Skip to main content
Top
Published in: Molecular Cancer 1/2009

Open Access 01-12-2009 | Research

CHK1 inhibition as a strategy for targeting fanconi anemia (FA) DNA repair pathway deficient tumors

Authors: Clark C Chen, Richard D Kennedy, Samuel Sidi, A Thomas Look, Alan D'Andrea

Published in: Molecular Cancer | Issue 1/2009

Login to get access

Abstract

Background

DNA repair deficient tumor cells have been shown to accumulate high levels of DNA damage. Consequently, these cells become hyper-dependent on DNA damage response pathways, including the CHK1-kinase-mediated response. These observations suggest that DNA repair deficient tumors should exhibit increased sensitivity to CHK1 inhibition. Here we offer experimental evidence in support of this hypothesis.

Results

Using isogenic pairs of cell lines differing only in the Fanconi Anemia (FA) DNA repair pathway, we showed that FA deficient cell lines were hypersensitive to CHK1 silencing by independent siRNAs as well as CHK1 pharmacologic inhibition by Gö6976 and UCN-01. In parallel, an siRNA screen designed to identify gene silencings synthetically lethal with CHK1 inhibition identified genes required for FA pathway function. To confirm these findings in vivo, we demonstrated that whole zebrafish embryos, depleted for FANCD2 by a morpholino approach, were hypersensitive to Gö6976. Silencing of FA genes led to hyper-activation of CHK1 and vice versa. Furthermore, inactivation of CHK1 in FA deficient cell lines caused increased accumulation of DNA strand and chromosomal breakages. These results suggest that the functions subserved by CHK1 and the FA pathway mutually compensate in maintaining genome integrity. As CHK1 inhibition has been under clinical trial in combination with cisplatin, we showed that the FA specific tumoricidal effect of CHK1 inhibition and cisplatin was synergistic.

Conclusion

Taken together, these results suggest CHK1 inhibition as a strategy for targeting FA deficient tumors.
Appendix
Available only for authorised users
Literature
1.
go back to reference Lengauer C, Kinzler KW, Vogelstein B: Genetic instabilities in human cancers. Nature. 1998, 396: 643-649. 10.1038/25292CrossRefPubMed Lengauer C, Kinzler KW, Vogelstein B: Genetic instabilities in human cancers. Nature. 1998, 396: 643-649. 10.1038/25292CrossRefPubMed
2.
go back to reference Kastan MB, Bartek J: Cell-cycle checkpoints and cancer. Nature. 2004, 432: 316-323. 10.1038/nature03097CrossRefPubMed Kastan MB, Bartek J: Cell-cycle checkpoints and cancer. Nature. 2004, 432: 316-323. 10.1038/nature03097CrossRefPubMed
3.
go back to reference Kennedy RD, D'Andrea AD: DNA repair pathways in clinical practice: lessons from pediatric cancer susceptibility syndromes. J Clin Oncol. 2006, 24: 3799-3808. 10.1200/JCO.2005.05.4171CrossRefPubMed Kennedy RD, D'Andrea AD: DNA repair pathways in clinical practice: lessons from pediatric cancer susceptibility syndromes. J Clin Oncol. 2006, 24: 3799-3808. 10.1200/JCO.2005.05.4171CrossRefPubMed
4.
go back to reference Wang W: Emergence of a DNA-damage response network consisting of Fanconi anaemia and BRCA proteins. Nat Rev Genet. 2007, 10: 735-748. 10.1038/nrg2159. 10.1038/nrg2159CrossRef Wang W: Emergence of a DNA-damage response network consisting of Fanconi anaemia and BRCA proteins. Nat Rev Genet. 2007, 10: 735-748. 10.1038/nrg2159. 10.1038/nrg2159CrossRef
5.
go back to reference Chen CC, Taniguchi T, D'Andrea A: The Fanconi anemia (FA) pathway confers glioma resistance to DNA alkylating agents. J Mol Med. 2007, 85: 497-509. Epub 2007 Jan 2013., 10.1007/s00109-006-0153-2CrossRefPubMed Chen CC, Taniguchi T, D'Andrea A: The Fanconi anemia (FA) pathway confers glioma resistance to DNA alkylating agents. J Mol Med. 2007, 85: 497-509. Epub 2007 Jan 2013., 10.1007/s00109-006-0153-2CrossRefPubMed
6.
go back to reference Taniguchi T, Tischkowitz M, Ameziane N, Hodgson SV, Mathew CG, Joenje H, Mok SC, D'Andrea AD: Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nat Med. 2003, 9: 568-574. 10.1038/nm852CrossRefPubMed Taniguchi T, Tischkowitz M, Ameziane N, Hodgson SV, Mathew CG, Joenje H, Mok SC, D'Andrea AD: Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nat Med. 2003, 9: 568-574. 10.1038/nm852CrossRefPubMed
7.
go back to reference Narayan G, Arias-Pulido H, Nandula SV, Basso K, Sugirtharaj DD, Vargas H, Mansukhani M, Villella J, Meyer L, Schneider A, Gissmann L, Durst M, Pothuri B, Murty VV: Promoter hypermethylation of FANCF: disruption of Fanconi Anemia-BRCA pathway in cervical cancer. Cancer Res. 2004, 64: 2994-2997. 10.1158/0008-5472.CAN-04-0245CrossRefPubMed Narayan G, Arias-Pulido H, Nandula SV, Basso K, Sugirtharaj DD, Vargas H, Mansukhani M, Villella J, Meyer L, Schneider A, Gissmann L, Durst M, Pothuri B, Murty VV: Promoter hypermethylation of FANCF: disruption of Fanconi Anemia-BRCA pathway in cervical cancer. Cancer Res. 2004, 64: 2994-2997. 10.1158/0008-5472.CAN-04-0245CrossRefPubMed
8.
go back to reference Marsit CJ, Liu M, Nelson HH, Posner M, Suzuki M, Kelsey KT: Inactivation of the Fanconi anemia/BRCA pathway in lung and oral cancers: implications for treatment and survival. Oncogene. 2003, 23: 1000-1004. 10.1038/sj.onc.1207256. 10.1038/sj.onc.1207256CrossRef Marsit CJ, Liu M, Nelson HH, Posner M, Suzuki M, Kelsey KT: Inactivation of the Fanconi anemia/BRCA pathway in lung and oral cancers: implications for treatment and survival. Oncogene. 2003, 23: 1000-1004. 10.1038/sj.onc.1207256. 10.1038/sj.onc.1207256CrossRef
9.
go back to reference Tischkowitz M, Ameziane N, Waisfisz Q, De Winter JP, Harris R, Taniguchi T, D'Andrea A, Hodgson SV, Mathew CG, Joenje H: Bi-allelic silencing of the Fanconi anaemia gene FANCF in acute myeloid leukaemia. Br J Haematol. 2003, 123: 469-471. 10.1046/j.1365-2141.2003.04640.xCrossRefPubMed Tischkowitz M, Ameziane N, Waisfisz Q, De Winter JP, Harris R, Taniguchi T, D'Andrea A, Hodgson SV, Mathew CG, Joenje H: Bi-allelic silencing of the Fanconi anaemia gene FANCF in acute myeloid leukaemia. Br J Haematol. 2003, 123: 469-471. 10.1046/j.1365-2141.2003.04640.xCrossRefPubMed
10.
go back to reference Taniguchi T, D'Andrea AD: Molecular pathogenesis of Fanconi anemia: recent progress. Blood. 2006, 107: 4223-4233. Epub 2006 Feb 4221., 10.1182/blood-2005-10-4240CrossRefPubMed Taniguchi T, D'Andrea AD: Molecular pathogenesis of Fanconi anemia: recent progress. Blood. 2006, 107: 4223-4233. Epub 2006 Feb 4221., 10.1182/blood-2005-10-4240CrossRefPubMed
11.
go back to reference Kennedy RD, Chen CC, Stuckert P, Archila EM, De la Vega MA, Moreau LA, Shimamura A, D'Andrea AD: Fanconi anemia pathway-deficient tumor cells are hypersensitive to inhibition of ataxia telangiectasia mutated. J Clin Invest. 2007, 117: 1440-1449. 10.1172/JCI31245PubMedCentralCrossRefPubMed Kennedy RD, Chen CC, Stuckert P, Archila EM, De la Vega MA, Moreau LA, Shimamura A, D'Andrea AD: Fanconi anemia pathway-deficient tumor cells are hypersensitive to inhibition of ataxia telangiectasia mutated. J Clin Invest. 2007, 117: 1440-1449. 10.1172/JCI31245PubMedCentralCrossRefPubMed
12.
go back to reference Latt SA, Kaiser TN, Lojewski A, Dougherty C, Juergens L, Brefach S, Sahar E, Gustashaw K, Schreck RR, Powers M, Lalande M: Cytogenetic and flow cytometric studies of cells from patients with Fanconi's anemia. Cytogenet Cell Genet. 1982, 33: 133-138. 10.1159/000131737CrossRefPubMed Latt SA, Kaiser TN, Lojewski A, Dougherty C, Juergens L, Brefach S, Sahar E, Gustashaw K, Schreck RR, Powers M, Lalande M: Cytogenetic and flow cytometric studies of cells from patients with Fanconi's anemia. Cytogenet Cell Genet. 1982, 33: 133-138. 10.1159/000131737CrossRefPubMed
13.
go back to reference Kupfer GM, D'Andrea AD: The effect of the Fanconi anemia polypeptide, FAC, upon p53 induction and G2 checkpoint regulation. Blood. 1996, 88: 1019-1025.PubMed Kupfer GM, D'Andrea AD: The effect of the Fanconi anemia polypeptide, FAC, upon p53 induction and G2 checkpoint regulation. Blood. 1996, 88: 1019-1025.PubMed
14.
go back to reference Pulsipher M, Kupfer GM, Naf D, Suliman A, Lee JS, Jakobs P, Grompe M, Joenje H, Sieff C, Guinan E, Mulligan R, D'Andrea AD: Subtyping analysis of Fanconi anemia by immunoblotting and retroviral gene transfer. Mol Med. 1998, 4: 468-479.PubMedCentralPubMed Pulsipher M, Kupfer GM, Naf D, Suliman A, Lee JS, Jakobs P, Grompe M, Joenje H, Sieff C, Guinan E, Mulligan R, D'Andrea AD: Subtyping analysis of Fanconi anemia by immunoblotting and retroviral gene transfer. Mol Med. 1998, 4: 468-479.PubMedCentralPubMed
15.
go back to reference Andreassen PR, D'Andrea AD, Taniguchi T: ATR couples FANCD2 monoubiquitination to the DNA-damage response. Genes Dev. 2004, 18: 1958-1963. 10.1101/gad.1196104PubMedCentralCrossRefPubMed Andreassen PR, D'Andrea AD, Taniguchi T: ATR couples FANCD2 monoubiquitination to the DNA-damage response. Genes Dev. 2004, 18: 1958-1963. 10.1101/gad.1196104PubMedCentralCrossRefPubMed
16.
go back to reference Bartek J, Lukas J: Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell. 2003, 3: 421-429. 10.1016/S1535-6108(03)00110-7CrossRefPubMed Bartek J, Lukas J: Chk1 and Chk2 kinases in checkpoint control and cancer. Cancer Cell. 2003, 3: 421-429. 10.1016/S1535-6108(03)00110-7CrossRefPubMed
17.
go back to reference Tse AN, Carvajal R, Schwartz GK: Targeting checkpoint kinase 1 in cancer therapeutics. Clin Cancer Res. 2007, 13: 1955-1960. 10.1158/1078-0432.CCR-06-2793CrossRefPubMed Tse AN, Carvajal R, Schwartz GK: Targeting checkpoint kinase 1 in cancer therapeutics. Clin Cancer Res. 2007, 13: 1955-1960. 10.1158/1078-0432.CCR-06-2793CrossRefPubMed
18.
go back to reference Zhou BB, Bartek J: Targeting the checkpoint kinases: chemosensitization versus chemoprotection. Nat Rev Cancer. 2004, 4: 216-225. 10.1038/nrc1296CrossRefPubMed Zhou BB, Bartek J: Targeting the checkpoint kinases: chemosensitization versus chemoprotection. Nat Rev Cancer. 2004, 4: 216-225. 10.1038/nrc1296CrossRefPubMed
19.
go back to reference Lara PN, Mack PC, Synold T, Frankel P, Longmate J, Gumerlock PH, Doroshow JH, Gandara DR: The cyclin-dependent kinase inhibitor UCN-01 plus cisplatin in advanced solid tumors: a California cancer consortium phase I pharmacokinetic and molecular correlative trial. Clin Cancer Res. 2005, 11: 4444-4450. 10.1158/1078-0432.CCR-04-2602CrossRefPubMed Lara PN, Mack PC, Synold T, Frankel P, Longmate J, Gumerlock PH, Doroshow JH, Gandara DR: The cyclin-dependent kinase inhibitor UCN-01 plus cisplatin in advanced solid tumors: a California cancer consortium phase I pharmacokinetic and molecular correlative trial. Clin Cancer Res. 2005, 11: 4444-4450. 10.1158/1078-0432.CCR-04-2602CrossRefPubMed
20.
go back to reference Kortmansky J, Shah MA, Kaubisch A, Weyerbacher A, Yi S, Tong W, Sowers R, Gonen M, O'Reilly E, Kemeny N, Ilson DI, Saltz LB, Maki RG, Kelsen DP, Schwartz GK: Phase I trial of the cyclin-dependent kinase inhibitor and protein kinase C inhibitor 7-hydroxystaurosporine in combination with Fluorouracil in patients with advanced solid tumors. J Clin Oncol. 2005, 23: 1875-1884. 10.1200/JCO.2005.03.116CrossRefPubMed Kortmansky J, Shah MA, Kaubisch A, Weyerbacher A, Yi S, Tong W, Sowers R, Gonen M, O'Reilly E, Kemeny N, Ilson DI, Saltz LB, Maki RG, Kelsen DP, Schwartz GK: Phase I trial of the cyclin-dependent kinase inhibitor and protein kinase C inhibitor 7-hydroxystaurosporine in combination with Fluorouracil in patients with advanced solid tumors. J Clin Oncol. 2005, 23: 1875-1884. 10.1200/JCO.2005.03.116CrossRefPubMed
21.
go back to reference Hotte SJ, Oza A, Winquist EW, Moore M, Chen EX, Brown S, Pond GR, Dancey JE, Hirte HW: Phase I trial of UCN-01 in combination with topotecan in patients with advanced solid cancers: a Princess Margaret Hospital Phase II Consortium study. Ann Oncol. 2006, 17: 334-340. 10.1093/annonc/mdj076CrossRefPubMed Hotte SJ, Oza A, Winquist EW, Moore M, Chen EX, Brown S, Pond GR, Dancey JE, Hirte HW: Phase I trial of UCN-01 in combination with topotecan in patients with advanced solid cancers: a Princess Margaret Hospital Phase II Consortium study. Ann Oncol. 2006, 17: 334-340. 10.1093/annonc/mdj076CrossRefPubMed
22.
go back to reference Sampath D, Cortes J, Estrov Z, Du M, Shi Z, Andreeff M, Gandhi V, Plunkett W: Pharmacodynamics of cytarabine alone and in combination with 7-hydroxystaurosporine (UCN-01) in AML blasts in vitro and during a clinical trial. Blood. 2006, 107: 2517-2524. 10.1182/blood-2005-08-3351PubMedCentralCrossRefPubMed Sampath D, Cortes J, Estrov Z, Du M, Shi Z, Andreeff M, Gandhi V, Plunkett W: Pharmacodynamics of cytarabine alone and in combination with 7-hydroxystaurosporine (UCN-01) in AML blasts in vitro and during a clinical trial. Blood. 2006, 107: 2517-2524. 10.1182/blood-2005-08-3351PubMedCentralCrossRefPubMed
23.
go back to reference Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H, Klevernic I, Arthur JS, Alessi DR, Cohen P: The selectivity of protein kinase inhibitors: a further update. Biochem J. 2007, 408: 297-315. 10.1042/BJ20070797PubMedCentralCrossRefPubMed Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H, Klevernic I, Arthur JS, Alessi DR, Cohen P: The selectivity of protein kinase inhibitors: a further update. Biochem J. 2007, 408: 297-315. 10.1042/BJ20070797PubMedCentralCrossRefPubMed
24.
go back to reference Kohn EA, Yoo CJ, Eastman A: The protein kinase C inhibitor Gö6976 is a potent inhibitor of DNA damage-induced S and G2 cell cycle checkpoints. Cancer Res. 2003, 63: 31-35.PubMed Kohn EA, Yoo CJ, Eastman A: The protein kinase C inhibitor Gö6976 is a potent inhibitor of DNA damage-induced S and G2 cell cycle checkpoints. Cancer Res. 2003, 63: 31-35.PubMed
25.
go back to reference Seynaeve CM, Kazanietz MG, Blumberg PM, Sausville EA, Worland PJ: Differential inhibition of protein kinase C isozymes by UCN-01, a staurosporine analogue. Molecular Pharmacology. 1994, 45: 1207-1214.PubMed Seynaeve CM, Kazanietz MG, Blumberg PM, Sausville EA, Worland PJ: Differential inhibition of protein kinase C isozymes by UCN-01, a staurosporine analogue. Molecular Pharmacology. 1994, 45: 1207-1214.PubMed
26.
go back to reference MacKeigan JP, Murphy LO, Blenis J: Sensitized RNAi screen of human kinases and phosphatases identifies new regulators of apoptosis and chemoresistance. Nature Cell Biology. 2005, 7: 591-600. 10.1038/ncb1258CrossRefPubMed MacKeigan JP, Murphy LO, Blenis J: Sensitized RNAi screen of human kinases and phosphatases identifies new regulators of apoptosis and chemoresistance. Nature Cell Biology. 2005, 7: 591-600. 10.1038/ncb1258CrossRefPubMed
27.
go back to reference Yarden RI, Pardo-Reoyo S, Sgagias M, Cowan KH, Brody LC: BRCA1 regulates the G2/M checkpoint by activating Chk1 kinase upon DNA damage. Nat Genet. 2002, 30: 285-289. 10.1038/ng837CrossRefPubMed Yarden RI, Pardo-Reoyo S, Sgagias M, Cowan KH, Brody LC: BRCA1 regulates the G2/M checkpoint by activating Chk1 kinase upon DNA damage. Nat Genet. 2002, 30: 285-289. 10.1038/ng837CrossRefPubMed
28.
go back to reference Kennedy RD, D'Andrea AD: The Fanconi Anemia/BRCA pathway: new faces in the crowd. Genes Dev. 2005, 19: 2925-2940. 10.1101/gad.1370505CrossRefPubMed Kennedy RD, D'Andrea AD: The Fanconi Anemia/BRCA pathway: new faces in the crowd. Genes Dev. 2005, 19: 2925-2940. 10.1101/gad.1370505CrossRefPubMed
29.
go back to reference Taniguchi T, Garcia-Higuera I, Andreassen PR, Gregory RC, Grompe M, D'Andrea AD: S-phase-specific interaction of the Fanconi anemia protein, FANCD2, with BRCA1 and RAD51. Blood. 2002, 100: 2414-2420. 10.1182/blood-2002-01-0278CrossRefPubMed Taniguchi T, Garcia-Higuera I, Andreassen PR, Gregory RC, Grompe M, D'Andrea AD: S-phase-specific interaction of the Fanconi anemia protein, FANCD2, with BRCA1 and RAD51. Blood. 2002, 100: 2414-2420. 10.1182/blood-2002-01-0278CrossRefPubMed
30.
go back to reference Syljuasen RG, Sorensen CS, Hansen LT, Fugger K, Lundin C, Johansson F, Helleday T, Sehested M, Lukas J, Bartek J: Inhibition of human Chk1 causes increased initiation of DNA replication, phosphorylation of ATR targets, and DNA breakage. Mol Cell Biol. 2005, 25: 3553-3562. 10.1128/MCB.25.9.3553-3562.2005PubMedCentralCrossRefPubMed Syljuasen RG, Sorensen CS, Hansen LT, Fugger K, Lundin C, Johansson F, Helleday T, Sehested M, Lukas J, Bartek J: Inhibition of human Chk1 causes increased initiation of DNA replication, phosphorylation of ATR targets, and DNA breakage. Mol Cell Biol. 2005, 25: 3553-3562. 10.1128/MCB.25.9.3553-3562.2005PubMedCentralCrossRefPubMed
31.
go back to reference Liu TX, Howlett NG, Deng M, Langenau DM, Hsu K, Rhodes J, Kanki JP, D'Andrea AD, Look AT: Knockdown of zebrafish Fancd2 causes developmental abnormalities via p53-dependent apoptosis. Dev Cell. 2003, 5: 903-914. 10.1016/S1534-5807(03)00339-3CrossRefPubMed Liu TX, Howlett NG, Deng M, Langenau DM, Hsu K, Rhodes J, Kanki JP, D'Andrea AD, Look AT: Knockdown of zebrafish Fancd2 causes developmental abnormalities via p53-dependent apoptosis. Dev Cell. 2003, 5: 903-914. 10.1016/S1534-5807(03)00339-3CrossRefPubMed
32.
go back to reference Sidi S, Sanda T, Kennedy RD, Hagen AT, Jette CA, Hoffmans R, Pascual J, Imamura S, Kishi S, Amatruda JF, Kanki JP, Green DR, D'Andrea AA, Look AT: Chk1 suppresses a caspase-2 apoptotic response to DNA damage that bypasses p53, Bcl-2, and caspase-3. Cell. 2008, 133: 864-877. 10.1016/j.cell.2008.03.037PubMedCentralCrossRefPubMed Sidi S, Sanda T, Kennedy RD, Hagen AT, Jette CA, Hoffmans R, Pascual J, Imamura S, Kishi S, Amatruda JF, Kanki JP, Green DR, D'Andrea AA, Look AT: Chk1 suppresses a caspase-2 apoptotic response to DNA damage that bypasses p53, Bcl-2, and caspase-3. Cell. 2008, 133: 864-877. 10.1016/j.cell.2008.03.037PubMedCentralCrossRefPubMed
33.
go back to reference Chou TC: Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006, 58: 621-681. 10.1124/pr.58.3.10CrossRefPubMed Chou TC: Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006, 58: 621-681. 10.1124/pr.58.3.10CrossRefPubMed
34.
go back to reference Sha SK, Sato T, Kobayashi H, Ishigaki M, Yamamoto S, Sato H, Takada A, Nakajyo S, Mochizuki Y, Friedman JM, Cheng FC, Okura T, Kimura R, Kufe DW, Vonhoff DD, Kawabe T: Cell cycle phenotype-based optimization of G2-abrogating peptides yields CBP501 with a unique mechanism of action at the G2 checkpoint. Mol Cancer Ther. 2007, 6: 147-153. 10.1158/1535-7163.MCT-06-0371CrossRefPubMed Sha SK, Sato T, Kobayashi H, Ishigaki M, Yamamoto S, Sato H, Takada A, Nakajyo S, Mochizuki Y, Friedman JM, Cheng FC, Okura T, Kimura R, Kufe DW, Vonhoff DD, Kawabe T: Cell cycle phenotype-based optimization of G2-abrogating peptides yields CBP501 with a unique mechanism of action at the G2 checkpoint. Mol Cancer Ther. 2007, 6: 147-153. 10.1158/1535-7163.MCT-06-0371CrossRefPubMed
35.
go back to reference Zabludoff SD, Deng C, Grondine MR, Sheehy AM, Ashwell S, Caleb BL, Green S, Haye HR, Horn CL, Janetka JW, Liu D, Mouchet E, Ready S, Rosenthal JL, Queva C, Schwartz GK, Taylor KJ, Tse AN, Walker GE, White AM: AZD a novel checkpoint kinase inhibitor, drives checkpoint abrogation and potentiates DNA-targeted therapies. Molecular Cancer Therapeutics. 7762, 7: 2955-2966. 10.1158/1535-7163.MCT-08-0492. 10.1158/1535-7163.MCT-08-0492CrossRef Zabludoff SD, Deng C, Grondine MR, Sheehy AM, Ashwell S, Caleb BL, Green S, Haye HR, Horn CL, Janetka JW, Liu D, Mouchet E, Ready S, Rosenthal JL, Queva C, Schwartz GK, Taylor KJ, Tse AN, Walker GE, White AM: AZD a novel checkpoint kinase inhibitor, drives checkpoint abrogation and potentiates DNA-targeted therapies. Molecular Cancer Therapeutics. 7762, 7: 2955-2966. 10.1158/1535-7163.MCT-08-0492. 10.1158/1535-7163.MCT-08-0492CrossRef
36.
go back to reference Liu Q, Guntuku S, Cui XS, Matsuoka S, Cortez D, Tamai K, Luo G, Carattini-Rivera S, DeMayo F, Bradley A, Donehower LA, Elledge SJ: Chk1 is an essential kinase that is regulated by Atr and required for the G(2)/M DNA damage checkpoint. Genes Dev. 2000, 14: 1448-1459. 10.1101/gad.840500PubMedCentralCrossRefPubMed Liu Q, Guntuku S, Cui XS, Matsuoka S, Cortez D, Tamai K, Luo G, Carattini-Rivera S, DeMayo F, Bradley A, Donehower LA, Elledge SJ: Chk1 is an essential kinase that is regulated by Atr and required for the G(2)/M DNA damage checkpoint. Genes Dev. 2000, 14: 1448-1459. 10.1101/gad.840500PubMedCentralCrossRefPubMed
37.
go back to reference Garcia-Higuera I, Kuang Y, Denham J, D'Andrea AD: The fanconi anemia proteins FANCA and FANCG stabilize each other and promote the nuclear accumulation of the Fanconi anemia complex. Blood. 2000, 96: 3224-3230.PubMed Garcia-Higuera I, Kuang Y, Denham J, D'Andrea AD: The fanconi anemia proteins FANCA and FANCG stabilize each other and promote the nuclear accumulation of the Fanconi anemia complex. Blood. 2000, 96: 3224-3230.PubMed
38.
go back to reference Garcia-Higuera I, Taniguchi T, Ganesan S, Meyn MS, Timmers C, Hejna J, Grompe M, D'Andrea AD: Interaction of the Fanconi anemia proteins and BRCA1 in a common pathway. Mol Cell. 2001, 7: 249-262. 10.1016/S1097-2765(01)00173-3CrossRefPubMed Garcia-Higuera I, Taniguchi T, Ganesan S, Meyn MS, Timmers C, Hejna J, Grompe M, D'Andrea AD: Interaction of the Fanconi anemia proteins and BRCA1 in a common pathway. Mol Cell. 2001, 7: 249-262. 10.1016/S1097-2765(01)00173-3CrossRefPubMed
39.
go back to reference Naf D, Kupfer GM, Suliman A, Lambert K, D'Andrea AD: Functional activity of the fanconi anemia protein FAA requires FAC binding and nuclear localization. Mol Cell Biol. 1998, 18: 5952-5960.PubMedCentralPubMed Naf D, Kupfer GM, Suliman A, Lambert K, D'Andrea AD: Functional activity of the fanconi anemia protein FAA requires FAC binding and nuclear localization. Mol Cell Biol. 1998, 18: 5952-5960.PubMedCentralPubMed
40.
go back to reference Stewart GS, Wang B, Bignell CR, Taylor AM, Elledge SJ: MDC1 is a mediator of the mammalian DNA damage checkpoint. Nature. 2003, 421: 961-966. 10.1038/nature01446CrossRefPubMed Stewart GS, Wang B, Bignell CR, Taylor AM, Elledge SJ: MDC1 is a mediator of the mammalian DNA damage checkpoint. Nature. 2003, 421: 961-966. 10.1038/nature01446CrossRefPubMed
41.
go back to reference Wang X, Kennedy RD, Ray K, Stuckert P, Ellenberger T, D'Andrea AD: Chk1-mediated phosphorylation of FANCE is required for the Fanconi anemia/BRCA pathway. Mol Cell Biol. 2007, 27: 3098-3108. 10.1128/MCB.02357-06PubMedCentralCrossRefPubMed Wang X, Kennedy RD, Ray K, Stuckert P, Ellenberger T, D'Andrea AD: Chk1-mediated phosphorylation of FANCE is required for the Fanconi anemia/BRCA pathway. Mol Cell Biol. 2007, 27: 3098-3108. 10.1128/MCB.02357-06PubMedCentralCrossRefPubMed
42.
go back to reference Kupfer GM, Naf D, Suliman A, Pulsipher M, D'Andrea AD: The Fanconi anaemia proteins, FAA and FAC, interact to form a nuclear complex. Nat Genet. 1997, 17: 487-490. 10.1038/ng1297-487CrossRefPubMed Kupfer GM, Naf D, Suliman A, Pulsipher M, D'Andrea AD: The Fanconi anaemia proteins, FAA and FAC, interact to form a nuclear complex. Nat Genet. 1997, 17: 487-490. 10.1038/ng1297-487CrossRefPubMed
43.
go back to reference Siddique MA, Nakanishi K, Taniguchi T, Grompe M, D'Andrea AD: Function of the Fanconi anemia pathway in Fanconi anemia complementation group F and D1 cells. Exp Hematol. 2001, 29: 1448-1455. 10.1016/S0301-472X(01)00754-8CrossRefPubMed Siddique MA, Nakanishi K, Taniguchi T, Grompe M, D'Andrea AD: Function of the Fanconi anemia pathway in Fanconi anemia complementation group F and D1 cells. Exp Hematol. 2001, 29: 1448-1455. 10.1016/S0301-472X(01)00754-8CrossRefPubMed
44.
go back to reference Yang Y, Kuang Y, Montes De Oca R, Hays T, Moreau L, Lu N, Seed B, D'Andrea AD: Targeted disruption of the murine Fanconi anemia gene, Fancg/Xrcc9. Blood. 2001, 98: 3435-3440. 10.1182/blood.V98.12.3435CrossRefPubMed Yang Y, Kuang Y, Montes De Oca R, Hays T, Moreau L, Lu N, Seed B, D'Andrea AD: Targeted disruption of the murine Fanconi anemia gene, Fancg/Xrcc9. Blood. 2001, 98: 3435-3440. 10.1182/blood.V98.12.3435CrossRefPubMed
Metadata
Title
CHK1 inhibition as a strategy for targeting fanconi anemia (FA) DNA repair pathway deficient tumors
Authors
Clark C Chen
Richard D Kennedy
Samuel Sidi
A Thomas Look
Alan D'Andrea
Publication date
01-12-2009
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2009
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-8-24

Other articles of this Issue 1/2009

Molecular Cancer 1/2009 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine