Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2016

Open Access 01-12-2016 | Research

Biological evaluation of antibody-maytansinoid conjugates as a strategy of RON targeted drug delivery for treatment of non-small cell lung cancer

Authors: Liang Feng, Hang-Ping Yao, Sharad Sharma, Yong-Qing Zhou, Jianwei Zhou, Ruiwen Zhang, Ming-Hai Wang

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2016

Login to get access

Abstract

Background

Aberrant expression of the RON receptor tyrosine kinase, a member of the MET proto-oncogene family, in breast cancer and non-small cell lung cancer (NSCLC) has therapeutic implication. Here we evaluated the efficacy of a novel anti-RON antibody-drug maytansinoid conjugate Zt/g4-DM1 for treatment of breast and NSCLC xenograft tumors in mouse models and explored a treatment strategy by combination of Zt/g4-DM1 with chemotherapeutics to achieve the maximal therapeutic activity.

Methods

Mouse monoclonal antibody Zt/g4 (IgG1a/κ) specific to human RON was conjugated to DM1 via thioether linkage to form Zt/g4-DM1 with a drug-antibody ratio of 4:1. Several breast cancer and NSCLC cell lines, expressing different levels of RON, were used as the model. Immunofluorescence was used to determine Zt/g4-induced RON internalization. Flow cytometric analysis and cell viability assay were used to determine the effect of Zt-g4-DM1 on cell cycle and death. Mouse xenograft NSCLC models were used in vivo to determine the therapeutic efficacy of Zt/g4-DM1 alone or in combination with chemotherapeutics.

Results

In vitro, Zt/g4 treatment of breast cancer and NSCLC cells rapidly induced cell surface RON internalization, which results in intracellular delivery of DM1 sufficient to arrest cell cycle at G2/M phase, reduce cell viability, and cause massive cell death. In mouse tumor xenograft models, Zt/g4-DM1 at 20 mg/kg in a Q12 × 2 regimen effectively blocked breast cancer and NSCLC cell- mediated tumor growth. More than 95 % inhibition of tumor growth among three tumor xenograft models tested was achieved according to the measured tumor volume. The minimal dose to balance the tumor growth and inhibition (tumoristatic concentration) was established at 2.02 mg/kg for H2228, 1.94 mg/kg for H358 cell, and 6.25 mg/kg for T-47D cell-mediated xenograft tumors.

Conclusion

Zt/g4 is highly effective in RON-directed drug delivery for targeted inhibition of NSCLC cell-derived tumor growth in mouse xenograft models. This work provides the basis for clinical development of humanized Zt/g4-DM1 for potential cancer therapy in the future.
Literature
1.
go back to reference Ducry L, Stump B. Antibody-drug conjugates: linking cytotoxic payloads to monoclonal antibodies. Bioconjug Chem. 2010;21:5–13.CrossRefPubMed Ducry L, Stump B. Antibody-drug conjugates: linking cytotoxic payloads to monoclonal antibodies. Bioconjug Chem. 2010;21:5–13.CrossRefPubMed
2.
go back to reference Teicher BA, Chari RV. Antibody conjugate therapeutics: challenges and potential. Clin Cancer Res. 2011;17:6389–97.CrossRefPubMed Teicher BA, Chari RV. Antibody conjugate therapeutics: challenges and potential. Clin Cancer Res. 2011;17:6389–97.CrossRefPubMed
3.
4.
go back to reference Lewis Phillips GD, Li G, Dugger DL, Crocker LM, Parsons KL, Mai E, et al. Targeting HER2- positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008;68:9280–90.CrossRefPubMed Lewis Phillips GD, Li G, Dugger DL, Crocker LM, Parsons KL, Mai E, et al. Targeting HER2- positive breast cancer with trastuzumab-DM1, an antibody-cytotoxic drug conjugate. Cancer Res. 2008;68:9280–90.CrossRefPubMed
5.
go back to reference Lambert JM, Chari RV. Ado-trastuzumab Emtansine (T-DM1): an antibody-drug conjugate (ADC) for HER2-positive breast cancer. J Med Chem. 2014;57:6949–64.CrossRefPubMed Lambert JM, Chari RV. Ado-trastuzumab Emtansine (T-DM1): an antibody-drug conjugate (ADC) for HER2-positive breast cancer. J Med Chem. 2014;57:6949–64.CrossRefPubMed
6.
go back to reference Younes A, Bartlett NL, Leonard JP, Kennedy DA, Lynch CM, Sievers EL, Forero-Torres A. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N Engl J Med. 2010;363:1812–21.CrossRefPubMed Younes A, Bartlett NL, Leonard JP, Kennedy DA, Lynch CM, Sievers EL, Forero-Torres A. Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas. N Engl J Med. 2010;363:1812–21.CrossRefPubMed
7.
go back to reference de Claro RA, McGinn K, Kwitkowski V, Bullock J, Khandelwal A, Habtemariam B, Ouyang Y, Saber H, Lee K, Koti K, Rothmann M, Shapiro M, Borrego F, Clouse K, Chen XH, Brown J, Akinsanya L, Kane R, Kaminskas E, Farrell A, Pazdur R. U.S. Food and Drug Administration approval summary: brentuximab vedotin for the treatment of relapsed Hodgkin lymphoma or relapsed systemic anaplastic large-cell lymphoma. Clin Cancer Res. 2012;18:5845–9.CrossRefPubMed de Claro RA, McGinn K, Kwitkowski V, Bullock J, Khandelwal A, Habtemariam B, Ouyang Y, Saber H, Lee K, Koti K, Rothmann M, Shapiro M, Borrego F, Clouse K, Chen XH, Brown J, Akinsanya L, Kane R, Kaminskas E, Farrell A, Pazdur R. U.S. Food and Drug Administration approval summary: brentuximab vedotin for the treatment of relapsed Hodgkin lymphoma or relapsed systemic anaplastic large-cell lymphoma. Clin Cancer Res. 2012;18:5845–9.CrossRefPubMed
8.
go back to reference Wang X, Ma D, Olson WC, Heston WD. In vitro and in vivo responses of advanced prostate tumors to PSMA ADC, an auristatin-conjugated antibody to prostate-specific membrane antigen. Mol Cancer Ther. 2011;10:1728–39.CrossRefPubMed Wang X, Ma D, Olson WC, Heston WD. In vitro and in vivo responses of advanced prostate tumors to PSMA ADC, an auristatin-conjugated antibody to prostate-specific membrane antigen. Mol Cancer Ther. 2011;10:1728–39.CrossRefPubMed
9.
go back to reference Ma D, Hopf CE, Malewicz AD, Donovan GP, Senter PD, Goeckeler WF, Maddon PJ, Olson WC. Potent antitumor activity of an auristatin-conjugated, fully human monoclonal antibody to prostate-specific membrane antigen. Clin Cancer Res. 2006;12:2591–6.CrossRefPubMed Ma D, Hopf CE, Malewicz AD, Donovan GP, Senter PD, Goeckeler WF, Maddon PJ, Olson WC. Potent antitumor activity of an auristatin-conjugated, fully human monoclonal antibody to prostate-specific membrane antigen. Clin Cancer Res. 2006;12:2591–6.CrossRefPubMed
10.
go back to reference Damelin M, Bankovich A, Park A, Aguilar J, Anderson WC, Santaguida M, Aujay M, Fong S, Khandke K, Pulito V, Ernstoff E, Escarpe P, Bernstein J, Pysz MA, Zhong W, Upeslacis E, Lucas J, Lucas J, Nichols T, Loving K, Foord O, Hampl J, Stull R, Barletta F, Falahatpisheh H, Sapra P, Gerber HP, Dylla SJ. Anti-EFNA4 calicheamicin conjugates effectively target triple-negative breast and ovarian tumor-initiating cells to result in sustained tumor regressions. Clin Cancer Res. 2015. Epub ahead of print. Damelin M, Bankovich A, Park A, Aguilar J, Anderson WC, Santaguida M, Aujay M, Fong S, Khandke K, Pulito V, Ernstoff E, Escarpe P, Bernstein J, Pysz MA, Zhong W, Upeslacis E, Lucas J, Lucas J, Nichols T, Loving K, Foord O, Hampl J, Stull R, Barletta F, Falahatpisheh H, Sapra P, Gerber HP, Dylla SJ. Anti-EFNA4 calicheamicin conjugates effectively target triple-negative breast and ovarian tumor-initiating cells to result in sustained tumor regressions. Clin Cancer Res. 2015. Epub ahead of print.
11.
go back to reference Dornan D, Bennett F, Chen Y, Dennis M, Eaton D, Elkins K, French D, Go MA, Jack A, Junutula JR, Koeppen H, Lau J, McBride J, Rawstron A, Shi X, Yu N, Yu SF, Yue P, Zheng B, Ebens A, Polson AG. Therapeutic potential of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphoma. Blood. 2009;114:2721–9.CrossRefPubMed Dornan D, Bennett F, Chen Y, Dennis M, Eaton D, Elkins K, French D, Go MA, Jack A, Junutula JR, Koeppen H, Lau J, McBride J, Rawstron A, Shi X, Yu N, Yu SF, Yue P, Zheng B, Ebens A, Polson AG. Therapeutic potential of an anti-CD79b antibody-drug conjugate, anti-CD79b-vc-MMAE, for the treatment of non-Hodgkin lymphoma. Blood. 2009;114:2721–9.CrossRefPubMed
12.
go back to reference Pfeifer M, Zheng B, Erdmann T, Koeppen H, McCord R, Grau M, Staiger A, Chai A, Sandmann T, Madle H, Dörken B, Chu YW, Chen AI, Lebovic D, Salles GA, Czuczman MS, Palanca-Wessels MC, Press OW, Advani R, Morschhauser F, Cheson BD, Lenz P, Ott G, Polson AG, Mundt KE, Lenz G. Anti-CD22 and anti-CD79B antibody drug conjugates are active in different molecular diffuse large B-cell lymphoma subtypes. Leukemia. 2015;29:1578–86.CrossRefPubMed Pfeifer M, Zheng B, Erdmann T, Koeppen H, McCord R, Grau M, Staiger A, Chai A, Sandmann T, Madle H, Dörken B, Chu YW, Chen AI, Lebovic D, Salles GA, Czuczman MS, Palanca-Wessels MC, Press OW, Advani R, Morschhauser F, Cheson BD, Lenz P, Ott G, Polson AG, Mundt KE, Lenz G. Anti-CD22 and anti-CD79B antibody drug conjugates are active in different molecular diffuse large B-cell lymphoma subtypes. Leukemia. 2015;29:1578–86.CrossRefPubMed
13.
go back to reference Raji R, Guzzo F, Carrara L, Varughese J, Cocco E, Bellone S, Betti M, Todeschini P, Gasparrini S, Ratner E, Silasi D-A, Azodi M, Schwartz P, Rutherford TJ, Buza N, Pecorelli S, Santin AD. Uterine and ovarian carcinomas overexpressing trop-2 are sensitive to hRS7, a humanized anti-Trop-2 antibody. J Exp Clin Cancer Res. 2011;30:106–16.CrossRefPubMedPubMedCentral Raji R, Guzzo F, Carrara L, Varughese J, Cocco E, Bellone S, Betti M, Todeschini P, Gasparrini S, Ratner E, Silasi D-A, Azodi M, Schwartz P, Rutherford TJ, Buza N, Pecorelli S, Santin AD. Uterine and ovarian carcinomas overexpressing trop-2 are sensitive to hRS7, a humanized anti-Trop-2 antibody. J Exp Clin Cancer Res. 2011;30:106–16.CrossRefPubMedPubMedCentral
14.
go back to reference Asundi J, Crocker L, Tremayne J, Chang P, Sakanaka C, Tanguay J, Spencer S, Chalasani S, Luis E, Gascoigne K, Desai R, Raja R, Friedman BA, Haverty PM, Polakis P, Firestein R. An antibody-drug conjugate directed against lymphocyte antigen 6 complex, locus E (LY6E) provides robust tumor killing in a wide range of solid tumor malignancies. Clin Cancer Res. 2015;21:3252–62.CrossRefPubMed Asundi J, Crocker L, Tremayne J, Chang P, Sakanaka C, Tanguay J, Spencer S, Chalasani S, Luis E, Gascoigne K, Desai R, Raja R, Friedman BA, Haverty PM, Polakis P, Firestein R. An antibody-drug conjugate directed against lymphocyte antigen 6 complex, locus E (LY6E) provides robust tumor killing in a wide range of solid tumor malignancies. Clin Cancer Res. 2015;21:3252–62.CrossRefPubMed
15.
go back to reference Ronsin C, Muscatelli F, Mattei MG, Breathnach R. A novel putative receptor protein tyrosine kinase of the met family. Oncogene. 1993;8:1195–202.PubMed Ronsin C, Muscatelli F, Mattei MG, Breathnach R. A novel putative receptor protein tyrosine kinase of the met family. Oncogene. 1993;8:1195–202.PubMed
16.
go back to reference Yao HP, Zhou YQ, Zhang R, Wang MH. MSP-RON signaling in cancer: pathogenesis and therapeutic potential. Nat Rev Cancer. 2013;13:466–81.CrossRefPubMed Yao HP, Zhou YQ, Zhang R, Wang MH. MSP-RON signaling in cancer: pathogenesis and therapeutic potential. Nat Rev Cancer. 2013;13:466–81.CrossRefPubMed
17.
go back to reference Zhao H, Chen MS, Lo YH, Waltz SE, Wang J, Ho PC, Vasiliauskas J, Plattner R, Wang YL, Wang SC. The Ron receptor tyrosine kinase activates c-Abl to promote cell proliferation through tyrosine phosphorylation of PCNA in breast cancer. Oncogene. 2014;33:1429–37.CrossRefPubMed Zhao H, Chen MS, Lo YH, Waltz SE, Wang J, Ho PC, Vasiliauskas J, Plattner R, Wang YL, Wang SC. The Ron receptor tyrosine kinase activates c-Abl to promote cell proliferation through tyrosine phosphorylation of PCNA in breast cancer. Oncogene. 2014;33:1429–37.CrossRefPubMed
18.
go back to reference Liu X, Zhao L, Derose YS, Lin YC, Bieniasz M, Eyob H, Buys SS, Neumayer L, Welm AL. Short-form Ron promotes spontaneous breast cancer metastasis through interaction with phosphoinositide 3-Kinase. Genes Cancer. 2011;2:753–62.CrossRefPubMedPubMedCentral Liu X, Zhao L, Derose YS, Lin YC, Bieniasz M, Eyob H, Buys SS, Neumayer L, Welm AL. Short-form Ron promotes spontaneous breast cancer metastasis through interaction with phosphoinositide 3-Kinase. Genes Cancer. 2011;2:753–62.CrossRefPubMedPubMedCentral
19.
go back to reference Kawada I, Hasina R, Arif Q, Mueller J, Smithberger E, Husain AN, Vokes EE, Salgia R. Dramatic antitumor effects of the dual MET/RON small-molecule inhibitor LY2801653 in non-small cell lung cancer. Cancer Res. 2014;74:884–95.CrossRefPubMed Kawada I, Hasina R, Arif Q, Mueller J, Smithberger E, Husain AN, Vokes EE, Salgia R. Dramatic antitumor effects of the dual MET/RON small-molecule inhibitor LY2801653 in non-small cell lung cancer. Cancer Res. 2014;74:884–95.CrossRefPubMed
20.
go back to reference Kanteti R, Krishnaswamy S, Catenacci D, Tan YH, EL-Hashani E, Cervantes G, Husain AN, Tretiakova M, Vokes EE, Huet H, Salgia R. Differential expression of RON in small and non- small cell lung cancers. Genes Chromosomes Cancer. 2012;51:841–51.CrossRefPubMed Kanteti R, Krishnaswamy S, Catenacci D, Tan YH, EL-Hashani E, Cervantes G, Husain AN, Tretiakova M, Vokes EE, Huet H, Salgia R. Differential expression of RON in small and non- small cell lung cancers. Genes Chromosomes Cancer. 2012;51:841–51.CrossRefPubMed
21.
go back to reference Maggiora P, Marchio S, Stella MC, Giai M, Belfiore A, De Bortoli M, et al. Overexpression of the RON gene in human breast carcinoma. Oncogene. 1998;16:2927–33.CrossRefPubMed Maggiora P, Marchio S, Stella MC, Giai M, Belfiore A, De Bortoli M, et al. Overexpression of the RON gene in human breast carcinoma. Oncogene. 1998;16:2927–33.CrossRefPubMed
22.
go back to reference Lee WY, Chen HH, Chow NH, Su WC, Lin PW, Guo HR. Prognostic significance of co- expression of RON and MET receptors in node-negative breast cancer patients. Clin Cancer Res. 2005;11:2222–8.CrossRefPubMed Lee WY, Chen HH, Chow NH, Su WC, Lin PW, Guo HR. Prognostic significance of co- expression of RON and MET receptors in node-negative breast cancer patients. Clin Cancer Res. 2005;11:2222–8.CrossRefPubMed
23.
go back to reference Wang MH, Lee W, Luo YL, Weis MT, Yao HP. Altered expression of the RON receptor tyrosine kinase in various epithelial cancers and its contribution to tumorigenic phenotypes in thyroid cancer cells. J Pathol. 2007;213:402–11.CrossRefPubMed Wang MH, Lee W, Luo YL, Weis MT, Yao HP. Altered expression of the RON receptor tyrosine kinase in various epithelial cancers and its contribution to tumorigenic phenotypes in thyroid cancer cells. J Pathol. 2007;213:402–11.CrossRefPubMed
24.
go back to reference Yao HP, Zhuang CM, Zhou YQ, Zeng JY, Zhang RW, Wang MH. Oncogenic variant RON160 expression in breast cancer and its potential as a therapeutic target by small molecule tyrosine kinase inhibitor. Curr Cancer Drug Targets. 2013;13:686–97.CrossRefPubMed Yao HP, Zhuang CM, Zhou YQ, Zeng JY, Zhang RW, Wang MH. Oncogenic variant RON160 expression in breast cancer and its potential as a therapeutic target by small molecule tyrosine kinase inhibitor. Curr Cancer Drug Targets. 2013;13:686–97.CrossRefPubMed
25.
go back to reference Feng L, Yao HP, Wang W, Zhou YQ, Zhou J, Zhang R, Wang MH. Efficacy of anti-RON antibody Zt/g4-drug maytansinoid conjugation (Anti-RON ADC) as a novel therapeutics for targeted colorectal cancer therapy. Clin Cancer Res. 2014;20:6045–58.CrossRefPubMed Feng L, Yao HP, Wang W, Zhou YQ, Zhou J, Zhang R, Wang MH. Efficacy of anti-RON antibody Zt/g4-drug maytansinoid conjugation (Anti-RON ADC) as a novel therapeutics for targeted colorectal cancer therapy. Clin Cancer Res. 2014;20:6045–58.CrossRefPubMed
26.
go back to reference O’Toole JM, Rabenau KE, Burns K, Lu D, Mangalampalli V, Balderes P, Covino N, Bassi R, Prewett M, Gottfredsen KJ, Thobe MN, Cheng Y, Li Y, Hicklin DJ, Zhu Z, Waltz SE, Hayman MJ, Ludwig DL, Pereira DS. Therapeutic implications of a human neutralizing antibody to the macrophage-stimulating protein receptor tyrosine kinase (RON), a c-MET family member. Cancer Res. 2006;66:9162–70.CrossRefPubMed O’Toole JM, Rabenau KE, Burns K, Lu D, Mangalampalli V, Balderes P, Covino N, Bassi R, Prewett M, Gottfredsen KJ, Thobe MN, Cheng Y, Li Y, Hicklin DJ, Zhu Z, Waltz SE, Hayman MJ, Ludwig DL, Pereira DS. Therapeutic implications of a human neutralizing antibody to the macrophage-stimulating protein receptor tyrosine kinase (RON), a c-MET family member. Cancer Res. 2006;66:9162–70.CrossRefPubMed
27.
go back to reference Schroeder GM, An Y, Cai ZW, Chen XT, Clark C, Cornelius LA, et al. Discovery of N-(4-(2- amino-3-chloropyridin-4-yloxy)-3-fluorophenyl)-4-ethoxy-1-(4-fluorophenyl)-2-oxo-1,2- dihydropyridine-3-carboxamide (BMS-777607), a selective and orally efficacious inhibitor of the Met kinase superfamily. J Med Chem. 2009;52:1251–4.CrossRefPubMed Schroeder GM, An Y, Cai ZW, Chen XT, Clark C, Cornelius LA, et al. Discovery of N-(4-(2- amino-3-chloropyridin-4-yloxy)-3-fluorophenyl)-4-ethoxy-1-(4-fluorophenyl)-2-oxo-1,2- dihydropyridine-3-carboxamide (BMS-777607), a selective and orally efficacious inhibitor of the Met kinase superfamily. J Med Chem. 2009;52:1251–4.CrossRefPubMed
28.
go back to reference Raeppel SL, Raeppel F, Therrien E. Design and synthesis of close analogs of LCRF-0004, a potent and selective RON receptor tyrosine kinase inhibitor. Bioorg Med Chem Lett. 2015;25:2527–31.CrossRefPubMed Raeppel SL, Raeppel F, Therrien E. Design and synthesis of close analogs of LCRF-0004, a potent and selective RON receptor tyrosine kinase inhibitor. Bioorg Med Chem Lett. 2015;25:2527–31.CrossRefPubMed
29.
go back to reference Northrup AB, Katcher MH, Altman MD, Chenard M, Daniels MH, Deshmukh SV, Falcone D, Guerin DJ, Hatch H, Li C, Lu W, Lutterbach B, Allison TJ, Patel SB, Reilly JF, Reutershan M, Rickert KW, Rosenstein C, Soisson SM, Szewczak AA, Walker D, Wilson K, Young JR, Pan BS, Dinsmore CJ. Discovery of 1-[3-(1-methyl-1H-pyrazol-4-yl)-5-oxo-5H-benzo[4,5] cyclohepta[1,2-b]pyridin-7-yl]-N-(pyridin-2-ylmethyl)methanesulfonamide (MK-8033): A Specific c-Met/Ron dual kinase inhibitor with preferential affinity for the activated state of c- Met. J Med Chem. 2013;56:2294–310.CrossRefPubMed Northrup AB, Katcher MH, Altman MD, Chenard M, Daniels MH, Deshmukh SV, Falcone D, Guerin DJ, Hatch H, Li C, Lu W, Lutterbach B, Allison TJ, Patel SB, Reilly JF, Reutershan M, Rickert KW, Rosenstein C, Soisson SM, Szewczak AA, Walker D, Wilson K, Young JR, Pan BS, Dinsmore CJ. Discovery of 1-[3-(1-methyl-1H-pyrazol-4-yl)-5-oxo-5H-benzo[4,5] cyclohepta[1,2-b]pyridin-7-yl]-N-(pyridin-2-ylmethyl)methanesulfonamide (MK-8033): A Specific c-Met/Ron dual kinase inhibitor with preferential affinity for the activated state of c- Met. J Med Chem. 2013;56:2294–310.CrossRefPubMed
30.
go back to reference Guin S, Yao HP, Wang MH. RON receptor tyrosine kinase as a target for delivery of chemodrugs by antibody directed pathway for cancer cell cytotoxicity. Mol Pharm. 2010;7:386–97.CrossRefPubMed Guin S, Yao HP, Wang MH. RON receptor tyrosine kinase as a target for delivery of chemodrugs by antibody directed pathway for cancer cell cytotoxicity. Mol Pharm. 2010;7:386–97.CrossRefPubMed
31.
go back to reference Guin S, Ma Q, Padhye S, Zhou YQ, Yao HP, Wang MH. Targeting acute hypoxic cancer cells by doxorubicin-immunoliposomes directed by monoclonal antibodies specific to RON receptor tyrosine kinase. Cancer Chemother Pharmacol. 2011;67:1073–83.CrossRefPubMed Guin S, Ma Q, Padhye S, Zhou YQ, Yao HP, Wang MH. Targeting acute hypoxic cancer cells by doxorubicin-immunoliposomes directed by monoclonal antibodies specific to RON receptor tyrosine kinase. Cancer Chemother Pharmacol. 2011;67:1073–83.CrossRefPubMed
32.
go back to reference Padhye SS, Guin S, Yao HP, Zhou YQ, Zhang R, Wang MH. Sustained expression of the RON receptor tyrosine kinase by pancreatic cancer stem cells as a potential targeting moiety for antibody-directed chemotherapeutics. Mol Pharm. 2011;8:2310–9.CrossRefPubMed Padhye SS, Guin S, Yao HP, Zhou YQ, Zhang R, Wang MH. Sustained expression of the RON receptor tyrosine kinase by pancreatic cancer stem cells as a potential targeting moiety for antibody-directed chemotherapeutics. Mol Pharm. 2011;8:2310–9.CrossRefPubMed
33.
go back to reference Yao HP, Luo YL, Feng L, Cheng LF, Lu Y, Li W, Wang MH. Agonistic monoclonal antibodies potentiate tumorigenic and invasive activities of splicing variant of the RON receptor tyrosine kinase. Cancer Biol Ther. 2006;5:1179–86.CrossRefPubMed Yao HP, Luo YL, Feng L, Cheng LF, Lu Y, Li W, Wang MH. Agonistic monoclonal antibodies potentiate tumorigenic and invasive activities of splicing variant of the RON receptor tyrosine kinase. Cancer Biol Ther. 2006;5:1179–86.CrossRefPubMed
34.
go back to reference Li Z, Yao H, Guin S, Padhye SS, Zhou YQ, Wang MH. Monoclonal antibody (mAb)-induced down-regulation of RON receptor tyrosine kinase diminishes tumorigenic activities of colon cancer cells. Int J Oncol. 2010;37:473–82.PubMed Li Z, Yao H, Guin S, Padhye SS, Zhou YQ, Wang MH. Monoclonal antibody (mAb)-induced down-regulation of RON receptor tyrosine kinase diminishes tumorigenic activities of colon cancer cells. Int J Oncol. 2010;37:473–82.PubMed
35.
36.
go back to reference Sharma S, Zeng JY, Zhuang CM, Zhou YQ, Yao HP, Hu X, Wang MH. Small-molecule inhibitor BMS-777607 induces breast cancer cell polyploidy with increased resistance to cytotoxic chemotherapy agents. Mol Cancer Ther. 2013;12:725–36.CrossRefPubMed Sharma S, Zeng JY, Zhuang CM, Zhou YQ, Yao HP, Hu X, Wang MH. Small-molecule inhibitor BMS-777607 induces breast cancer cell polyploidy with increased resistance to cytotoxic chemotherapy agents. Mol Cancer Ther. 2013;12:725–36.CrossRefPubMed
37.
go back to reference Plunkett W, Huang P, Xu YZ, Heinemann V, Grunewald R, Gandhi V. Gemcitabine: metabolism, mechanisms of action, and self-potentiation. Semin Oncol. 1995;22 Suppl 11:3–10.PubMed Plunkett W, Huang P, Xu YZ, Heinemann V, Grunewald R, Gandhi V. Gemcitabine: metabolism, mechanisms of action, and self-potentiation. Semin Oncol. 1995;22 Suppl 11:3–10.PubMed
38.
go back to reference Saveria M, Montani G, Prodosmo A, Stagni V, Merli D, Monteonofrio L, Gatti V, Gentileschi MP, Barilà D, Soddu S. ATM-depletion in breast cancer cells confers sensitivity to PARP inhibition. J Exp Clin Cancer Res. 2013;32:95–105.CrossRef Saveria M, Montani G, Prodosmo A, Stagni V, Merli D, Monteonofrio L, Gatti V, Gentileschi MP, Barilà D, Soddu S. ATM-depletion in breast cancer cells confers sensitivity to PARP inhibition. J Exp Clin Cancer Res. 2013;32:95–105.CrossRef
39.
go back to reference Lopus M, Oroudjev E, Wilson L, Wilhelm S, Widdison W, Chari R, Ma J. Maytansine and cellular metabolites of antibody-maytansinoid conjugates strongly suppress microtubule dynamics by binding to microtubules. Mol Cancer Ther. 2010;9:2689–99.CrossRefPubMedPubMedCentral Lopus M, Oroudjev E, Wilson L, Wilhelm S, Widdison W, Chari R, Ma J. Maytansine and cellular metabolites of antibody-maytansinoid conjugates strongly suppress microtubule dynamics by binding to microtubules. Mol Cancer Ther. 2010;9:2689–99.CrossRefPubMedPubMedCentral
40.
go back to reference Jumbe NL, Xin Y, Leipold DD, Crocker L, Dugger D, Mai E, Sliwkowski M, Fielder PJ, Tibbitts J. Modeling the efficacy of trastuzumab-DM1, an antibody drug conjugate in mice. J Pharmacokinet Pharmacodyn. 2010;37:221–42.CrossRefPubMed Jumbe NL, Xin Y, Leipold DD, Crocker L, Dugger D, Mai E, Sliwkowski M, Fielder PJ, Tibbitts J. Modeling the efficacy of trastuzumab-DM1, an antibody drug conjugate in mice. J Pharmacokinet Pharmacodyn. 2010;37:221–42.CrossRefPubMed
41.
go back to reference Wang H, Rangan VS, Sung MC, Passmore D, Kempe T, Wang X, Thevanayagam L, Pan C, Rao C, Srinivasan M, Zhang Q, Gangwar S, Deshpande S, Cardarelli P, Marathe P, Yang Z. Pharmacokinetic characterization of BMS-936561, an anti-CD70 antibody-drug conjugate, in preclinical animal species and prediction of its pharmacokinetics in humans. Biopharm Drug Dispos. 2015. doi:10.1002/bdd.1953. Epub ahead of print. Wang H, Rangan VS, Sung MC, Passmore D, Kempe T, Wang X, Thevanayagam L, Pan C, Rao C, Srinivasan M, Zhang Q, Gangwar S, Deshpande S, Cardarelli P, Marathe P, Yang Z. Pharmacokinetic characterization of BMS-936561, an anti-CD70 antibody-drug conjugate, in preclinical animal species and prediction of its pharmacokinetics in humans. Biopharm Drug Dispos. 2015. doi:10.​1002/​bdd.​1953. Epub ahead of print.
42.
go back to reference Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006;58:621–81.CrossRefPubMed Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 2006;58:621–81.CrossRefPubMed
43.
go back to reference Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70:440–6.CrossRefPubMed Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70:440–6.CrossRefPubMed
Metadata
Title
Biological evaluation of antibody-maytansinoid conjugates as a strategy of RON targeted drug delivery for treatment of non-small cell lung cancer
Authors
Liang Feng
Hang-Ping Yao
Sharad Sharma
Yong-Qing Zhou
Jianwei Zhou
Ruiwen Zhang
Ming-Hai Wang
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2016
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-016-0347-6

Other articles of this Issue 1/2016

Journal of Experimental & Clinical Cancer Research 1/2016 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