Skip to main content
Erschienen in: Breast Cancer Research and Treatment 1/2014

01.11.2014 | Epidemiology

Dual specificity phosphatase 4 gene expression in association with triple-negative breast cancer outcome

verfasst von: Michelle L. Baglia, Qiuyin Cai, Ying Zheng, Jie Wu, Yinghao Su, Fei Ye, Ping-Ping Bao, Hui Cai, Zhiguo Zhao, Justin Balko, Wei Zheng, Wei Lu, Xiao-Ou Shu

Erschienen in: Breast Cancer Research and Treatment | Ausgabe 1/2014

Einloggen, um Zugang zu erhalten

Abstract

Triple-negative breast cancer (TNBC) is an aggressive cancer with limited treatment options. Dual specificity phosphatase 4 (DUSP4) has recently been suggested as a potential marker of chemotherapy resistance for TNBC. DUSP4 gene expression levels were measured in breast cancer tissue from 469 TNBC patients aged 20–75 years who participated in the Shanghai Breast Cancer Survival Study, and their association with recurrence/breast cancer mortality and total mortality was evaluated. Information on breast cancer diagnosis, treatment, and disease progression was collected via medical chart review and multiple in-person follow-up surveys. A Cox regression model was applied in the data analyses. Over a median follow-up of 5.3 years (range: 0.7–8.9 years), 100 deaths and 92 recurrences/breast cancer deaths were documented. Expression levels of transcript variant 1 (NM_001394) and transcript variant 2 (NM_057158) of the DUSP4 gene were studied and were highly correlated (r = 0.76). Low DUSP4 expression levels, particularly of variant 1, were associated with both increased recurrence/breast cancer mortality and increased overall mortality. Hazard ratios with adjustment for age at diagnosis and TNM stage associated with below versus above the median expression level were 1.97 (95 % confidence interval (CI): 1.27–3.05) for recurrence/breast cancer mortality and 2.09 (95 % CI: 1.38–3.17) for overall mortality. Additional adjustment for expression levels of MKI67 and TP53, common treatment types, breast cancer subtype, and grade did not materially alter the observed associations. Low DUSP4 expression levels predict recurrence and mortality in TNBC patients independently from known clinical and molecular predictors.
Literatur
3.
Zurück zum Zitat Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, Lickley LA, Rawlinson E, Sun P, Narod SA (2007) Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res 13(15 Pt 1):4429–4434. doi:10.1158/1078-0432.CCR-06-3045 PubMedCrossRef Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, Lickley LA, Rawlinson E, Sun P, Narod SA (2007) Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res 13(15 Pt 1):4429–4434. doi:10.​1158/​1078-0432.​CCR-06-3045 PubMedCrossRef
4.
Zurück zum Zitat Balko JM, Cook RS, Vaught DB, Kuba MG, Miller TW, Bhola NE, Sanders ME, Granja-Ingram NM, Smith JJ, Meszoely IM, Salter J, Dowsett M, Stemke-Hale K, Gonzalez-Angulo AM, Mills GB, Pinto JA, Gomez HL, Arteaga CL (2012) Profiling of residual breast cancers after neoadjuvant chemotherapy identifies DUSP4 deficiency as a mechanism of drug resistance. Nat Med 18(7):1052–1059. doi:10.1038/nm.2795 PubMedCrossRefPubMedCentral Balko JM, Cook RS, Vaught DB, Kuba MG, Miller TW, Bhola NE, Sanders ME, Granja-Ingram NM, Smith JJ, Meszoely IM, Salter J, Dowsett M, Stemke-Hale K, Gonzalez-Angulo AM, Mills GB, Pinto JA, Gomez HL, Arteaga CL (2012) Profiling of residual breast cancers after neoadjuvant chemotherapy identifies DUSP4 deficiency as a mechanism of drug resistance. Nat Med 18(7):1052–1059. doi:10.​1038/​nm.​2795 PubMedCrossRefPubMedCentral
5.
Zurück zum Zitat Jeffrey KL, Camps M, Rommel C, Mackay CR (2007) Targeting dual-specificity phosphatases: manipulating MAP kinase signalling and immune responses. Nat Rev Drug Discovery 6(5):391–403. doi:10.1038/nrd2289 CrossRef Jeffrey KL, Camps M, Rommel C, Mackay CR (2007) Targeting dual-specificity phosphatases: manipulating MAP kinase signalling and immune responses. Nat Rev Drug Discovery 6(5):391–403. doi:10.​1038/​nrd2289 CrossRef
6.
Zurück zum Zitat Lawan A, Al-Harthi S, Cadalbert L, McCluskey AG, Shweash M, Grassia G, Grant A, Boyd M, Currie S, Plevin R (2011) Deletion of the dual specific phosphatase-4 (DUSP-4) gene reveals an essential non-redundant role for MAP kinase phosphatase-2 (MKP-2) in proliferation and cell survival. J Biol Chem 286(15):12933–12943. doi:10.1074/jbc.M110.181370 PubMedCrossRefPubMedCentral Lawan A, Al-Harthi S, Cadalbert L, McCluskey AG, Shweash M, Grassia G, Grant A, Boyd M, Currie S, Plevin R (2011) Deletion of the dual specific phosphatase-4 (DUSP-4) gene reveals an essential non-redundant role for MAP kinase phosphatase-2 (MKP-2) in proliferation and cell survival. J Biol Chem 286(15):12933–12943. doi:10.​1074/​jbc.​M110.​181370 PubMedCrossRefPubMedCentral
7.
Zurück zum Zitat Waha A, Felsberg J, Hartmann W, von dem Knesebeck A, Mikeska T, Joos S, Wolter M, Koch A, Yan PS, Endl E, Wiestler OD, Reifenberger G, Pietsch T (2010) Epigenetic downregulation of mitogen-activated protein kinase phosphatase MKP-2 relieves its growth suppressive activity in glioma cells. Cancer Res 70(4):1689–1699. doi:10.1158/0008-5472.CAN-09-3218 PubMedCrossRef Waha A, Felsberg J, Hartmann W, von dem Knesebeck A, Mikeska T, Joos S, Wolter M, Koch A, Yan PS, Endl E, Wiestler OD, Reifenberger G, Pietsch T (2010) Epigenetic downregulation of mitogen-activated protein kinase phosphatase MKP-2 relieves its growth suppressive activity in glioma cells. Cancer Res 70(4):1689–1699. doi:10.​1158/​0008-5472.​CAN-09-3218 PubMedCrossRef
8.
Zurück zum Zitat Cagnol S, Rivard N (2013) Oncogenic KRAS and BRAF activation of the MEK/ERK signaling pathway promotes expression of dual-specificity phosphatase 4 (DUSP4/MKP2) resulting in nuclear ERK1/2 inhibition. Oncogene 32(5):564–576. doi:10.1038/onc.2012.88 PubMedCrossRef Cagnol S, Rivard N (2013) Oncogenic KRAS and BRAF activation of the MEK/ERK signaling pathway promotes expression of dual-specificity phosphatase 4 (DUSP4/MKP2) resulting in nuclear ERK1/2 inhibition. Oncogene 32(5):564–576. doi:10.​1038/​onc.​2012.​88 PubMedCrossRef
12.
Zurück zum Zitat Geiss GK, Bumgarner RE, Birditt B, Dahl T, Dowidar N, Dunaway DL, Fell HP, Ferree S, George RD, Grogan T, James JJ, Maysuria M, Mitton JD, Oliveri P, Osborn JL, Peng T, Ratcliffe AL, Webster PJ, Davidson EH, Hood L, Dimitrov K (2008) Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat Biotechnol 26(3):317–325. doi:10.1038/nbt1385 PubMedCrossRef Geiss GK, Bumgarner RE, Birditt B, Dahl T, Dowidar N, Dunaway DL, Fell HP, Ferree S, George RD, Grogan T, James JJ, Maysuria M, Mitton JD, Oliveri P, Osborn JL, Peng T, Ratcliffe AL, Webster PJ, Davidson EH, Hood L, Dimitrov K (2008) Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat Biotechnol 26(3):317–325. doi:10.​1038/​nbt1385 PubMedCrossRef
13.
Zurück zum Zitat Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T, Davies S, Fauron C, He X, Hu Z, Quackenbush JF, Stijleman IJ, Palazzo J, Marron JS, Nobel AB, Mardis E, Nielsen TO, Ellis MJ, Perou CM, Bernard PS (2009) Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 27(8):1160–1167. doi:10.1200/JCO.2008.18.1370 PubMedCrossRefPubMedCentral Parker JS, Mullins M, Cheang MC, Leung S, Voduc D, Vickery T, Davies S, Fauron C, He X, Hu Z, Quackenbush JF, Stijleman IJ, Palazzo J, Marron JS, Nobel AB, Mardis E, Nielsen TO, Ellis MJ, Perou CM, Bernard PS (2009) Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 27(8):1160–1167. doi:10.​1200/​JCO.​2008.​18.​1370 PubMedCrossRefPubMedCentral
15.
Zurück zum Zitat Guarneri V, Piacentini F, Ficarra G, Frassoldati A, D’Amico R, Giovannelli S, Maiorana A, Jovic G, Conte P (2009) A prognostic model based on nodal status and Ki-67 predicts the risk of recurrence and death in breast cancer patients with residual disease after preoperative chemotherapy. Ann Oncol Off J European Soc Med Oncol 20(7):1193–1198. doi:10.1093/annonc/mdn761 CrossRef Guarneri V, Piacentini F, Ficarra G, Frassoldati A, D’Amico R, Giovannelli S, Maiorana A, Jovic G, Conte P (2009) A prognostic model based on nodal status and Ki-67 predicts the risk of recurrence and death in breast cancer patients with residual disease after preoperative chemotherapy. Ann Oncol Off J European Soc Med Oncol 20(7):1193–1198. doi:10.​1093/​annonc/​mdn761 CrossRef
16.
Zurück zum Zitat Peng DJ, Zhou JY, Wu GS (2010) Post-translational regulation of mitogen-activated protein kinase phosphatase-2 (MKP-2) by ERK. Cell Cycle 9(23):4650–4655PubMedCrossRef Peng DJ, Zhou JY, Wu GS (2010) Post-translational regulation of mitogen-activated protein kinase phosphatase-2 (MKP-2) by ERK. Cell Cycle 9(23):4650–4655PubMedCrossRef
18.
Zurück zum Zitat Byler S, Goldgar S, Heerboth S, Leary M, Housman G, Moulton K, Sarkar S (2014) Genetic and epigenetic aspects of breast cancer progression and therapy. Anticancer Res 34(3):1071–1077PubMed Byler S, Goldgar S, Heerboth S, Leary M, Housman G, Moulton K, Sarkar S (2014) Genetic and epigenetic aspects of breast cancer progression and therapy. Anticancer Res 34(3):1071–1077PubMed
20.
Zurück zum Zitat Baquero MT, Lostritto K, Gustavson MD, Bassi KA, Appia F, Camp RL, Molinaro AM, Harris LN, Rimm DL (2011) Evaluation of prognostic and predictive value of microtubule associated protein tau in two independent cohorts. Breast Cancer Res 13(5):R85. doi:10.1186/bcr2937 PubMedCrossRefPubMedCentral Baquero MT, Lostritto K, Gustavson MD, Bassi KA, Appia F, Camp RL, Molinaro AM, Harris LN, Rimm DL (2011) Evaluation of prognostic and predictive value of microtubule associated protein tau in two independent cohorts. Breast Cancer Res 13(5):R85. doi:10.​1186/​bcr2937 PubMedCrossRefPubMedCentral
21.
Zurück zum Zitat van de Vijver MJ, He YD, van’t Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, Parrish M, Atsma D, Witteveen A, Glas A, Delahaye L, van der Velde T, Bartelink H, Rodenhuis S, Rutgers ET, Friend SH, Bernards R (2002) A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 347(25):1999–2009. doi:10.1056/NEJMoa021967 PubMedCrossRef van de Vijver MJ, He YD, van’t Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ, Parrish M, Atsma D, Witteveen A, Glas A, Delahaye L, van der Velde T, Bartelink H, Rodenhuis S, Rutgers ET, Friend SH, Bernards R (2002) A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 347(25):1999–2009. doi:10.​1056/​NEJMoa021967 PubMedCrossRef
Metadaten
Titel
Dual specificity phosphatase 4 gene expression in association with triple-negative breast cancer outcome
verfasst von
Michelle L. Baglia
Qiuyin Cai
Ying Zheng
Jie Wu
Yinghao Su
Fei Ye
Ping-Ping Bao
Hui Cai
Zhiguo Zhao
Justin Balko
Wei Zheng
Wei Lu
Xiao-Ou Shu
Publikationsdatum
01.11.2014
Verlag
Springer US
Erschienen in
Breast Cancer Research and Treatment / Ausgabe 1/2014
Print ISSN: 0167-6806
Elektronische ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-014-3127-z

Weitere Artikel der Ausgabe 1/2014

Breast Cancer Research and Treatment 1/2014 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.