Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 5/2011

01-05-2011 | Original Article

Targeting acute hypoxic cancer cells by doxorubicin-immunoliposomes directed by monoclonal antibodies specific to RON receptor tyrosine kinase

Authors: Sunny Guin, Qi Ma, Snehal Padhye, Yong-Qing Zhou, Hang-Ping Yao, Ming-Hai Wang

Published in: Cancer Chemotherapy and Pharmacology | Issue 5/2011

Login to get access

Abstract

Purpose

Hypoxia contributes to acquired drug resistance in various cancer cells. The underlying mechanism is cellular insensitivity regulated by hypoxia-inducible factors (HIF), which impairs drug uptake, transport, and metabolism. The current study determines anti-RON antibody-directed cytotoxicity of doxorubicin (Dox)-immunoliposomes (IL) in hypoxic colon cancer cells.

Methods

Cells were cultured under hypoxia (1% O2, 5% CO2, and 96% N2) for 24 h. Dox-loaded IL were formulated followed by post-insertion of monoclonal antibody Zt/g4 specific to RON. Western blotting was used to detect HIF-1α and RON expression. Cellular uptake of Zt/g4-conjugated IL was determined by confocal and internalization assays. Cell viability was assessed by the MTT assay.

Results

RON and HIF-1α expression were observed in hypoxic colon HCT116 and SW620 cells. Resistance to Dox-induced cytotoxicity was acquired in hypoxic cells with increased IC50 values. However, acquired resistance was attenuated by Zt/g4-directed Dox-IL, which displays increased cytotoxic activities. IL binding and uptake revealed that hypoxic RON expression is functional, which mediates high levels of Zt/g4-Dox-IL binding and cytoplasmic internalization. Zt/g4-Dox-IL is effective in killing hypoxic HCT116 and SW620 cells with reduced IC50 values compared to Dox and pegylated-liposomal Dox. These effects were dependent on hypoxic RON expression. HCC1937 cells with diminished RON expression under hypoxia were insensitive to Zt/g4-Dox-IL-induced cytotoxic effect.

Conclusions

RON expressed by hypoxic colon cancer cells is thus a potential targeting molecule for delivery of chemotherapeutics. The ability of anti-RON mAb to direct Dox-IL cytotoxicity could be developed for attenuating hypoxia-acquired drug resistance in various cancer cells.
Literature
1.
go back to reference Thomlinson RH, Gray LH (1955) The histological structure of some human lung cancers and the possible implications for radiotherapy. Br J Cancer 9:539–549PubMedCrossRef Thomlinson RH, Gray LH (1955) The histological structure of some human lung cancers and the possible implications for radiotherapy. Br J Cancer 9:539–549PubMedCrossRef
2.
go back to reference Semenza GL (2010) Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 29:625–634PubMedCrossRef Semenza GL (2010) Defining the role of hypoxia-inducible factor 1 in cancer biology and therapeutics. Oncogene 29:625–634PubMedCrossRef
3.
4.
go back to reference Rankin EB, Giaccia AJ (2008) The role of hypoxia-inducible factors in tumorigenesis. Cell Death Differ 15:678–685PubMedCrossRef Rankin EB, Giaccia AJ (2008) The role of hypoxia-inducible factors in tumorigenesis. Cell Death Differ 15:678–685PubMedCrossRef
5.
go back to reference Hickey MM, Simon MC (2006) Regulation of angiogenesis by hypoxia and hypoxia-inducible factors. Curr Top Dev Biol 76:217–257PubMedCrossRef Hickey MM, Simon MC (2006) Regulation of angiogenesis by hypoxia and hypoxia-inducible factors. Curr Top Dev Biol 76:217–257PubMedCrossRef
7.
go back to reference Trédan O, Galmarini CM, Patel K, Tannock IF (2007) Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst 99:1441–1454PubMedCrossRef Trédan O, Galmarini CM, Patel K, Tannock IF (2007) Drug resistance and the solid tumor microenvironment. J Natl Cancer Inst 99:1441–1454PubMedCrossRef
8.
go back to reference Yokoi K, Fidler IJ (2004) Hypoxia increases resistance of human pancreatic cancer cells to apoptosis induced by gemcitabine. Clin Cancer Res 10:2299–2306PubMedCrossRef Yokoi K, Fidler IJ (2004) Hypoxia increases resistance of human pancreatic cancer cells to apoptosis induced by gemcitabine. Clin Cancer Res 10:2299–2306PubMedCrossRef
9.
go back to reference Rockwell S, Dobrucki IT, Kim EY, Marrison ST, Vu VT (2009) Hypoxia and radiation therapy: past history, ongoing research, and future promise. Curr Mol Med 9:442–458PubMedCrossRef Rockwell S, Dobrucki IT, Kim EY, Marrison ST, Vu VT (2009) Hypoxia and radiation therapy: past history, ongoing research, and future promise. Curr Mol Med 9:442–458PubMedCrossRef
10.
go back to reference Ronsin C, Muscatelli F, Mattei MG, Breathnach R (1993) A novel putative receptor protein tyrosine kinase of the met family. Oncogene 8:1195–1202PubMed Ronsin C, Muscatelli F, Mattei MG, Breathnach R (1993) A novel putative receptor protein tyrosine kinase of the met family. Oncogene 8:1195–1202PubMed
11.
go back to reference Wang M-H, Ronsin C, Gesnel M-C, Coupey L, Skeel A, Leonard EJ, Breathnach R (1994) Identification of the RON gene product as the receptor for the human macrophage stimulating protein. Science 266:117–119PubMedCrossRef Wang M-H, Ronsin C, Gesnel M-C, Coupey L, Skeel A, Leonard EJ, Breathnach R (1994) Identification of the RON gene product as the receptor for the human macrophage stimulating protein. Science 266:117–119PubMedCrossRef
12.
go back to reference Wagh PK, Peace BE, Waltz SE (2008) Met-related receptor tyrosine kinase Ron in tumor growth and metastasis. Adv Cancer Res 100:1–33PubMedCrossRef Wagh PK, Peace BE, Waltz SE (2008) Met-related receptor tyrosine kinase Ron in tumor growth and metastasis. Adv Cancer Res 100:1–33PubMedCrossRef
13.
go back to reference Wang MH, Lee W, Luo YL, Weis MT, Yao HP (2007) Altered expression of the RON receptor tyrosine kinase in various epithelial cancers and its contribution to tumourigenic phenotypes in thyroid cancer cells. J Pathol 213:402–411PubMedCrossRef Wang MH, Lee W, Luo YL, Weis MT, Yao HP (2007) Altered expression of the RON receptor tyrosine kinase in various epithelial cancers and its contribution to tumourigenic phenotypes in thyroid cancer cells. J Pathol 213:402–411PubMedCrossRef
14.
go back to reference Thomas RM, Toney K, Fenoglio-Preiser C, Revelo-Penafiel MP, Hingorani SR, Tuveson DA, Waltz SE, Lowy AM (2007) The RON receptor tyrosine kinase mediates oncogenic phenotypes in pancreatic cancer cells and is increasingly expressed during pancreatic cancer progression. Cancer Res 67:6075–6082PubMedCrossRef Thomas RM, Toney K, Fenoglio-Preiser C, Revelo-Penafiel MP, Hingorani SR, Tuveson DA, Waltz SE, Lowy AM (2007) The RON receptor tyrosine kinase mediates oncogenic phenotypes in pancreatic cancer cells and is increasingly expressed during pancreatic cancer progression. Cancer Res 67:6075–6082PubMedCrossRef
15.
go back to reference Welm AL, Sneddon JB, Taylor C, Nuyten DS, van de Vijver MJ, Hasegawa BH, Bishop JM (2007) The macrophage-stimulating protein pathway promotes metastasis in a mouse model for breast cancer and predicts poor prognosis in humans. Proc Natl Acad Sci USA 104:7570–7575PubMedCrossRef Welm AL, Sneddon JB, Taylor C, Nuyten DS, van de Vijver MJ, Hasegawa BH, Bishop JM (2007) The macrophage-stimulating protein pathway promotes metastasis in a mouse model for breast cancer and predicts poor prognosis in humans. Proc Natl Acad Sci USA 104:7570–7575PubMedCrossRef
16.
go back to reference Wang MH, Wang D, Chen YQ (2003) Oncogenic and invasive potentials of human macrophage-stimulating protein receptor, the RON receptor tyrosine kinase. Carcinogenesis 24:1291–1300PubMedCrossRef Wang MH, Wang D, Chen YQ (2003) Oncogenic and invasive potentials of human macrophage-stimulating protein receptor, the RON receptor tyrosine kinase. Carcinogenesis 24:1291–1300PubMedCrossRef
17.
go back to reference Camp ER, Liu W, Fan F, Yang A, Somcio R, Ellis LM (2005) RON, a tyrosine kinase receptor involved in tumor progression and metastasis. Ann Surg Oncol 12:273–281PubMedCrossRef Camp ER, Liu W, Fan F, Yang A, Somcio R, Ellis LM (2005) RON, a tyrosine kinase receptor involved in tumor progression and metastasis. Ann Surg Oncol 12:273–281PubMedCrossRef
18.
go back to reference Logan-Collins J, Thomas RM, Yu P et al (2010) Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res 70:1130–1140PubMedCrossRef Logan-Collins J, Thomas RM, Yu P et al (2010) Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res 70:1130–1140PubMedCrossRef
19.
go back to reference Lee WY, Chen HH, Chow NH, Su WC, Lin PW, Guo HR (2005) Prognostic significance of co-expression of RON and MET receptors in node-negative breast cancer patients. Clin Cancer Res 11:2222–2228PubMedCrossRef Lee WY, Chen HH, Chow NH, Su WC, Lin PW, Guo HR (2005) Prognostic significance of co-expression of RON and MET receptors in node-negative breast cancer patients. Clin Cancer Res 11:2222–2228PubMedCrossRef
20.
go back to reference Dussault I, Bellon SF (2009) From concept to reality: the long road to c-Met and RON receptor tyrosine kinase inhibitors for the treatment of cancer. Anticancer Agents Med Chem 9:221–229PubMed Dussault I, Bellon SF (2009) From concept to reality: the long road to c-Met and RON receptor tyrosine kinase inhibitors for the treatment of cancer. Anticancer Agents Med Chem 9:221–229PubMed
21.
go back to reference O’Toole JM, Rabenau KE, Burns K et al (2006) Therapeutic implications of a human neutralizing antibody to the macrophage-stimulating protein receptor tyrosine kinase (RON), a c-MET family member. Cancer Res 66:9162–9170PubMedCrossRef O’Toole JM, Rabenau KE, Burns K et al (2006) Therapeutic implications of a human neutralizing antibody to the macrophage-stimulating protein receptor tyrosine kinase (RON), a c-MET family member. Cancer Res 66:9162–9170PubMedCrossRef
22.
go back to reference Zhang Y, Kaplan-Lefko PJ et al (2008) Identification of a novel recepteur d’origine nantais/c-met small-molecule kinase inhibitor with antitumor activity in vivo. Cancer Res 68:6680–6687PubMedCrossRef Zhang Y, Kaplan-Lefko PJ et al (2008) Identification of a novel recepteur d’origine nantais/c-met small-molecule kinase inhibitor with antitumor activity in vivo. Cancer Res 68:6680–6687PubMedCrossRef
23.
go back to reference Christensen JG, Schreck R, Burrows J et al (2003) A selective small molecule inhibitor of c-Met kinase inhibits c-Met-dependent phenotypes in vitro and exhibits cytoreductive antitumor activity in vivo. Cancer Res 63:7345–7355PubMed Christensen JG, Schreck R, Burrows J et al (2003) A selective small molecule inhibitor of c-Met kinase inhibits c-Met-dependent phenotypes in vitro and exhibits cytoreductive antitumor activity in vivo. Cancer Res 63:7345–7355PubMed
24.
go back to reference Guin S, Yao HP, Wang MH (2010) RON Receptor Tyrosine Kinase as a Target for Delivery of Chemodrugs by Antibody Directed Pathway for Cancer Cell Cytotoxicity. Mol Pharm (in press) Guin S, Yao HP, Wang MH (2010) RON Receptor Tyrosine Kinase as a Target for Delivery of Chemodrugs by Antibody Directed Pathway for Cancer Cell Cytotoxicity. Mol Pharm (in press)
25.
go back to reference Xu RH, Pelicano H, Zhou Y, Carew JS, Feng L, Bhalla KN, Keating MJ, Huang P (2005) Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer Res 65:613–621PubMedCrossRef Xu RH, Pelicano H, Zhou Y, Carew JS, Feng L, Bhalla KN, Keating MJ, Huang P (2005) Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer Res 65:613–621PubMedCrossRef
26.
27.
go back to reference Iden DL, Allen TM (2001) In vitro and in vivo comparison of immunoliposomes made by conventional coupling techniques with those made by a new post-insertion approach. Biochim Biophys Acta 1513:207–216PubMedCrossRef Iden DL, Allen TM (2001) In vitro and in vivo comparison of immunoliposomes made by conventional coupling techniques with those made by a new post-insertion approach. Biochim Biophys Acta 1513:207–216PubMedCrossRef
28.
go back to reference Soundararajan A, Bao A, Phillips WT, Perez R III, Goins BA (2009) [186Re]Liposomal doxorubicin (Doxil): in vitro stability, pharmacokinetics, imaging and biodistribution in a head and neck squamous cell carcinoma xenograft model. Nuc Med Biol 36:515–524CrossRef Soundararajan A, Bao A, Phillips WT, Perez R III, Goins BA (2009) [186Re]Liposomal doxorubicin (Doxil): in vitro stability, pharmacokinetics, imaging and biodistribution in a head and neck squamous cell carcinoma xenograft model. Nuc Med Biol 36:515–524CrossRef
29.
go back to reference Miller CR, Bondurant B, McLean SD, McGovern KA, O’Brien DF (1998) Liposome-cell interactions in vitro: effect of liposome surface charge on the binding and endocytosis of conventional and sterically stabilized liposomes. Biochemistry 37:12875–12883PubMedCrossRef Miller CR, Bondurant B, McLean SD, McGovern KA, O’Brien DF (1998) Liposome-cell interactions in vitro: effect of liposome surface charge on the binding and endocytosis of conventional and sterically stabilized liposomes. Biochemistry 37:12875–12883PubMedCrossRef
30.
go back to reference Wang Y, Roche O, Yan MS et al (2009) Regulation of endocytosis via the oxygen-sensing pathway. Nat Med 15:319–324PubMedCrossRef Wang Y, Roche O, Yan MS et al (2009) Regulation of endocytosis via the oxygen-sensing pathway. Nat Med 15:319–324PubMedCrossRef
31.
go back to reference Teicher BA (2009) Acute and chronic in vivo therapeutic resistance. Biochem Pharmacol 77:1665–1673PubMedCrossRef Teicher BA (2009) Acute and chronic in vivo therapeutic resistance. Biochem Pharmacol 77:1665–1673PubMedCrossRef
32.
go back to reference Yao HP, Luo YL, Feng L, Cheng LF, Lu Y, Li W, Wang MH (2006) Agonistic monoclonal antibodies potentiate tumorigenic and invasive activities of splicing variant of the RON receptor tyrosine kinase. Cancer Biol Ther 5:1179–1186PubMedCrossRef Yao HP, Luo YL, Feng L, Cheng LF, Lu Y, Li W, Wang MH (2006) Agonistic monoclonal antibodies potentiate tumorigenic and invasive activities of splicing variant of the RON receptor tyrosine kinase. Cancer Biol Ther 5:1179–1186PubMedCrossRef
33.
go back to reference Brown JM, Wilson WR (2004) Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer 4:437–447PubMedCrossRef Brown JM, Wilson WR (2004) Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer 4:437–447PubMedCrossRef
34.
go back to reference Patterson LH, McKeown SR (2000) AQ4 N: a new approach to hypoxia-activated cancer chemotherapy. Br J Cancer 83:1589–1593PubMedCrossRef Patterson LH, McKeown SR (2000) AQ4 N: a new approach to hypoxia-activated cancer chemotherapy. Br J Cancer 83:1589–1593PubMedCrossRef
35.
go back to reference Shibata T, Giaccia AJ, Brown JM (2002) Hypoxia-inducible regulation of a prodrug-activating enzyme for tumor-specific gene therapy. Neoplasia 4:40–48PubMedCrossRef Shibata T, Giaccia AJ, Brown JM (2002) Hypoxia-inducible regulation of a prodrug-activating enzyme for tumor-specific gene therapy. Neoplasia 4:40–48PubMedCrossRef
36.
go back to reference Patterson AV, Williams KJ, Cowen RL et al (2002) Oxygen-sensitive enzyme-prodrug gene therapy for the eradication of radiation-resistant solid tumours. Gene Ther 9:946–954PubMedCrossRef Patterson AV, Williams KJ, Cowen RL et al (2002) Oxygen-sensitive enzyme-prodrug gene therapy for the eradication of radiation-resistant solid tumours. Gene Ther 9:946–954PubMedCrossRef
38.
go back to reference Ricart AD, Tolcher AW (2007) Technology insight: cytotoxic drug immunoconjugates for cancer therapy. Nat Clin Pract Oncol 4:245–255PubMedCrossRef Ricart AD, Tolcher AW (2007) Technology insight: cytotoxic drug immunoconjugates for cancer therapy. Nat Clin Pract Oncol 4:245–255PubMedCrossRef
39.
go back to reference Mamot C, Drummond DC, Noble CO, Kallab V, Guo Z, Hong K, Kirpotin DB, Park JW (2005) Epidermal growth factor receptor-targeted immunoliposomes significantly enhance the efficacy of multiple anticancer drugs in vivo. Cancer Res 65:11631–11638PubMedCrossRef Mamot C, Drummond DC, Noble CO, Kallab V, Guo Z, Hong K, Kirpotin DB, Park JW (2005) Epidermal growth factor receptor-targeted immunoliposomes significantly enhance the efficacy of multiple anticancer drugs in vivo. Cancer Res 65:11631–11638PubMedCrossRef
40.
go back to reference Patra CR, Bhattacharya R, Wang E et al (2008) Targeted delivery of gemcitabine to pancreatic adenocarcinoma using cetuximab as a targeting agent. Cancer Res 68:1970–1978PubMedCrossRef Patra CR, Bhattacharya R, Wang E et al (2008) Targeted delivery of gemcitabine to pancreatic adenocarcinoma using cetuximab as a targeting agent. Cancer Res 68:1970–1978PubMedCrossRef
41.
go back to reference Mamot C, Drummond DC, Greiser U, Hong K, Kirpotin DB, Marks JD, Park JW (2003) Epidermal growth factor receptor (EGFR)-targeted immunoliposomes mediate specific and efficient drug delivery to EGFR- and EGFRvIII-overexpressing tumor cells. Cancer Res 63:3154–3161PubMed Mamot C, Drummond DC, Greiser U, Hong K, Kirpotin DB, Marks JD, Park JW (2003) Epidermal growth factor receptor (EGFR)-targeted immunoliposomes mediate specific and efficient drug delivery to EGFR- and EGFRvIII-overexpressing tumor cells. Cancer Res 63:3154–3161PubMed
42.
go back to reference Cho K, Wang X, Nie S, Chen ZG, Shin DM (2008) Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res 14:1310–1316PubMedCrossRef Cho K, Wang X, Nie S, Chen ZG, Shin DM (2008) Therapeutic nanoparticles for drug delivery in cancer. Clin Cancer Res 14:1310–1316PubMedCrossRef
43.
go back to reference Carter P (2001) Improving the efficacy of antibody-based cancer therapies. Nat Rev Cancer 1:118–129PubMedCrossRef Carter P (2001) Improving the efficacy of antibody-based cancer therapies. Nat Rev Cancer 1:118–129PubMedCrossRef
44.
go back to reference Thangasamy A, Rogge J, Ammanamanchi S (2009) Recepteur d’origine nantais tyrosine kinase is a direct target of hypoxia-inducible factor-1alpha-mediated invasion of breast carcinoma cells. J Biol Chem 284:14001–14010PubMedCrossRef Thangasamy A, Rogge J, Ammanamanchi S (2009) Recepteur d’origine nantais tyrosine kinase is a direct target of hypoxia-inducible factor-1alpha-mediated invasion of breast carcinoma cells. J Biol Chem 284:14001–14010PubMedCrossRef
45.
go back to reference Greijer AE, de Jong MC, Scheffer GL, Shvarts A, van Diest PJ, van der Wall E (2005) Hypoxia-induced acidification causes mitoxantrone resistance not mediated by drug transporters in human breast cancer cells. Cell Oncol 27:43–49PubMed Greijer AE, de Jong MC, Scheffer GL, Shvarts A, van Diest PJ, van der Wall E (2005) Hypoxia-induced acidification causes mitoxantrone resistance not mediated by drug transporters in human breast cancer cells. Cell Oncol 27:43–49PubMed
46.
go back to reference Comerford KM, Wallace TJ, Karhausen J, Louis NA, Montalto MC, Colgan SP (2002) Hypoxia-inducible factor-1-dependent regulation of the multidrug resistance (MDR1) gene. Cancer Res 62:3387–3394PubMed Comerford KM, Wallace TJ, Karhausen J, Louis NA, Montalto MC, Colgan SP (2002) Hypoxia-inducible factor-1-dependent regulation of the multidrug resistance (MDR1) gene. Cancer Res 62:3387–3394PubMed
47.
go back to reference Sauvant C, Nowak M, Wirth C, Schneider B, Riemann A, Gekle M, Thews O (2008) Acidosis induces multi-drug resistance in rat prostate cancer cells (AT1) in vitro and in vivo by increasing the activity of the p-glycoprotein via activation of p38. Int J Cancer 123:2532–2542PubMedCrossRef Sauvant C, Nowak M, Wirth C, Schneider B, Riemann A, Gekle M, Thews O (2008) Acidosis induces multi-drug resistance in rat prostate cancer cells (AT1) in vitro and in vivo by increasing the activity of the p-glycoprotein via activation of p38. Int J Cancer 123:2532–2542PubMedCrossRef
Metadata
Title
Targeting acute hypoxic cancer cells by doxorubicin-immunoliposomes directed by monoclonal antibodies specific to RON receptor tyrosine kinase
Authors
Sunny Guin
Qi Ma
Snehal Padhye
Yong-Qing Zhou
Hang-Ping Yao
Ming-Hai Wang
Publication date
01-05-2011
Publisher
Springer-Verlag
Published in
Cancer Chemotherapy and Pharmacology / Issue 5/2011
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-010-1408-8

Other articles of this Issue 5/2011

Cancer Chemotherapy and Pharmacology 5/2011 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