Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 5/2013

01-05-2013 | Original Article

The interaction of bortezomib with multidrug transporters: implications for therapeutic applications in advanced multiple myeloma and other neoplasias

Authors: Robert O’Connor, Melissa G. Ooi, Justine Meiller, Jana Jakubikova, Steffen Klippel, Jake Delmore, Paul Richardson, Kenneth Anderson, Martin Clynes, Constantine S. Mitsiades, Peter O’Gorman

Published in: Cancer Chemotherapy and Pharmacology | Issue 5/2013

Login to get access

Abstract

Purpose

Bortezomib is an important agent in multiple myeloma treatment, but resistance in cell lines and patients has been described. The main mechanisms of resistance described in cancer fall into one of two categories, pharmacokinetic resistance (PK), e.g. over expression of drug efflux pumps and pharmacodynamic resistance, e.g. apoptosis resistance or altered survival pathways, where the agent reaches an appropriate concentration, but this fails to propagate an appropriate cell death response. Of the known pump mechanisms, P-glycoprotein (P-gp) is the best studied and considered to be the most important in contributing to general PK drug resistance. Resistance to bortezomib is multifactorial and there are conflicting indications that cellular overexpression of P-gp may contribute to resistance agent. Hence, better characterization of the interactions of this drug with classical resistance mechanisms should identify improved treatment applications.

Methods

Cell lines with different P-gp expression levels were used to determine the relationship between bortezomib and P-gp. Coculture system with stromal cells was used to determine the effect of the local microenvironment on the bortezomib–elacridar combination. To further assess P-gp function, intracellular accumulation of P-gp probe rhodamine-123 was utilised.

Results

In the present study, we show that bortezomib is a substrate for P-gp, but not for the other drug efflux transporters. Bortezomib activity is affected by P-gp expression and conversely, the expression of P-gp affect bortezomib’s ability to act as a P-gp substrate. The local microenvironment did not alter the cellular response to bortezomib. We also demonstrate that bortezomib directly affects the expression and function of P-gp.

Conclusions

Our findings strongly support a role for P-gp in bortezomib resistance and, therefore, suggest that combination of a P-gp inhibitor and bortezomib in P-gp positive myeloma would be a reasonable treatment combination to extend efficacy of this important drug.
Appendix
Available only for authorised users
Literature
1.
go back to reference Richardson PG, Mitsiades C, Schlossman R, Munshi N, Anderson K (2007) New drugs for myeloma. Oncologist 12:664–689PubMedCrossRef Richardson PG, Mitsiades C, Schlossman R, Munshi N, Anderson K (2007) New drugs for myeloma. Oncologist 12:664–689PubMedCrossRef
2.
go back to reference O’Connor R (2007) The pharmacology of cancer resistance. Anticancer Res 27:1267–1272PubMed O’Connor R (2007) The pharmacology of cancer resistance. Anticancer Res 27:1267–1272PubMed
3.
go back to reference Yang HH, Ma MH, Vescio RA, Berenson JR (2003) Overcoming drug resistance in multiple myeloma: the emergence of therapeutic approaches to induce apoptosis. J Clin Oncol 21:4239–4247PubMedCrossRef Yang HH, Ma MH, Vescio RA, Berenson JR (2003) Overcoming drug resistance in multiple myeloma: the emergence of therapeutic approaches to induce apoptosis. J Clin Oncol 21:4239–4247PubMedCrossRef
4.
go back to reference Grogan TM, Spier CM, Salmon SE et al (1993) P-glycoprotein expression in human plasma cell myeloma: correlation with prior chemotherapy. Blood 81:490–495PubMed Grogan TM, Spier CM, Salmon SE et al (1993) P-glycoprotein expression in human plasma cell myeloma: correlation with prior chemotherapy. Blood 81:490–495PubMed
5.
go back to reference Sonneveld P, Suciu S, Weijermans P et al (2001) Cyclosporin A combined with vincristine, doxorubicin and dexamethasone (VAD) compared with VAD alone in patients with advanced refractory multiple myeloma: an EORTC–HOVON randomized phase III study (06914). Br J Haematol 115:895–902PubMedCrossRef Sonneveld P, Suciu S, Weijermans P et al (2001) Cyclosporin A combined with vincristine, doxorubicin and dexamethasone (VAD) compared with VAD alone in patients with advanced refractory multiple myeloma: an EORTC–HOVON randomized phase III study (06914). Br J Haematol 115:895–902PubMedCrossRef
6.
go back to reference Dalton WS, Crowley JJ, Salmon SS et al (1995) A phase III randomized study of oral verapamil as a chemosensitizer to reverse drug resistance in patients with refractory myeloma. A Southwest Oncology Group study. Cancer 75:815–820PubMedCrossRef Dalton WS, Crowley JJ, Salmon SS et al (1995) A phase III randomized study of oral verapamil as a chemosensitizer to reverse drug resistance in patients with refractory myeloma. A Southwest Oncology Group study. Cancer 75:815–820PubMedCrossRef
7.
go back to reference Friedenberg WR, Rue M, Blood EA et al (2006) Phase III study of PSC-833 (valspodar) in combination with vincristine, doxorubicin, and dexamethasone (valspodar/VAD) versus VAD alone in patients with recurring or refractory multiple myeloma (E1A95): a trial of the Eastern Cooperative Oncology Group. Cancer 106:830–838PubMedCrossRef Friedenberg WR, Rue M, Blood EA et al (2006) Phase III study of PSC-833 (valspodar) in combination with vincristine, doxorubicin, and dexamethasone (valspodar/VAD) versus VAD alone in patients with recurring or refractory multiple myeloma (E1A95): a trial of the Eastern Cooperative Oncology Group. Cancer 106:830–838PubMedCrossRef
8.
go back to reference Sonneveld P, Marie JP, Huisman C et al (1996) Reversal of multidrug resistance by SDZ PSC 833, combined with VAD (vincristine, doxorubicin, dexamethasone) in refractory multiple myeloma. A phase I study. Leukemia 10:1741–1750PubMed Sonneveld P, Marie JP, Huisman C et al (1996) Reversal of multidrug resistance by SDZ PSC 833, combined with VAD (vincristine, doxorubicin, dexamethasone) in refractory multiple myeloma. A phase I study. Leukemia 10:1741–1750PubMed
9.
go back to reference Rumpold H, Salvador C, Wolf AM, Tilg H, Gastl G, Wolf D (2007) Knockdown of PgP resensitizes leukemic cells to proteasome inhibitors. Biochem Biophys Res Commun 361:549–554PubMedCrossRef Rumpold H, Salvador C, Wolf AM, Tilg H, Gastl G, Wolf D (2007) Knockdown of PgP resensitizes leukemic cells to proteasome inhibitors. Biochem Biophys Res Commun 361:549–554PubMedCrossRef
10.
go back to reference Loo TW, Clarke DM (1999) The human multidrug resistance P-glycoprotein is inactive when its maturation is inhibited: potential for a role in cancer chemotherapy. FASEB J 13:1724–1732PubMed Loo TW, Clarke DM (1999) The human multidrug resistance P-glycoprotein is inactive when its maturation is inhibited: potential for a role in cancer chemotherapy. FASEB J 13:1724–1732PubMed
11.
go back to reference Duffy CP, Elliott CJ, O’Connor RA et al (1998) Enhancement of chemotherapeutic drug toxicity to human tumour cells in vitro by a subset of non-steroidal anti-inflammatory drugs (NSAIDs). Eur J Cancer 34:1250–1259PubMedCrossRef Duffy CP, Elliott CJ, O’Connor RA et al (1998) Enhancement of chemotherapeutic drug toxicity to human tumour cells in vitro by a subset of non-steroidal anti-inflammatory drugs (NSAIDs). Eur J Cancer 34:1250–1259PubMedCrossRef
12.
go back to reference Clynes M, Redmond A, Moran E, Gilvarry U (1992) Multiple drug-resistance in variant of a human non-small cell lung carcinoma cell line, DLKP-A. Cytotechnology 10:75–89PubMedCrossRef Clynes M, Redmond A, Moran E, Gilvarry U (1992) Multiple drug-resistance in variant of a human non-small cell lung carcinoma cell line, DLKP-A. Cytotechnology 10:75–89PubMedCrossRef
13.
go back to reference Murphy L, Clynes M, Keenan J (2007) Proteomic analysis to dissect mitoxantrone resistance-associated proteins in a squamous lung carcinoma. Anticancer Res 27:1277–1284PubMed Murphy L, Clynes M, Keenan J (2007) Proteomic analysis to dissect mitoxantrone resistance-associated proteins in a squamous lung carcinoma. Anticancer Res 27:1277–1284PubMed
14.
go back to reference Collins DM, Crown J, O'Donovan N et al (2010) Tyrosine kinase inhibitors potentiate the cytotoxicity of MDR-substrate anticancer agents independent of growth factor receptor status in lung cancer cell lines. Invest New Drugs 28(4):433–444 Collins DM, Crown J, O'Donovan N et al (2010) Tyrosine kinase inhibitors potentiate the cytotoxicity of MDR-substrate anticancer agents independent of growth factor receptor status in lung cancer cell lines. Invest New Drugs 28(4):433–444
15.
go back to reference Roovers DJ, van Vliet M, Bloem AC, Lokhorst HM (1999) Idarubicin overcomes P-glycoprotein-related multidrug resistance: comparison with doxorubicin and daunorubicin in human multiple myeloma cell lines. Leuk Res 23:539–548PubMedCrossRef Roovers DJ, van Vliet M, Bloem AC, Lokhorst HM (1999) Idarubicin overcomes P-glycoprotein-related multidrug resistance: comparison with doxorubicin and daunorubicin in human multiple myeloma cell lines. Leuk Res 23:539–548PubMedCrossRef
16.
go back to reference Sarver JG, Klis WA, Byers JP, Erhardt PW (2002) Microplate screening of the differential effects of test agents on Hoechst 33342, rhodamine 123, and rhodamine 6G accumulation in breast cancer cells that overexpress P-glycoprotein. J Biomol Screen 7:29–34PubMed Sarver JG, Klis WA, Byers JP, Erhardt PW (2002) Microplate screening of the differential effects of test agents on Hoechst 33342, rhodamine 123, and rhodamine 6G accumulation in breast cancer cells that overexpress P-glycoprotein. J Biomol Screen 7:29–34PubMed
17.
go back to reference Martin A, Clynes M (1991) Acid phosphatase: endpoint for in vitro toxicity tests. In Vitro Cell Dev Biol 27A:183–184PubMedCrossRef Martin A, Clynes M (1991) Acid phosphatase: endpoint for in vitro toxicity tests. In Vitro Cell Dev Biol 27A:183–184PubMedCrossRef
18.
go back to reference McMillin DW, Delmore J, Weisberg E et al (2010) Tumor cell-specific bioluminescence platform to identify stroma-induced changes to anticancer drug activity. Nat Med 16:483–489 McMillin DW, Delmore J, Weisberg E et al (2010) Tumor cell-specific bioluminescence platform to identify stroma-induced changes to anticancer drug activity. Nat Med 16:483–489
19.
go back to reference McMillin DW, Ooi M, Delmore J et al (2009) Antimyeloma activity of the orally bioavailable dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235. Cancer Res 69:5835–5842PubMedCrossRef McMillin DW, Ooi M, Delmore J et al (2009) Antimyeloma activity of the orally bioavailable dual phosphatidylinositol 3-kinase/mammalian target of rapamycin inhibitor NVP-BEZ235. Cancer Res 69:5835–5842PubMedCrossRef
20.
go back to reference Ooi MG, Hayden PJ, Kotoula V et al (2009) Interactions of the Hdm2/p53 and proteasome pathways may enhance the antitumor activity of bortezomib. Clin Cancer Res 15(23):7153–7160 Ooi MG, Hayden PJ, Kotoula V et al (2009) Interactions of the Hdm2/p53 and proteasome pathways may enhance the antitumor activity of bortezomib. Clin Cancer Res 15(23):7153–7160
21.
go back to reference Schmidmaier R, Baumann P, Meinhardt G (2006) Cell-cell contact mediated signalling—no fear of contact. Exp Oncol 28:12–15PubMed Schmidmaier R, Baumann P, Meinhardt G (2006) Cell-cell contact mediated signalling—no fear of contact. Exp Oncol 28:12–15PubMed
22.
go back to reference Chou TC, Talalay P (1983) Analysis of combined drug effects: a new look at a very old problem. Trends Pharmacol Sci 4:450–454CrossRef Chou TC, Talalay P (1983) Analysis of combined drug effects: a new look at a very old problem. Trends Pharmacol Sci 4:450–454CrossRef
23.
go back to reference Chou TC, Talalay P (1984) Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzym Regul 22:27–55CrossRef Chou TC, Talalay P (1984) Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzym Regul 22:27–55CrossRef
24.
go back to reference Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC (2007) Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer 7:585–598PubMedCrossRef Hideshima T, Mitsiades C, Tonon G, Richardson PG, Anderson KC (2007) Understanding multiple myeloma pathogenesis in the bone marrow to identify new therapeutic targets. Nat Rev Cancer 7:585–598PubMedCrossRef
25.
go back to reference Nakamura T, Tanaka K, Matsunobu T et al (2007) The mechanism of cross-resistance to proteasome inhibitor bortezomib and overcoming resistance in Ewing’s family tumor cells. Int J Oncol 31:803–811PubMed Nakamura T, Tanaka K, Matsunobu T et al (2007) The mechanism of cross-resistance to proteasome inhibitor bortezomib and overcoming resistance in Ewing’s family tumor cells. Int J Oncol 31:803–811PubMed
26.
go back to reference Iijima M, Momose I, Ikeda D (2010) Increased ABCB1 expression in TP-110-resistant RPMI-8226 cells. Biosci Biotechnol Biochem 74:1913–1919PubMedCrossRef Iijima M, Momose I, Ikeda D (2010) Increased ABCB1 expression in TP-110-resistant RPMI-8226 cells. Biosci Biotechnol Biochem 74:1913–1919PubMedCrossRef
27.
go back to reference Styczynski J, Olszewska-Slonina D, Kolodziej B, Napieraj M, Wysocki M (2006) Activity of bortezomib in glioblastoma. Anticancer Res 26:4499–4503PubMed Styczynski J, Olszewska-Slonina D, Kolodziej B, Napieraj M, Wysocki M (2006) Activity of bortezomib in glioblastoma. Anticancer Res 26:4499–4503PubMed
28.
go back to reference Wiberg K, Carlson K, Aleskog A, Larsson R, Nygren P, Lindhagen E (2009) In vitro activity of bortezomib in cultures of patient tumour cells—potential utility in haematological malignancies. Med Oncol 26:193–201PubMedCrossRef Wiberg K, Carlson K, Aleskog A, Larsson R, Nygren P, Lindhagen E (2009) In vitro activity of bortezomib in cultures of patient tumour cells—potential utility in haematological malignancies. Med Oncol 26:193–201PubMedCrossRef
29.
go back to reference Fujita T, Washio K, Takabatake D et al (2005) Proteasome inhibitors can alter the signaling pathways and attenuate the P-glycoprotein-mediated multidrug resistance. Int J Cancer 117:670–682PubMedCrossRef Fujita T, Washio K, Takabatake D et al (2005) Proteasome inhibitors can alter the signaling pathways and attenuate the P-glycoprotein-mediated multidrug resistance. Int J Cancer 117:670–682PubMedCrossRef
30.
go back to reference Lu S, Chen Z, Yang J et al (2010) The effects of proteasome inhibitor bortezomib on a P-gp positive leukemia cell line K562/A02. Int J Lab Hematol 32(1 pt 1):e123–131 Lu S, Chen Z, Yang J et al (2010) The effects of proteasome inhibitor bortezomib on a P-gp positive leukemia cell line K562/A02. Int J Lab Hematol 32(1 pt 1):e123–131
31.
go back to reference Minderman H, Zhou Y, O’Loughlin KL, Baer MR (2007) Bortezomib activity and in vitro interactions with anthracyclines and cytarabine in acute myeloid leukemia cells are independent of multidrug resistance mechanisms and p53 status. Cancer Chemother Pharmacol 60:245–255PubMedCrossRef Minderman H, Zhou Y, O’Loughlin KL, Baer MR (2007) Bortezomib activity and in vitro interactions with anthracyclines and cytarabine in acute myeloid leukemia cells are independent of multidrug resistance mechanisms and p53 status. Cancer Chemother Pharmacol 60:245–255PubMedCrossRef
32.
go back to reference Mitsiades N, Mitsiades CS, Richardson PG et al (2003) The proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conventional chemotherapeutic agents: therapeutic applications. Blood 101:2377–2380PubMedCrossRef Mitsiades N, Mitsiades CS, Richardson PG et al (2003) The proteasome inhibitor PS-341 potentiates sensitivity of multiple myeloma cells to conventional chemotherapeutic agents: therapeutic applications. Blood 101:2377–2380PubMedCrossRef
33.
go back to reference Schwarzenbach H (2002) Expression of MDR1/P-glycoprotein, the multidrug resistance protein MRP, and the lung-resistance protein LRP in multiple myeloma. Med Oncol 19:87–104PubMedCrossRef Schwarzenbach H (2002) Expression of MDR1/P-glycoprotein, the multidrug resistance protein MRP, and the lung-resistance protein LRP in multiple myeloma. Med Oncol 19:87–104PubMedCrossRef
34.
go back to reference Kuo MT, Liu Z, Wei Y et al (2002) Induction of human MDR1 gene expression by 2-acetylaminofluorene is mediated by effectors of the phosphoinositide 3-kinase pathway that activate NF-kappaB signaling. Oncogene 21:1945–1954PubMedCrossRef Kuo MT, Liu Z, Wei Y et al (2002) Induction of human MDR1 gene expression by 2-acetylaminofluorene is mediated by effectors of the phosphoinositide 3-kinase pathway that activate NF-kappaB signaling. Oncogene 21:1945–1954PubMedCrossRef
35.
go back to reference Thevenod F, Friedmann JM, Katsen AD, Hauser IA (2000) Up-regulation of multidrug resistance P-glycoprotein via nuclear factor-kappaB activation protects kidney proximal tubule cells from cadmium- and reactive oxygen species-induced apoptosis. J Biol Chem 275:1887–1896PubMedCrossRef Thevenod F, Friedmann JM, Katsen AD, Hauser IA (2000) Up-regulation of multidrug resistance P-glycoprotein via nuclear factor-kappaB activation protects kidney proximal tubule cells from cadmium- and reactive oxygen species-induced apoptosis. J Biol Chem 275:1887–1896PubMedCrossRef
36.
go back to reference Ros JE, Schuetz JD, Geuken M et al (2001) Induction of Mdr1b expression by tumor necrosis factor-alpha in rat liver cells is independent of p53 but requires NF-kappaB signaling. Hepatology 33:1425–1431PubMedCrossRef Ros JE, Schuetz JD, Geuken M et al (2001) Induction of Mdr1b expression by tumor necrosis factor-alpha in rat liver cells is independent of p53 but requires NF-kappaB signaling. Hepatology 33:1425–1431PubMedCrossRef
37.
go back to reference Zhang Y, Shi Y, Li X et al (2008) Proteasome inhibitor MG132 reverses multidrug resistance of gastric cancer through enhancing apoptosis and inhibiting P-gp. Cancer Biol Ther 7:540–546PubMedCrossRef Zhang Y, Shi Y, Li X et al (2008) Proteasome inhibitor MG132 reverses multidrug resistance of gastric cancer through enhancing apoptosis and inhibiting P-gp. Cancer Biol Ther 7:540–546PubMedCrossRef
38.
go back to reference Bentires-Alj M, Barbu V, Fillet M et al (2003) NF-kappaB transcription factor induces drug resistance through MDR1 expression in cancer cells. Oncogene 22:90–97PubMedCrossRef Bentires-Alj M, Barbu V, Fillet M et al (2003) NF-kappaB transcription factor induces drug resistance through MDR1 expression in cancer cells. Oncogene 22:90–97PubMedCrossRef
39.
go back to reference Werts ED, DeGowin RL, Knapp SK, Gibson DP (1980) Characterization of marrow stromal (fibroblastoid) cells and their association with erythropoiesis. Exp Hematol 8:423–433PubMed Werts ED, DeGowin RL, Knapp SK, Gibson DP (1980) Characterization of marrow stromal (fibroblastoid) cells and their association with erythropoiesis. Exp Hematol 8:423–433PubMed
40.
go back to reference Chauhan D, Uchiyama H, Akbarali Y et al (1996) Multiple myeloma cell adhesion-induced interleukin-6 expression in bone marrow stromal cells involves activation of NF-kappa B. Blood 87:1104–1112PubMed Chauhan D, Uchiyama H, Akbarali Y et al (1996) Multiple myeloma cell adhesion-induced interleukin-6 expression in bone marrow stromal cells involves activation of NF-kappa B. Blood 87:1104–1112PubMed
41.
go back to reference Hideshima T, Chauhan D, Schlossman R, Richardson P, Anderson KC (2001) The role of tumor necrosis factor alpha in the pathophysiology of human multiple myeloma: therapeutic applications. Oncogene 20:4519–4527PubMedCrossRef Hideshima T, Chauhan D, Schlossman R, Richardson P, Anderson KC (2001) The role of tumor necrosis factor alpha in the pathophysiology of human multiple myeloma: therapeutic applications. Oncogene 20:4519–4527PubMedCrossRef
42.
go back to reference Urashima M, Ogata A, Chauhan D et al (1996) Transforming growth factor-beta1: differential effects on multiple myeloma versus normal B cells. Blood 87:1928–1938PubMed Urashima M, Ogata A, Chauhan D et al (1996) Transforming growth factor-beta1: differential effects on multiple myeloma versus normal B cells. Blood 87:1928–1938PubMed
43.
go back to reference Podar K, Anderson KC (2005) The pathophysiologic role of VEGF in hematologic malignancies: therapeutic implications. Blood 105:1383–1395PubMedCrossRef Podar K, Anderson KC (2005) The pathophysiologic role of VEGF in hematologic malignancies: therapeutic implications. Blood 105:1383–1395PubMedCrossRef
44.
go back to reference Jakubikova J, Adamia S, Kost-Alimova M et al (2011) Lenalidomide targets clonogenic side population in multiple myeloma: pathophysiologic and clinical implications. Blood 117:4409–4419PubMedCrossRef Jakubikova J, Adamia S, Kost-Alimova M et al (2011) Lenalidomide targets clonogenic side population in multiple myeloma: pathophysiologic and clinical implications. Blood 117:4409–4419PubMedCrossRef
45.
go back to reference McMillin DW, Negri JM, Mitsiades CS (2013) The role of tumour-stromal interactions in modifying drug response: challenges and oppurtunities. Nat Rev Drug Discov 12(3):217–228 McMillin DW, Negri JM, Mitsiades CS (2013) The role of tumour-stromal interactions in modifying drug response: challenges and oppurtunities. Nat Rev Drug Discov 12(3):217–228
46.
go back to reference McMillin DW, Delmore J, Negri J, Ooi M, Klippel S, Miduturu CV, Gray NS, Richardson PG, Anderson KC, Kung AL, Mitsiades CS (2011) Microenvironmental influence on pre-clinical activity of polo-like kinase inhibition in multiple myeloma: implications for clinical translation. PLoS One 6(7):e2022647 McMillin DW, Delmore J, Negri J, Ooi M, Klippel S, Miduturu CV, Gray NS, Richardson PG, Anderson KC, Kung AL, Mitsiades CS (2011) Microenvironmental influence on pre-clinical activity of polo-like kinase inhibition in multiple myeloma: implications for clinical translation. PLoS One 6(7):e2022647
47.
go back to reference Pessina A, Piccirillo M, Mineo E et al (1999) Role of SR-4987 stromal cells in the modulation of doxorubicin toxicity to in vitro granulocyte-macrophage progenitors (CFU-GM). Life Sci 65:513–523PubMedCrossRef Pessina A, Piccirillo M, Mineo E et al (1999) Role of SR-4987 stromal cells in the modulation of doxorubicin toxicity to in vitro granulocyte-macrophage progenitors (CFU-GM). Life Sci 65:513–523PubMedCrossRef
48.
go back to reference Perez LE, Parquet N, Meads M, Anasetti C, Dalton W (2010) Bortezomib restores stroma-mediated APO2L/TRAIL apoptosis resistance in multiple myeloma. Eur J Haematol 84(3):212–222 Perez LE, Parquet N, Meads M, Anasetti C, Dalton W (2010) Bortezomib restores stroma-mediated APO2L/TRAIL apoptosis resistance in multiple myeloma. Eur J Haematol 84(3):212–222
49.
go back to reference Garcia MG, Alaniz LD, Cordo Russo RI, Alvarez E, Hajos SE (2009) PI3K/Akt inhibition modulates multidrug resistance and activates NF-kappaB in murine lymphoma cell lines. Leuk Res 33:288–296PubMedCrossRef Garcia MG, Alaniz LD, Cordo Russo RI, Alvarez E, Hajos SE (2009) PI3K/Akt inhibition modulates multidrug resistance and activates NF-kappaB in murine lymphoma cell lines. Leuk Res 33:288–296PubMedCrossRef
50.
go back to reference Petrylak DP, Scher HI, Reuter V, O’Brien JP, Cordon-Cardo C (1994) P-glycoprotein expression in primary and metastatic transitional cell carcinoma of the bladder. Ann Oncol 5:835–840PubMed Petrylak DP, Scher HI, Reuter V, O’Brien JP, Cordon-Cardo C (1994) P-glycoprotein expression in primary and metastatic transitional cell carcinoma of the bladder. Ann Oncol 5:835–840PubMed
51.
go back to reference Mitsiades CS, Davies FE, Laubach JP et al (2011) Future directions of next-generation novel therapies, combination approaches, and the development of personalized medicine in myeloma. J Clin Oncol 29:1916–1923PubMedCrossRef Mitsiades CS, Davies FE, Laubach JP et al (2011) Future directions of next-generation novel therapies, combination approaches, and the development of personalized medicine in myeloma. J Clin Oncol 29:1916–1923PubMedCrossRef
Metadata
Title
The interaction of bortezomib with multidrug transporters: implications for therapeutic applications in advanced multiple myeloma and other neoplasias
Authors
Robert O’Connor
Melissa G. Ooi
Justine Meiller
Jana Jakubikova
Steffen Klippel
Jake Delmore
Paul Richardson
Kenneth Anderson
Martin Clynes
Constantine S. Mitsiades
Peter O’Gorman
Publication date
01-05-2013
Publisher
Springer-Verlag
Published in
Cancer Chemotherapy and Pharmacology / Issue 5/2013
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-013-2136-7

Other articles of this Issue 5/2013

Cancer Chemotherapy and Pharmacology 5/2013 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