostate cancer. Oncogene 2001;20:7342–51.CrossRefGoogle ScholarPubMed
Heckman, CA, Mehew, JW, Boxer, LM. NF-kappaB activates Bcl-2 expression in t(14;18) lymphoma cells. Oncogene 2002;21:3898–908.CrossRefGoogle Scholar
Kucharczak, J, Simmons, MJ, Fan, Y, Gelinas, C. To be, or not to be: NF-kappaB is the answer-role of Rel/NF-kappaB in the regulation of apoptosis. Oncogene 2003;22:8961–82.CrossRefGoogle ScholarPubMed
Kurland, JF, Kodym, R, Story, MD, et al. NF-kappaB1 (p50) homodimers contribute to transcription of the bcl-2 oncogene. Journal of Biological Chemistry 2001;276:45 380–6.CrossRefGoogle ScholarPubMed
Horton, TM, Pati, D, Plon, SE, et al. A phase 1 study of the proteasome inhibitor bortezomib in pediatric patients with refractory leukemia: a Children's Oncology Group study. Clinical Cancer Research 2007;13:1516–22.CrossRefGoogle ScholarPubMed
Hasle, H. Pattern of malignant disorders in individuals with Down's syndrome. Lancet Oncology 2001;2:429–36.CrossRefGoogle ScholarPubMed
Malinge, S, Izraeli, S, Crispino, JD. Insights into the manifestations, outcomes, and mechanisms of leukemogenesis in Down syndrome. Blood 2009;113:2619–28.CrossRefGoogle ScholarPubMed
Bercovich, D, Ganmore, I, Scott, LM, et al. Mutations of JAK2 in acute lymphoblastic leukaemias associated with Down's syndrome. Lancet 2008;372:1484–92.CrossRefGoogle ScholarPubMed
Kearney, L, Gonzalez, DeCastro, D, Yeung, J, et al. Specific JAK2 mutation (JAK2R683) and multiple gene deletions in Down syndrome acute lymphoblastic leukemia. Blood 2009;113:646–8.CrossRefGoogle ScholarPubMed
Mullighan, CG, Collins-Underwood, JR, Phillips, LAA, et al. Rearrangement of CRLF2 in B-progenitor- and Down syndrome-associated acute lymphoblastic leukemia. Nature Genetics 2009;41:1243–6.CrossRefGoogle ScholarPubMed
Russell, LJ, Capasso, M, Vater, I, et al. Deregulated expression of cytokine receptor gene, CRLF2, is involved in lymphoid transformation in B-cell precursor acute lymphoblastic leukemia. Blood 2009;114:2688–98.CrossRefGoogle ScholarPubMed
Mullighan, CG, Goorha, S, Radtke, I, et al. Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia. Nature 2007;446:758–64.CrossRefGoogle ScholarPubMed
Kuiper, RP, Schoenmakers, EFPM, van Reijmersdal, SV, et al. High-resolution genomic profiling of childhood ALL reveals novel recurrent genetic lesions affecting pathways involved in lymphocyte differentiation and cell cycle progression. Leukemia 2007;21:1258–66.CrossRefGoogle ScholarPubMed
Kawamata, N, Ogawa, S, Zimmermann, M, et al. Cloning of genes involved in chromosomal translocations by high-resolution single nucleotide polymorphism genomic microarray. Proceedings of the National Academy of Sciences USA 2008;105:11 921–6.CrossRefGoogle ScholarPubMed
Mullighan, CG, Su, X, Zhang, J, et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. New England Journal of Medicine 2009;360:470–80.CrossRefGoogle ScholarPubMed
Urbanek, P, Wang, ZQ, Fetka, I, Wagner, EF, Busslinger, M. Complete block of early B cell differentiation and altered patterning of the posterior midbrain in mice lacking Pax5/BSAP. Cell 1994;79:901–12.CrossRefGoogle Scholar
Paulsson, K, Johansson, B. High hyperdiploid childhood acute lymphoblastic leukemia. Genes, Chromosomes and Cancer 2009;48:637–60.CrossRefGoogle ScholarPubMed
Sharathkumar, A, DeCamillo, D, Bhambhani, K, et al. Children with hyperdiploid but not triple trisomy (+4,+10,+17) acute lymphoblastic leukemia have an increased incidence of extramedullary relapse on current therapies: a single institution experience. American Journal of Hematology 2008;83:34–40.CrossRefGoogle Scholar
Sutcliffe, MJ, Shuster, JJ, Sather, HN, et al. High concordance from independent studies by the Children's Cancer Group (CCG) and Pediatric Oncology Group (POG) associating favorable prognosis with combined trisomies 4, 10, and 17 in children with NCI Standard-Risk B-precursor acute lymphoblastic leukemia: a Children's Oncology Group (COG) initiative. Leukemia 2005;19:734–40.CrossRefGoogle ScholarPubMed
Pui, C-H, Robison, LL, Look, AT. Acute lymphoblastic leukaemia. Lancet 2008;371:1030–43.CrossRefGoogle ScholarPubMed
Armstrong, SA, Look, AT. Molecular genetics of acute lymphoblastic leukemia. Journal of Clinical Oncology 2005;23:6306–15.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×