Skip to main content
Top
Published in: BMC Cancer 1/2018

Open Access 01-12-2018 | Research article

Combination of lutetium-177 labelled anti-L1CAM antibody chCE7 with the clinically relevant protein kinase inhibitor MK1775: a novel combination against human ovarian carcinoma

Authors: Dennis Lindenblatt, Nastassja Terraneo, Giovanni Pellegrini, Susan Cohrs, Philipp René Spycher, David Vukovic, Martin Béhé, Roger Schibli, Jürgen Grünberg

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

Protein kinase inhibitors (PKIs) are currently tested in clinical studies (phase I-III) as an alternative strategy against (recurrent) ovarian cancer. Besides their anti-tumour efficacy, several PKIs have also shown radiosensitizing effects when combined with external beam radiation. Based on these results we asked if the addition of PKIs offers a therapeutic opportunity to improve radioimmunotherapy (RIT) against ovarian cancer. Five PKIs (alisertib, MK1775, MK2206, saracatinib, temsirolimus) were chosen for cytotoxicity screenings based on their current clinical trials in the treatment of ovarian cancer and their influence on cell cycle regulation and DNA damage repair pathways. We combined selected PKIs with 177Lu-labelled anti-L1CAM monoclonal antibody chCE7 for our investigations.

Methods

PKIs cytotoxicity was determined via cell colony-forming assays. Biomarker of DNA double-strand breaks (DSBs, γH2A.X) was analysed by western blot and fluorescence microscopy. Flow cytometric measurements were performed to evaluate levels of apoptosis based on mono- or combination treatments. The best combination was used for in vivo combination therapy studies in nude mice with SKOV3ip and IGROV1 human ovarian cancer xenografts. Bonferroni correction was used to determine statistical significance for multiple comparisons.

Results

The highest cytotoxicity against both cell lines was observed for MK1775 and alisertib. Combinations including 177Lu-labelled mAb chCE7 and MK1775 decreased 177Lu-DOTA-chCE7 IC60-values 14-fold, compared to 6-fold, when the radioimmunoconjugate was combined with alisertib. The most effective PKI MK1775 was further evaluated and demonstrated synergistic effects in combination with 177Lu-DOTA-chCE7 against IGROV1 cells. Significantly higher amounts of DSBs were detected in IGROV1 cells after combination (91%) compared to either treatment alone (MK1775: 52%; radioimmunoconjugate: 72%; p < 0.0125). Early-apoptosis was significantly enhanced in IGROV1 cells correlating with induced DSBs (177Lu-DOTA-chCE7: 8%, MK1775: 28%, 177Lu-DOTA-chCE7 + MK1775: 40%, p < 0.0125). Immunohistochemistry analysis of γH2A.X expression levels after therapy in SKOV3ip xenografts revealed a high sensitivity of the tumour cells to MK1775 and a high radioresistance. A prominent effect of tumour growth inhibition of the RIT and of the combination therapy was observed in vivo in a late stage IGROV1 xenograft model.

Conclusions

Our results warrant further evaluation of combination of MK1775 and radioimmunotherapy.
Appendix
Available only for authorised users
Literature
1.
go back to reference Jelovac D, Armstrong DK. Recent Progress in the diagnosis and treatment of ovarian Cancer. Cancer J Clin. 2011;61(3):183–203.CrossRef Jelovac D, Armstrong DK. Recent Progress in the diagnosis and treatment of ovarian Cancer. Cancer J Clin. 2011;61(3):183–203.CrossRef
2.
go back to reference McGuire WP. Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IV ovarian cancer (vol 334, pg 1, 1996). New Engl J Med. 1996, 334(16):1070–0. McGuire WP. Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IV ovarian cancer (vol 334, pg 1, 1996). New Engl J Med. 1996, 334(16):1070–0.
3.
go back to reference Guarneri V, Piacentini F, Barbieri E, Conte PF. Achievements and unmet needs in the management of advanced ovarian cancer. Gynecol Oncol. 2010;117(2):152–8.CrossRef Guarneri V, Piacentini F, Barbieri E, Conte PF. Achievements and unmet needs in the management of advanced ovarian cancer. Gynecol Oncol. 2010;117(2):152–8.CrossRef
4.
go back to reference Ferrandina G, Ludovisi M, Lorusso D, Pignata S, Breda E, Savarese A, Medico PD, Scaltriti L, Katsaros D, Priolo D, et al. Phase III trial of gemcitabine compared with Pegylated liposomal doxorubicin in progressive or recurrent ovarian Cancer. J Clin Oncol. 2008;26(6):890–6.CrossRef Ferrandina G, Ludovisi M, Lorusso D, Pignata S, Breda E, Savarese A, Medico PD, Scaltriti L, Katsaros D, Priolo D, et al. Phase III trial of gemcitabine compared with Pegylated liposomal doxorubicin in progressive or recurrent ovarian Cancer. J Clin Oncol. 2008;26(6):890–6.CrossRef
5.
go back to reference Huinink WT, Lane SR, Ross GA, Grp ITS. Long-term survival in a phase III, randomised study of topotecan versus paclitaxel in advanced epithelial ovarian carcinoma. Ann Oncol. 2004;15(1):100–3.CrossRef Huinink WT, Lane SR, Ross GA, Grp ITS. Long-term survival in a phase III, randomised study of topotecan versus paclitaxel in advanced epithelial ovarian carcinoma. Ann Oncol. 2004;15(1):100–3.CrossRef
6.
go back to reference Rose PG, Blessing JA, Mayer AR, Homesley HD. Prolonged oral etoposide as second-line therapy for platinum-resistant and platinum-sensitive ovarian carcinoma: a gynecologic oncology group study. J Clin Oncol. 1998;16(2):405–10.CrossRef Rose PG, Blessing JA, Mayer AR, Homesley HD. Prolonged oral etoposide as second-line therapy for platinum-resistant and platinum-sensitive ovarian carcinoma: a gynecologic oncology group study. J Clin Oncol. 1998;16(2):405–10.CrossRef
7.
go back to reference Itamochi H, Oishi T, Shimada M, Sato S, Uegaki K, Naniwa J, Sato S, Nonaka M, Terakawa N, Kigawa J, et al. Inhibiting the mTOR pathway synergistically enhances cytotoxicity in ovarian Cancer cells induced by etoposide through upregulation of c-Jun. Clin Cancer Res. 2011;17(14):4742–50.CrossRef Itamochi H, Oishi T, Shimada M, Sato S, Uegaki K, Naniwa J, Sato S, Nonaka M, Terakawa N, Kigawa J, et al. Inhibiting the mTOR pathway synergistically enhances cytotoxicity in ovarian Cancer cells induced by etoposide through upregulation of c-Jun. Clin Cancer Res. 2011;17(14):4742–50.CrossRef
8.
go back to reference Banerjee S, Kaye SB. New strategies in the treatment of ovarian Cancer: current clinical perspectives and future potential. Clin Cancer Res. 2013;19(5):961–8.CrossRef Banerjee S, Kaye SB. New strategies in the treatment of ovarian Cancer: current clinical perspectives and future potential. Clin Cancer Res. 2013;19(5):961–8.CrossRef
9.
go back to reference Frederick PJ, Straughn JM, Alvarez RD, Buchsbaum DJ. Preclinical studies and clinical utilization of monoclonal antibodies in epithelial ovarian cancer. Gynecol Oncol. 2009;113(3):384–90.CrossRef Frederick PJ, Straughn JM, Alvarez RD, Buchsbaum DJ. Preclinical studies and clinical utilization of monoclonal antibodies in epithelial ovarian cancer. Gynecol Oncol. 2009;113(3):384–90.CrossRef
10.
go back to reference Berek J, Taylor P, McGuire W, Smith LM, Schultes B, Nicodemus CF. Oregovomab maintenance Monoimmunotherapy does not improve outcomes in advanced ovarian Cancer. J Clin Oncol. 2009;27(3):418–25.CrossRef Berek J, Taylor P, McGuire W, Smith LM, Schultes B, Nicodemus CF. Oregovomab maintenance Monoimmunotherapy does not improve outcomes in advanced ovarian Cancer. J Clin Oncol. 2009;27(3):418–25.CrossRef
11.
go back to reference Schilder RJ, Pathak HB, Lokshin AE, Holloway RW, Alvarez RD, Aghajanian C, Min H, Devarajan K, Ross E, Drescher CW, et al. Phase II trial of single agent cetuximab in patients with persistent or recurrent epithelial ovarian or primary peritoneal carcinoma with the potential for dose escalation to rash. Gynecol Oncol. 2009;113(1):21–7.CrossRef Schilder RJ, Pathak HB, Lokshin AE, Holloway RW, Alvarez RD, Aghajanian C, Min H, Devarajan K, Ross E, Drescher CW, et al. Phase II trial of single agent cetuximab in patients with persistent or recurrent epithelial ovarian or primary peritoneal carcinoma with the potential for dose escalation to rash. Gynecol Oncol. 2009;113(1):21–7.CrossRef
12.
go back to reference Bookman MA, Darcy KM, Clarke-Pearson D, Boothby RA, Horowitz IR. Evaluation of monoclonal humanized anti-HER2 antibody, Trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the gynecologic oncology group. J Clin Oncol. 2003;21(2):283–90.CrossRef Bookman MA, Darcy KM, Clarke-Pearson D, Boothby RA, Horowitz IR. Evaluation of monoclonal humanized anti-HER2 antibody, Trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the gynecologic oncology group. J Clin Oncol. 2003;21(2):283–90.CrossRef
13.
go back to reference Leone Roberti Maggiore U, Bellati F, Ruscito I, Gasparri ML, Alessandri F, Venturini PL, Ferrero S. Monoclonal antibodies therapies for ovarian cancer. Expert Opin Biol Ther. 2013;13(5):739–64.CrossRef Leone Roberti Maggiore U, Bellati F, Ruscito I, Gasparri ML, Alessandri F, Venturini PL, Ferrero S. Monoclonal antibodies therapies for ovarian cancer. Expert Opin Biol Ther. 2013;13(5):739–64.CrossRef
14.
go back to reference Hong X, O’Donnell JP, Salazar CR, Van Brocklyn JR, Barnett KD, Pearl DK, deCarvalho AC, Ecsedy JA, Brown SL, Mikkelsen T, et al. The selective Aurora-a kinase inhibitor MLN8237 (alisertib) potently inhibits proliferation of glioblastoma neurosphere tumor stem-like cells and potentiates the effects of temozolomide and ionizing radiation. Cancer Chemother Pharmacol. 2014;73(5):983–90.PubMedPubMedCentral Hong X, O’Donnell JP, Salazar CR, Van Brocklyn JR, Barnett KD, Pearl DK, deCarvalho AC, Ecsedy JA, Brown SL, Mikkelsen T, et al. The selective Aurora-a kinase inhibitor MLN8237 (alisertib) potently inhibits proliferation of glioblastoma neurosphere tumor stem-like cells and potentiates the effects of temozolomide and ionizing radiation. Cancer Chemother Pharmacol. 2014;73(5):983–90.PubMedPubMedCentral
15.
go back to reference Carrassa L, Chilà R, Lupi M, Ricci F, Celenza C, Mazzoletti M, Broggini M, Damia G. Combined inhibition of Chk1 and Wee1: in vitro synergistic effect translates to tumor growth inhibition in vivo. Cell Cycle. 2012;11(13):2507–17.CrossRef Carrassa L, Chilà R, Lupi M, Ricci F, Celenza C, Mazzoletti M, Broggini M, Damia G. Combined inhibition of Chk1 and Wee1: in vitro synergistic effect translates to tumor growth inhibition in vivo. Cell Cycle. 2012;11(13):2507–17.CrossRef
16.
go back to reference Purnell PR, Mack PC, Tepper CG, Evans CP, Green TP, Gumerlock PH, Lara PN, Gandara DR, Kung H-J, Gautschi O. The Src inhibitor AZD0530 blocks invasion and may act as a Radiosensitizer in lung Cancer cells. J Thorac Oncol. 2009;4(4):448–54.CrossRef Purnell PR, Mack PC, Tepper CG, Evans CP, Green TP, Gumerlock PH, Lara PN, Gandara DR, Kung H-J, Gautschi O. The Src inhibitor AZD0530 blocks invasion and may act as a Radiosensitizer in lung Cancer cells. J Thorac Oncol. 2009;4(4):448–54.CrossRef
17.
go back to reference Fischer E, Grünberg J, Cohrs S, Hohn A, Waldner-Knogler K, Jeger S, Zimmermann K, Novak-Hofer I, Schibli R. L1-CAM-targeted antibody therapy and 177Lu-radioimmunotherapy of disseminated ovarian cancer. Int J Cancer. 2012;130(11):2715–21.CrossRef Fischer E, Grünberg J, Cohrs S, Hohn A, Waldner-Knogler K, Jeger S, Zimmermann K, Novak-Hofer I, Schibli R. L1-CAM-targeted antibody therapy and 177Lu-radioimmunotherapy of disseminated ovarian cancer. Int J Cancer. 2012;130(11):2715–21.CrossRef
18.
go back to reference Knogler K, Grünberg J, Zimmermann K, Cohrs S, Honer M, Ametamey S, Altevogt P, Fogel M, Schubiger PA, Novak-Hofer I. Copper-67 Radioimmunotherapy and growth inhibition by anti–L1-cell adhesion molecule monoclonal antibodies in a therapy model of ovarian Cancer metastasis. Clin Cancer Res. 2007;13(2):603–11.CrossRef Knogler K, Grünberg J, Zimmermann K, Cohrs S, Honer M, Ametamey S, Altevogt P, Fogel M, Schubiger PA, Novak-Hofer I. Copper-67 Radioimmunotherapy and growth inhibition by anti–L1-cell adhesion molecule monoclonal antibodies in a therapy model of ovarian Cancer metastasis. Clin Cancer Res. 2007;13(2):603–11.CrossRef
19.
go back to reference Lindenblatt D, Fischer E, Cohrs S, Schibli R, Grünberg J. Paclitaxel improved anti-L1CAM lutetium-177 radioimmunotherapy in an ovarian cancer xenograft model. EJNMMI Res. 2014;4(1):54.CrossRef Lindenblatt D, Fischer E, Cohrs S, Schibli R, Grünberg J. Paclitaxel improved anti-L1CAM lutetium-177 radioimmunotherapy in an ovarian cancer xenograft model. EJNMMI Res. 2014;4(1):54.CrossRef
20.
go back to reference Grünberg J, Lindenblatt D, Dorrer H, Cohrs S, Zhernosekov K, Koster U, Turler A, Fischer E, Schibli R. Anti-L1CAM radioimmunotherapy is more effective with the radiolanthanide terbium-161 compared to lutetium-177 in an ovarian cancer model. Eur J Nucl Med Mol Imaging. 2014;41(10):1907–15.CrossRef Grünberg J, Lindenblatt D, Dorrer H, Cohrs S, Zhernosekov K, Koster U, Turler A, Fischer E, Schibli R. Anti-L1CAM radioimmunotherapy is more effective with the radiolanthanide terbium-161 compared to lutetium-177 in an ovarian cancer model. Eur J Nucl Med Mol Imaging. 2014;41(10):1907–15.CrossRef
21.
go back to reference Fogel M, Gutwein P, Mechtersheimer S, Riedle S, Stoeck A, Smirnov A, Edler L, Ben-Arie A, Huszar M, Altevogt P. L1 expression as a predictor of progression and survival in patients with uterine and ovarian carcinomas. Lancet. 2003;362(9387):869–75.CrossRef Fogel M, Gutwein P, Mechtersheimer S, Riedle S, Stoeck A, Smirnov A, Edler L, Ben-Arie A, Huszar M, Altevogt P. L1 expression as a predictor of progression and survival in patients with uterine and ovarian carcinomas. Lancet. 2003;362(9387):869–75.CrossRef
22.
go back to reference Kiefel H, Bondong S, Hazin J, Ridinger J, Schirmer U, Riedle S, Altevogt P. L1CAM: a major driver for tumor cell invasion and motility. Cell Adhes Migr. 2012;6(4):374–84.CrossRef Kiefel H, Bondong S, Hazin J, Ridinger J, Schirmer U, Riedle S, Altevogt P. L1CAM: a major driver for tumor cell invasion and motility. Cell Adhes Migr. 2012;6(4):374–84.CrossRef
23.
go back to reference Matulonis UA, Sharma S, Ghamande S, Gordon MS, Del Prete SA, Ray-Coquard I, Kutarska E, Liu H, Fingert H, Zhou X, et al. Phase II study of MLN8237 (alisertib), an investigational Aurora a kinase inhibitor, in patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. Gynecol Oncol. 2012;127(1):63–9.CrossRef Matulonis UA, Sharma S, Ghamande S, Gordon MS, Del Prete SA, Ray-Coquard I, Kutarska E, Liu H, Fingert H, Zhou X, et al. Phase II study of MLN8237 (alisertib), an investigational Aurora a kinase inhibitor, in patients with platinum-resistant or -refractory epithelial ovarian, fallopian tube, or primary peritoneal carcinoma. Gynecol Oncol. 2012;127(1):63–9.CrossRef
24.
go back to reference Parker L, Piwnica-Worms H. Inactivation of the p34cdc2-cyclin B complex by the human WEE1 tyrosine kinase. Science. 1992;257(5078):1955–7.CrossRef Parker L, Piwnica-Worms H. Inactivation of the p34cdc2-cyclin B complex by the human WEE1 tyrosine kinase. Science. 1992;257(5078):1955–7.CrossRef
25.
go back to reference Watanabe N, Broome M, Hunter T. Regulation of the human WEE1Hu CDK tyrosine 15-kinase during the cell cycle. EMBO J. 1995;14(9):1878–91.CrossRef Watanabe N, Broome M, Hunter T. Regulation of the human WEE1Hu CDK tyrosine 15-kinase during the cell cycle. EMBO J. 1995;14(9):1878–91.CrossRef
26.
go back to reference De Witt Hamer PC, Mir SE, Noske D, Van Noorden CJ, Wurdinger T. WEE1 kinase targeting combined with DNA-damaging cancer therapy catalyzes mitotic catastrophe. Clin Cancer Res. 2011;17(13):4200–7.CrossRef De Witt Hamer PC, Mir SE, Noske D, Van Noorden CJ, Wurdinger T. WEE1 kinase targeting combined with DNA-damaging cancer therapy catalyzes mitotic catastrophe. Clin Cancer Res. 2011;17(13):4200–7.CrossRef
27.
go back to reference Caretti V, Hiddingh L, Lagerweij T, Schellen P, Koken PW, Hulleman E, van Vuurden DG, Vandertop WP, Kaspers GJ, Noske DP, et al. WEE1 kinase inhibition enhances the radiation response of diffuse intrinsic pontine gliomas. Mol Cancer Ther. 2013;12(2):141–50.CrossRef Caretti V, Hiddingh L, Lagerweij T, Schellen P, Koken PW, Hulleman E, van Vuurden DG, Vandertop WP, Kaspers GJ, Noske DP, et al. WEE1 kinase inhibition enhances the radiation response of diffuse intrinsic pontine gliomas. Mol Cancer Ther. 2013;12(2):141–50.CrossRef
28.
go back to reference Behbakht K, Sill MW, Darcy KM, Rubin SC, Mannel RS, Waggoner S, Schilder RJ, Cai KQ, Godwin AK, Alpaugh RK. Phase II trial of the mTOR inhibitor, temsirolimus and evaluation of circulating tumor cells and tumor biomarkers in persistent and recurrent epithelial ovarian and primary peritoneal malignancies: a gynecologic oncology group study. Gynecol Oncol. 2011;123(1):19–26.CrossRef Behbakht K, Sill MW, Darcy KM, Rubin SC, Mannel RS, Waggoner S, Schilder RJ, Cai KQ, Godwin AK, Alpaugh RK. Phase II trial of the mTOR inhibitor, temsirolimus and evaluation of circulating tumor cells and tumor biomarkers in persistent and recurrent epithelial ovarian and primary peritoneal malignancies: a gynecologic oncology group study. Gynecol Oncol. 2011;123(1):19–26.CrossRef
29.
go back to reference Emons G, Kurzeder C, Schmalfeldt B, Neuser P, de Gregorio N, Pfisterer J, Park-Simon TW, Mahner S, Schroder W, Luck HJ, et al. Temsirolimus in women with platinum-refractory/resistant ovarian cancer or advanced/recurrent endometrial carcinoma. A phase II study of the AGO-study group (AGO-GYN8). Gynecol Oncol. 2016;140(3):450–6.CrossRef Emons G, Kurzeder C, Schmalfeldt B, Neuser P, de Gregorio N, Pfisterer J, Park-Simon TW, Mahner S, Schroder W, Luck HJ, et al. Temsirolimus in women with platinum-refractory/resistant ovarian cancer or advanced/recurrent endometrial carcinoma. A phase II study of the AGO-study group (AGO-GYN8). Gynecol Oncol. 2016;140(3):450–6.CrossRef
30.
go back to reference Gil del Alcazar CR, Hardebeck MC, Mukherjee B, Tomimatsu N, Gao X, Yan J, Xie XJ, Bachoo R, Li L, Habib AA, et al. Inhibition of DNA double-strand break repair by the dual PI3K/mTOR inhibitor NVP-BEZ235 as a strategy for radiosensitization of glioblastoma. Clin Cancer Res. 2014;20(5):1235–48.CrossRef Gil del Alcazar CR, Hardebeck MC, Mukherjee B, Tomimatsu N, Gao X, Yan J, Xie XJ, Bachoo R, Li L, Habib AA, et al. Inhibition of DNA double-strand break repair by the dual PI3K/mTOR inhibitor NVP-BEZ235 as a strategy for radiosensitization of glioblastoma. Clin Cancer Res. 2014;20(5):1235–48.CrossRef
31.
go back to reference Hirai H, Sootome H, Nakatsuru Y, Miyama K, Taguchi S, Tsujioka K, Ueno Y, Hatch H, Majumder PK, Pan BS, et al. MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard chemotherapeutic agents or molecular targeted drugs in vitro and in vivo. Mol Cancer Ther. 2010;9(7):1956–67.CrossRef Hirai H, Sootome H, Nakatsuru Y, Miyama K, Taguchi S, Tsujioka K, Ueno Y, Hatch H, Majumder PK, Pan BS, et al. MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard chemotherapeutic agents or molecular targeted drugs in vitro and in vivo. Mol Cancer Ther. 2010;9(7):1956–67.CrossRef
32.
go back to reference Connolly E, Sun Y, Chao K, Hei T. Abstract PD09-09: the Akt inhibitor MK-2206 is an effective radio-sensitizer of p53 deficient triple negative breast cancer (TNBC) cells. Cancer Res. 2012;72(24 Supplement):PD09–09.CrossRef Connolly E, Sun Y, Chao K, Hei T. Abstract PD09-09: the Akt inhibitor MK-2206 is an effective radio-sensitizer of p53 deficient triple negative breast cancer (TNBC) cells. Cancer Res. 2012;72(24 Supplement):PD09–09.CrossRef
33.
go back to reference Yu D, Wolf JK, Scanlon M, Price JE, Hung MC. Enhanced c-erbB-2/neu expression in human ovarian cancer cells correlates with more severe malignancy that can be suppressed by E1A. Cancer Res. 1993;53(4):891–8.PubMed Yu D, Wolf JK, Scanlon M, Price JE, Hung MC. Enhanced c-erbB-2/neu expression in human ovarian cancer cells correlates with more severe malignancy that can be suppressed by E1A. Cancer Res. 1993;53(4):891–8.PubMed
34.
go back to reference Grünberg J, Knogler K, Waibel R, Novak-Hofer I. High-yield production of recombinant antibody fragments in HEK-293 cells using sodium butyrate. BioTechniques. 2003;34(5):968–72.CrossRef Grünberg J, Knogler K, Waibel R, Novak-Hofer I. High-yield production of recombinant antibody fragments in HEK-293 cells using sodium butyrate. BioTechniques. 2003;34(5):968–72.CrossRef
35.
go back to reference Lindmo T, Boven E, Cuttitta F, Fedorko J, Bunn PA,J. Determination of the immunoreactive fraction of radiolabeled monoclonal antibodies by linear extrapolation to binding at infinite antigen excess. J Immunol Methods. 1984;72(1):77–89.CrossRef Lindmo T, Boven E, Cuttitta F, Fedorko J, Bunn PA,J. Determination of the immunoreactive fraction of radiolabeled monoclonal antibodies by linear extrapolation to binding at infinite antigen excess. J Immunol Methods. 1984;72(1):77–89.CrossRef
36.
go back to reference Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315–9.CrossRef Franken NA, Rodermond HM, Stap J, Haveman J, van Bree C. Clonogenic assay of cells in vitro. Nat Protoc. 2006;1(5):2315–9.CrossRef
37.
go back to reference Moser R, Xu C, Kao M, Annis J, Lerma LA, Schaupp CM, Gurley KE, Jang IS, Biktasova A, Yarbrough WG, et al. Functional kinomics identifies candidate therapeutic targets in head and neck cancer. Clin Cancer Res. 2014;20(16):4274–88.CrossRef Moser R, Xu C, Kao M, Annis J, Lerma LA, Schaupp CM, Gurley KE, Jang IS, Biktasova A, Yarbrough WG, et al. Functional kinomics identifies candidate therapeutic targets in head and neck cancer. Clin Cancer Res. 2014;20(16):4274–88.CrossRef
38.
go back to reference Hoggatt AF, Hoggatt J, Honerlaw M, Pelus LM. A spoonful of sugar helps the medicine go down: a novel technique to improve oral gavage in mice. J Amer Assoc Lab Anim Sci. 2010;49(3):329–34. Hoggatt AF, Hoggatt J, Honerlaw M, Pelus LM. A spoonful of sugar helps the medicine go down: a novel technique to improve oral gavage in mice. J Amer Assoc Lab Anim Sci. 2010;49(3):329–34.
39.
go back to reference Zhao L, Au JL, Wientjes MG. Comparison of methods for evaluating drug-drug interaction. Front Biosci. 2010;2:241–9. Zhao L, Au JL, Wientjes MG. Comparison of methods for evaluating drug-drug interaction. Front Biosci. 2010;2:241–9.
40.
go back to reference Geenen JJJ, Schellens JHM. Molecular Pathways: Targeting the Protein Kinase Wee1 in Cancer. Clin Cancer Res. 2017;23(16):4540–544.CrossRef Geenen JJJ, Schellens JHM. Molecular Pathways: Targeting the Protein Kinase Wee1 in Cancer. Clin Cancer Res. 2017;23(16):4540–544.CrossRef
41.
go back to reference Karnak D, Engelke CG, Parsels LA, Kausar T, Wei D, Robertson JR, Marsh KB, Davis MA, Zhao L, Maybaum J, et al. Combined inhibition of Wee1 and PARP1/2 for radiosensitization in pancreatic cancer. Clin Cancer Res. 2014;20(19):5085–96.CrossRef Karnak D, Engelke CG, Parsels LA, Kausar T, Wei D, Robertson JR, Marsh KB, Davis MA, Zhao L, Maybaum J, et al. Combined inhibition of Wee1 and PARP1/2 for radiosensitization in pancreatic cancer. Clin Cancer Res. 2014;20(19):5085–96.CrossRef
42.
go back to reference Hirai H, Arai T, Okada M, Nishibata T, Kobayashi M, Sakai N, Imagaki K, Ohtani J, Sakai T, Yoshizumi T, et al. MK-1775, a small molecule Wee1 inhibitor, enhances anti-tumor efficacy of various DNA-damaging agents, including 5-fluorouracil. Cancer Biol Ther. 2010;9(7):514–22.CrossRef Hirai H, Arai T, Okada M, Nishibata T, Kobayashi M, Sakai N, Imagaki K, Ohtani J, Sakai T, Yoshizumi T, et al. MK-1775, a small molecule Wee1 inhibitor, enhances anti-tumor efficacy of various DNA-damaging agents, including 5-fluorouracil. Cancer Biol Ther. 2010;9(7):514–22.CrossRef
43.
go back to reference PosthumaDeBoer J, Wurdinger T, Graat HC, van Beusechem VW, Helder MN, van Royen BJ, Kaspers GJ. WEE1 inhibition sensitizes osteosarcoma to radiotherapy. BMC Cancer. 2011;11:156.CrossRef PosthumaDeBoer J, Wurdinger T, Graat HC, van Beusechem VW, Helder MN, van Royen BJ, Kaspers GJ. WEE1 inhibition sensitizes osteosarcoma to radiotherapy. BMC Cancer. 2011;11:156.CrossRef
44.
go back to reference Kim MJ, Lee JY, Lee SJ. Transient suppression of nuclear Cdc2 activity in response to ionizing radiation. Oncol Rep. 2008;19(5):1323–9.PubMed Kim MJ, Lee JY, Lee SJ. Transient suppression of nuclear Cdc2 activity in response to ionizing radiation. Oncol Rep. 2008;19(5):1323–9.PubMed
45.
go back to reference Sharma A, Singh K, Almasan A. Histone H2AX phosphorylation: a marker for DNA damage. Methods Mol Biol. 2012;920:613–26.CrossRef Sharma A, Singh K, Almasan A. Histone H2AX phosphorylation: a marker for DNA damage. Methods Mol Biol. 2012;920:613–26.CrossRef
46.
go back to reference Beck H, Nahse-Kumpf V, Larsen MS, O'Hanlon KA, Patzke S, Holmberg C, Mejlvang J, Groth A, Nielsen O, Syljuasen RG, et al. Cyclin-dependent kinase suppression by WEE1 kinase protects the genome through control of replication initiation and nucleotide consumption. Mol Cell Biol. 2012;32(20):4226–36.CrossRef Beck H, Nahse-Kumpf V, Larsen MS, O'Hanlon KA, Patzke S, Holmberg C, Mejlvang J, Groth A, Nielsen O, Syljuasen RG, et al. Cyclin-dependent kinase suppression by WEE1 kinase protects the genome through control of replication initiation and nucleotide consumption. Mol Cell Biol. 2012;32(20):4226–36.CrossRef
47.
go back to reference Dominguez-Kelly R, Martin Y, Koundrioukoff S, Tanenbaum ME, Smits VA, Medema RH, Debatisse M, Freire R. Wee1 controls genomic stability during replication by regulating the Mus81-Eme1 endonuclease. J Cell Biol. 2011;194(4):567–79.CrossRef Dominguez-Kelly R, Martin Y, Koundrioukoff S, Tanenbaum ME, Smits VA, Medema RH, Debatisse M, Freire R. Wee1 controls genomic stability during replication by regulating the Mus81-Eme1 endonuclease. J Cell Biol. 2011;194(4):567–79.CrossRef
48.
go back to reference Petru E, Sevin BU, Gottlieb Ch. Radiosensitivity Patterns of Four Human Ovarian Cancer Cell Linesin Vitro. Gynecol Oncol. 1997;64(3):490–92.CrossRef Petru E, Sevin BU, Gottlieb Ch. Radiosensitivity Patterns of Four Human Ovarian Cancer Cell Linesin Vitro. Gynecol Oncol. 1997;64(3):490–92.CrossRef
49.
go back to reference Cojoc M, Mäbert K, Muders MH, Dubrovska A. A role for cancer stem cells in therapy resistance: Cellular and molecular mechanisms. Semin Cancer Biol. 2015;31:16–27.CrossRef Cojoc M, Mäbert K, Muders MH, Dubrovska A. A role for cancer stem cells in therapy resistance: Cellular and molecular mechanisms. Semin Cancer Biol. 2015;31:16–27.CrossRef
50.
go back to reference Sarcar B, Kahali S, Prabhu AH, Shumway SD, Xu Y, Demuth T, Chinnaiyan P. Targeting Radiation-Induced G2 Checkpoint Activation with the Wee-1 Inhibitor MK-1775 in Glioblastoma Cell Lines. Mol Cancer Ther. 2011;10(12):2405–414.CrossRef Sarcar B, Kahali S, Prabhu AH, Shumway SD, Xu Y, Demuth T, Chinnaiyan P. Targeting Radiation-Induced G2 Checkpoint Activation with the Wee-1 Inhibitor MK-1775 in Glioblastoma Cell Lines. Mol Cancer Ther. 2011;10(12):2405–414.CrossRef
51.
go back to reference Rubin RH, Young LS, Hansen WP, Nedelman M, Wilkinson R, Nelles MJ, Callahan R, Khaw BA, Strauss HW: Specific and nonspecific imaging of localized Fisher immunotype 1 Pseudomonas aeruginosa infection with radiolabeled monoclonal antibody. J Nucl Med. 1988;29(5):651–56. Rubin RH, Young LS, Hansen WP, Nedelman M, Wilkinson R, Nelles MJ, Callahan R, Khaw BA, Strauss HW: Specific and nonspecific imaging of localized Fisher immunotype 1 Pseudomonas aeruginosa infection with radiolabeled monoclonal antibody. J Nucl Med. 1988;29(5):651–56.
52.
go back to reference Lisa M. Coussens, Zena Werb. Inflammation and cancer. Nature. 2002;420 (6917):860–67.CrossRef Lisa M. Coussens, Zena Werb. Inflammation and cancer. Nature. 2002;420 (6917):860–67.CrossRef
Metadata
Title
Combination of lutetium-177 labelled anti-L1CAM antibody chCE7 with the clinically relevant protein kinase inhibitor MK1775: a novel combination against human ovarian carcinoma
Authors
Dennis Lindenblatt
Nastassja Terraneo
Giovanni Pellegrini
Susan Cohrs
Philipp René Spycher
David Vukovic
Martin Béhé
Roger Schibli
Jürgen Grünberg
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4836-1

Other articles of this Issue 1/2018

BMC Cancer 1/2018 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