477
Views
9
CrossRef citations to date
0
Altmetric
Biology

Thermal sensitisation by lonidamine of human melanoma cells grown at low extracellular pH

, , , , &
Pages 75-78 | Received 26 Apr 2013, Accepted 20 Oct 2013, Published online: 03 Dec 2013

Abstract

Purpose: This study tested the ability of lonidamine (LND), a clinically applicable inhibitor of monocarboxylate transporters (MCT), to thermally sensitise human melanoma cells cultured at a tumour-like extracellular pH (pHe) 6.7.

Materials and methods: Human melanoma DB-1 cells cultured at pHe 6.7 and pHe 7.3 were exposed to 150 µM LND for 3 h, beginning 1 h prior to heating at 42 °C (2 h). Intracellular pH (pHi) was determined using 2′,7′-bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein (BCECF) and whole spectrum analysis. Levels of heat shock proteins (HSPs) were determined by immunoblot analysis. Cell survival was determined by colony formation.

Results: Treatment with LND at pHe 6.7 reduced pHi to 6.30 ± 0.21, reduced thermal induction of HSPs, and sensitised cells growing at pHe 6.7 to 42 °C. When LND was combined with an acute acidification from pHe 6.7 to pHe 6.5, pHi was reduced to 6.09 ± 0.26, and additional sensitisation was observed. LND had negligible effects on cells cultured at pH 7.3.

Conclusions: The results show that LND can reduce pHi in human melanoma cells cultured at a tumour-like low pHe so that the 42 °C induction of HSPs are abrogated and the cells are sensitised to thermal therapy. Cells cultured at a normal tissue-like pHe 7.3 were not sensitised to 42 °C by LND. These findings support the strategy that human melanoma cells growing in an acidic environment can be sensitised to thermal therapy in vivo by exposure to an MCT inhibitor such as LND.

Introduction

Lonidamine (LND) inhibits the plasma membrane monocarboxylate transporter 1 (MCT-1) [Citation1] and the mitochondrial pyruvate carrier (MPC) [Citation2,Citation3]. Treatment of malignant cells and tumours with LND increases lactate production and inhibits lactate efflux, reduces intracellular pH (pHi), and decreases the rate of oxygen utilisation [Citation1,Citation3–7]. LND has been reported to sensitise cells and tumours to hyperthermia [Citation4,Citation6,Citation8] and a variety of chemotherapeutic drugs [Citation3,Citation9–12]. Thermal sensitisation by LND can be attributable primarily to intracellular acidification.

Acute extracellular acidification is a potent thermal sensitiser, with sensitisation greater for temperatures below 43 °C than above 43 °C [Citation13,Citation14]. However, it is the decrease in pHi accompanying acute lowering of extracellular pH (pHe) that correlates with thermal sensitisation [Citation15–19]. Acute acidification sensitises to hyperthermia in part by abrogating thermal induction of heat shock proteins (HSP) [Citation19,Citation20]. Human melanoma cells growing at a tumour-like pHe 6.7 contain higher constitutive levels of HSP than in cells growing at pHe 7.3. This explains why human melanoma cells growing at pHe 6.7 are resistant to hyperthermia compared to cells growing at pHe 7.3 [Citation19,Citation20]. It is important, therefore, to block additional synthesis of HSPs during thermal therapy. Acute intracellular acidification sensitises cells and tumours to chemotherapy with platinum [Citation21] and nitrogen mustards [Citation22–25], as well as to hyperthermia [Citation8,Citation9,Citation18,Citation19].

Human melanoma cells depend primarily on MCT-1 to maintain pH homeostasis [Citation20,Citation26]. Since intracellular acidification sensitised DB1 human melanoma cells to mild hyperthermia (42 °C) [Citation19], it was proposed that inhibition of MCT-1 would sensitise the cells to hyperthermia. Inhibition of MCT-1 with alpha-cyano-4-hydroxy-cinnamic acid (CHC) reduced pHi in DB-1 cells cultured at pHe 6.7, abrogated induction of HSPs, and sensitised the cells to 42 °C [Citation20]. However, CHC is not translatable to the clinic. The purpose of this study was to determine whether LND, by virtue of its ability to reduce pHi, would sensitise DB1 cells grown at a tumour-like low pH to hyperthermia.

The clinical experience on combining LND and chemotherapy and/or radiation therapy in the treatment of solid tumours has been reviewed [Citation12]. The available data demonstrate a significant role of LND in modulating anthracycline and platinum compound activity. The encouraging results of phase II and III trials for the treatment of advanced breast, ovarian and lung cancer still must be confirmed by larger studies.

Materials and methods

Cell culture

The human melanoma cell line, DB-1, was derived from a patient biopsied at Thomas Jefferson University [Citation19]. DB-1 cells express human melanoma antigens [Citation27]. Cells were maintained in exponential growth at pHe 7.3 or 6.7 in alpha-MEM/10% FBS (pH adjusted by modifying the sodium bicarbonate concentration) as described previously [Citation19]. The osmolality was kept constant.

Inhibitor

LND (1-[(2,4-dichlorobenzyl)methyl]-1H-indazole-3-carboxylic acid) (Sigma, St Louis, MO, USA) was solubilised in DMSO. Cells were exposed to 150 µM LND for 1 h at 37 °C in complete medium prior to heating at 42 °C (2 h). DMSO controls were performed for all experiments.

Measurement of pHi

Cells plated onto 18 mm glass coverslips (attached to the bottom of a 1 cm diameter hole in the center of plastic Petri dishes) were loaded with BCECF-AM as described previously [Citation19,Citation20] with the following exceptions. After allowing for hydrolysis of the dye ester, the Petri dish was placed into a temperature-controlled microscope stage chamber (Instec, Boulder, CO) for study at 37 °C under humidified air containing 5% CO2. pHi was calculated based on analysis of whole excitation spectra of attached cells as has been described [Citation19,Citation20,Citation28–31]. Felix32 software (Photon Technology International (PTI), Lawrenceville, NJ) was used to generate whole excitation spectra using the RatioMaster Model RM-5/2003 microscope-based ratio fluorescence spectrometer system (PTI) integrated with a Nikon Eclipse TS100 (Nikon Instruments Inc., Melville, NY) inverted fluorescence microscope and a Plan Fluor 40X objective. The initial steady-state pHi at pHe 7.3, 7.1, 6.7 or 6.5, was measured several times on a field of 8–15 cells and then the medium was replaced with medium containing 150 µM LND. The pHi was measured on a single field of 8–15 cells every 5 min for up to 1 h. The reported pHi values are those determined after 1 h. The pHe values of the medium in the microscope chamber Petri dishes were measured immediately following the pHi measurements to confirm that the pHe of the medium had not changed.

Clonal survival

All tissue culture flasks (25 cm2) used for colony survival assays had a total of 105 cells, comprised of irradiated (30 Gy) Chinese hamster ovarian carcinoma feeder cells and unirradiated DB-1 cells. All media in tissue culture flasks were replaced with fresh, pre-warmed (37 °C), CO2-equilibrated media with or without LND at the treatment pHe 1 h prior to the hyperthermia treatment. The cells were heated by submersion of flasks in 42 °C water baths. Media was replaced with fresh, pre-warmed (37 °C), CO2-equilibrated media without LND at the growth pHe following heat treatment. The flasks were maintained in a humidified CO2 incubator at 37 °C for colony formation.

Immunoblot analysis

Samples were prepared for SDS-PAGE and western immunoblot analysis as described in detail previously [Citation19,Citation20]. Cohorts of cells were solubilised for SDS-PAGE just before addition of LND, and immediately following the 2-h 42 °C treatment. Cells exposed to LND alone were processed following the 3-h exposure to drug. Immunodetection was performed by enhanced chemiluminescence using a Tropix Western-Star protein detection kit (Applied Biosystems, Foster City, CA). The relative protein content of individual bands on X-ray film was determined by scanning laser densitometry using ImageQuant software (Molecular Dynamics, Sunnyvale, CA). The absorbance (A) obtained for each HSP band was normalised to the A of the GAPDH band in that lane and expressed as a percentage of the respective normalised HSP band from unheated cells at their growth pHe.

Results

Effect of LND on pHi

The steady-state pHi of DB-1 cells cultured at pHe 7.3 or pHe 6.7 was 7.20 ± 0.07 or 6.76 ± 0.04, respectively. A 1-h, 37 °C treatment with LND reduced the steady-state pHi in cells cultured at pHe 7.3 to 7.15 ± 0.16. However, a 1-h LND treatment reduced the steady-state pHi in cells cultured at pHe 6.7 to 6.30 ± 0.21. A 0.2-pH unit acute acidification combined with exposure to LND reduced pHi in cells grown at pHe 7.3 or pHe 6.7 to 6.93 ± 0.15 or 6.09 ± 0.26, respectively. The reduction in pHi plateaued after 15 min and was maintained for the next 45 min of exposure after which measurement was discontinued. This decrease in steady-state pHi indicates that MCT transport is constitutively active at pHe 6.7. summarises the results of the pHi studies.

Table I. Summary of the effects of 150 μM LND on pHi in DB-1 cells.

Effect of LND on 42 °C-induction of HSP

summarises the effects of LND on the induction of Hsp70 and Hsp27 by hyperthermia. Hsp70 and Hsp27 levels were measured before and immediately following 2 h of heating. HSP levels increased within 10–40 min from the start of 42 °C heating of mammalian cells [Citation32] and the HSP levels continued to increase during 6 h of heating [Citation32,Citation33]. Exposure to 150 µM LND minimally suppressed the 42 °C induction of Hsp70 and Hsp27 in cells grown at pHe 7.3. However, LND treatment abrogated the heat-induced increase in levels of HSP, especially Hsp70, in cells grown at pHe 6.7.

Figure 1. Abrogation of induction of HSP70 (A) and HSP27 (B) during heating by 150 μM LND. Cells grown at pHe 7.3 and pHe 6.7 were exposed to LND for 60 min prior to and during heating (42 °C, 2 h) without or with a 0.2-pH unit acute acidification. Flasks of cells were processed for immunoblot studies before treatments and immediately after the 2 h of heating. HSP levels are presented as percent of HSP levels in unheated cells at their growth pHe. The means (bars) and standard errors represent results from two different experiments. 37 °C (white); LND, 37 °C (stippled); 42 °C (gray); LND + 42 °C (black).

Figure 1. Abrogation of induction of HSP70 (A) and HSP27 (B) during heating by 150 μM LND. Cells grown at pHe 7.3 and pHe 6.7 were exposed to LND for 60 min prior to and during heating (42 °C, 2 h) without or with a 0.2-pH unit acute acidification. Flasks of cells were processed for immunoblot studies before treatments and immediately after the 2 h of heating. HSP levels are presented as percent of HSP levels in unheated cells at their growth pHe. The means (bars) and standard errors represent results from two different experiments. 37 °C (white); LND, 37 °C (stippled); 42 °C (gray); LND + 42 °C (black).

Effect of LND on cell survival

summarises the effects of treatments on cell survival. Exposure to LND at 37 °C had little effect on survival of cells grown at either pHe 7.3 or 6.7. Heating alone reduced survival for all conditions. While LND exposure sensitised cells grown at pHe 6.7 to 42 °C by a factor of 10, the inhibitor did not sensitise cells grown at pHe 7.3 to the heat treatment. Furthermore, an acute 0.2-pH unit acute acidification of cells grown at pHe 6.7 was not required for sensitisation by LND as it was for CHC.

Figure 2. Enhancement of cell killing by exposure of DB-1 cells to 150 μM LND prior to and during heating (42 °C, 2 h). Cells grown at pHe 7.3 and pHe 6.7 were exposed to LND for 60 min prior to and during heating (42 °C, 2 h) without or with a 0.2-pH unit acute acidification. The means (bars) and standard errors represent results from two different experiments. 37 °C (white); LND, 37 °C (stippled); 42 °C (gray); LND + 42 °C (black).

Figure 2. Enhancement of cell killing by exposure of DB-1 cells to 150 μM LND prior to and during heating (42 °C, 2 h). Cells grown at pHe 7.3 and pHe 6.7 were exposed to LND for 60 min prior to and during heating (42 °C, 2 h) without or with a 0.2-pH unit acute acidification. The means (bars) and standard errors represent results from two different experiments. 37 °C (white); LND, 37 °C (stippled); 42 °C (gray); LND + 42 °C (black).

Discussion and conclusion

This study documents in vitro that the MCT inhibitor LND reduces pHi in melanoma cells growing in a tumour-like acidic pHe environment such that they are sensitised to hyperthermia. The previously studied MCT inhibitor, CHC, required a 0.2-pH unit acute acidification to sensitise DB-1 cells growing at pHe 6.7 to 42 °C [Citation20]. However, the LND-induced reduction of pHi in DB-1 cells, resulting in abrogation of thermally induced HSP and thermal sensitisation, did not require an acute extracellular acidification. These findings suggest that hyperglycaemia may not be necessary to drive glycolysis in order to achieve tumour acidification by LND. Indeed, it was recently reported that LND reduced the pHi of DB-1 xenografts from 6.90 ± 0.05 to 6.33 ± 0.10 in nude mice without a prior induction of hyperglycaemia [Citation3].

Thermal sensitisation by LND is attributed to reduction in pHi [Citation4,Citation6,Citation7] and reduction in energy status [Citation5]. The effect of LND on intracellular acidification in cells and tumours is attributed to increased production of lactate and inhibition of lactate efflux [Citation1,Citation3,Citation7], inhibition of mitochondrial respiration by inhibiting pyruvate entry into mitochondria [Citation2,Citation3], and reduction in energy status [Citation3,Citation5]. These effects can be attributed to the drug’s inhibition of MCT [Citation3,Citation34–36] and MPC [Citation2,Citation3].

In conclusion, the results with LND support our previous findings with the MCT inhibitor CHC. However, an accompanying acute acidification is not required for reduction of pHi by LND to a value that abrogates the 42 °C-induced increase of HSP levels and sensitises to 42 °C hyperthermia. Accordingly, LND should be a good candidate for sensitisation of solid human tumours to thermal therapy [Citation37].

Declaration of interest

This research was supported by grant no. P01 CA56690 from the National Cancer Institute, the National Institutes of Health, of the US Department of Health and Human Services. The authors alone are responsible for the content and writing of the paper.

Acknowledgements

We thank Phyllis R. Wachsberger for proofreading this manuscript.

References

  • Fang J, Quinones QJ, Holman TL, Morowitz MJ, Wang Q, Zhao H, et al. The H+-linked monocarboxylate transporter (MCT1/SLC16A1): A potential therapeutic target for high-risk neuroblastoma. Mol Pharmacol 2006;70:2108–15
  • Floridi A, Lehninger AL. Action of the antitumor and antispermatogenic agent lonidamine on electron transport in Ehrlich ascites tumor mitochondria. Arch Biochem Biophys 1983;226:73–83
  • Nath K, Nelson DS, Ho AM, Lee, S-C, Darpolor MM, Pickup S, et al. 31P and 1H MRS of DB-1 melanoma xenografts: lonidamine selectively decreases tumor intracellular pH and energy status and sensitizes tumors to melphalan. NMR Biomed 2012;26:98–105
  • Kim JH, Kim SH, Alfieri A, Young CW, Silvestrini B. Lonidamine: A hyperthermic sensitizer of HeLa cells in culture and of the Meth-A tumor in vivo. Oncology 1984;41:30–5
  • Stryker JA, Gerweck LE. Lonidamine-induced, pH dependent inhibition of cellular oxygen utilization. Radiat Res 1988;113:356–61
  • Raaphorst GP, Feeley MM, Martin L, Danjoux CE, Maroun J, Desanctis AJ, et al. Enhancement of sensitivity to hyperthermia by lonidamine in human cancer cells. Int J Hyperthermia 1991;7:763–72
  • Ben-Yoseph O, Lyons JC, Song CW, Ross BD. Mechanism of action of lonidamine in the 9L brain tumor model involves inhibition of lactate efflux and intracellular acidification. J Neurooncol 1998;36:149–57
  • Silvestrini B, Hahn GM, Cioli V, De Martino C. Effects of lonidamine alone or combined with hyperthermia in some experimental cell and tumour systems. Br J Cancer 1983;47:221–31
  • Caputo A, Silvestrini B. Lonidamine, a new approach to cancer therapy. Oncology 1984;41:2–6
  • Citro G, Cucco C, Verdina A, Zupi G. Reversal of adriamycin resistance by lonidamine in a human breast cancer cell line. Br J Cancer 1991;64:534–6
  • Teicher BA, Herman TS, Holden SA, Epelbaum R, Liu S, Frei III E. Lonidamine as a modulator of alkylating agent activity in vitro and in vivo. Cancer Res 1991;51:780–4
  • Di Cosimo S, Ferretti G, Papaldo P, Carlini P, Fabi A, Cognetti F. Lonidamine: Efficacy and safety in clinical trials for the treatment of solid tumors. Drugs Today (Barc) 2003;39:157–74
  • Gerweck LE. Modification of cell lethality at elevated temperatures. The pH effect. Radiat Res 1977;70:224–35
  • Freeman ML, Raaphorst GP, Hopwood LE, Dewey WC. The effect of pH on cell lethality induced by hyperthermic treatment. Cancer 1980;45:2291–300
  • Hofer KG, Mivechi NF. Tumor cell sensitivity to hyperthermia as a function of extracellular and intracellular pH. J Natl Cancer Inst 1980;65:621–5
  • Chu G, Dewey WC. The role of low intracellular or extracellular pH in sensitization to hyperthermia. Radiat Res 1988;114:154–67
  • Chu GL, Wang Z, Hyun WC, Pershadsingh HA, Fulwyler MJ, Dewey WC. The role of intracellular pH and its variance in low pH sensitization of killing by hyperthermia. Radiat Res 1990;122:288–93
  • Lyons JC, Kim GE, Song CW. Modification of intracellular pH and thermosensitivity. Radiat Res 1992;129:79–87
  • Coss RA, Storck CW, Wachsberger PR, Reilly J, Leeper DB, Berd D, et al. Acute extracellular acidification reduces intracellular pH, 42 °C-induction of heat shock proteins and clonal survival of human melanoma cells grown at pH 6.7. Int J Hyperthermia 2004;20:93–106
  • Coss, RA, Storck CW, Daskalakis C, Berd D, Wahl ML. Intracellular acidification abrogates the heat shock response and compromises survival of human melanoma cells. Mol Cancer Ther 2003;2:383–8
  • Atema A, Buurman KJ, Noteboom E, Smets LA. Potentiation of DNA-adduct formation and cytotoxicity of platinum-containing drugs by low pH. Int J Cancer 1993;54:166–72
  • Jahde E, Glusenkamp KH, Klunder I, Hulser DF, Tietze LF, Rajewsky MF. Hydrogen ion-mediated enhancement of cytotoxicity of bis-chloroethylating drugs in rat mammary carcinoma cells in vitro. Cancer Res 1989;49:2965–72
  • Jahde E, Glusenkamp KH, Rajewsky MF. Nigericin enhances mafosfamide cytotoxicity at low extracellular pH. Cancer Chemother Pharmacol 1991;27:440–4
  • Jahde E, Roszinski S, Volk T, Glusenkamp KH, Wiedemann G, Rajewsky MF. Metabolic response of AH13r rat tumours to cyclophosphamide as monitored by pO2 and pH semi-microelectrodes. Eur J Cancer 1992;29:116–22
  • Gerweck LE, Vijayappa S, Kozin S. Tumor pH controls the in vivo efficacy of weak acid and base chemotherapeutics. Mol Cancer Ther 2006;5:1275–9
  • Wahl ML, Owen JA, Burd R, Herlands RA, Nogami SS, Rodeck U, et al. Regulation of intracellular pH in human melanoma: potential therapeutic implications. Mol Cancer Ther 2002;1:617–28
  • Hill LL, Korngold R, Jaworsky C, Murphy G, McCue P, Berd D. Growth and metastasis of fresh human melanoma tissue in mice with severe combined immunodeficiency. Cancer Res 1991;51:4937–41
  • Owen CS, Wahl ML, Leeper DB, Perry HD, Bobyock SB, Russel M, et al. Accurate whole spectrum measurements of intracellular pH and [Na+]. J Fluorescence 1995;5:329–35
  • Wahl ML, Coss RA, Bobyock SB, Leeper DB, Owen CS. Thermotolerance and intracellular pH in two Chinese hamster cell lines adapted to growth at low pH. J Cell Physiol 1996;166:438–45
  • Owen CS, Pooler PM, Wahl ML, Coss RA, Leeper DB. Altered proton extrusion in cells adapted to growth at low extracellular pH. J Cell Physiol 1997;167:397–405
  • Wahl ML, Pooler PM, Briand P, Leeper DB, Owen CS. Intracellular pH regulation in a nonmalignant and a derived malignant human breast cell line. J Cell Physiol 2000;18:373–89
  • Hickey ED, Weber LA. Modulation of heat-shock polypeptide synthesis in HeLa cells during hyperthermia and recovery. Biochemistry 1982;21:1513–21
  • Han J-S, Storck CW, Wachsberger PR, Leeper DB, Berd D, Wahl ML, et al. Acute extracellular acidification increases nuclear associated protein levels in human melanoma cells during 42 °C hyperthermia and enhances cell killing. Int J Hyperthermia 2002;18:404–15
  • Ben-Horin H, Tassini M, Vivi A, Navon G, Kaplan O. Mechanism of action of the antineoplastic drug lonidamine: 31P and 13C nuclear magnetic resonance studies. Cancer Res 1995;55:2814–21
  • Vivi A, Tassini M, Ben-Horin H, Navon G, Kaplan O. Comparison of action of the anti-neoplastic drug lonidamine on drug-sensitive and drug-resistant human breast cancer cells: 31P and 13C nuclear magnetic resonance studies. Breast Cancer Res Treat 1997;43:15–25
  • Mardor Y, Kaplan O, Sterin M, Ruiz-Cabello J, Ash E, Roth Y, et al. Noninvasive real-time monitoring of intracellular cancer cell metabolism and response to lonidamine treatment using diffusion weighted proton magnetic resonance spectroscopy. Cancer Res 2000;60:5179–86
  • Coss RA. Inhibiting induction of heat shock proteins as a strategy to enhance cancer therapy. Int J Hyperthermia 2005;21:695–701

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.