Skip to main content
Erschienen in: Molecular Cancer 1/2015

Open Access 01.12.2015 | Short communication

The sodium channel-blocking antiepileptic drug phenytoin inhibits breast tumour growth and metastasis

verfasst von: Michaela Nelson, Ming Yang, Adam A Dowle, Jerry R Thomas, William J Brackenbury

Erschienen in: Molecular Cancer | Ausgabe 1/2015

Abstract

Background

Voltage-gated Na+ channels (VGSCs) are heteromeric protein complexes containing pore-forming α subunits and smaller, non-pore-forming β subunits. VGSCs are classically expressed in electrically excitable cells, e.g. neurons. VGSCs are also expressed in tumour cells, including breast cancer (BCa) cells, where they enhance cellular migration and invasion. However, despite extensive work defining in detail the molecular mechanisms underlying the expression of VGSCs and their pro-invasive role in cancer cells, there has been a notable lack of clinically relevant in vivo data exploring their value as potential therapeutic targets.

Findings

We have previously reported that the VGSC-blocking antiepileptic drug phenytoin inhibits the migration and invasion of metastatic MDA-MB-231 cells in vitro. The purpose of the present study was to establish whether VGSCs might be viable therapeutic targets by testing the effect of phenytoin on tumour growth and metastasis in vivo. We found that expression of Nav1.5, previously detected in MDA-MB-231 cells in vitro, was retained on cells in orthotopic xenografts. Treatment with phenytoin, at a dose equivalent to that used to treat epilepsy (60 mg/kg; daily), significantly reduced tumour growth, without affecting animal weight. Phenytoin also reduced cancer cell proliferation in vivo and invasion into surrounding mammary tissue. Finally, phenytoin significantly reduced metastasis to the liver, lungs and spleen.

Conclusions

This is the first study showing that phenytoin reduces breast tumour growth and metastasis in vivo. We propose that pharmacologically targeting VGSCs, by repurposing antiepileptic or antiarrhythmic drugs, should be further studied as a potentially novel anti-cancer therapy.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12943-014-0277-x) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MN and MY carried out the tumour growth and drug studies. AAD and JRT carried out and analysed the LC-SRM-MS. WJB designed the study, performed immunohistochemistry, and drafted the manuscript. All authors read and approved the final manuscript.
Abkürzungen
AED
Antiepileptic drug
BCa
Breast cancer
CAM
Cell adhesion molecule
LC-SRM-MS
Liquid chromatography-mass spectrometry with single reaction monitoring
MMP9
Matrix metalloproteinase-9
VGSC
Voltage-gated Na+ channel

Findings

Despite recent advances, breast cancer (BCa) is still the leading cause of cancer-related deaths in women [1]. Metastasis, the spread of tumours to secondary sites, is responsible for 90% of these deaths and is rarely curable [2]. Thus, there is an urgent need to identify new molecular targets and curative therapies. Voltage-gated Na+ channels (VGSCs) contain a pore-forming α subunit with smaller β subunits. There are nine α subunits, Nav1.1-Nav1.9, and four β subunits, β1-β4. The β subunits modulate channel function and are cell adhesion molecules (CAMs) [3]. VGSCs transmit electrical activity in cells in the nervous system and regulate neuronal growth and migration during CNS development [4]. VGSCs are clinical targets for a range of disorders, including epilepsy, cardiac arrhythmias, neuropathic pain and depression [5].
VGSCs are widely expressed in traditionally non-excitable cells, including microglia, astrocytes, immune cells, fibroblasts and cancer cells [6]. In the latter, a number of studies have shown that VGSCs contribute to cellular migration and invasion [7]. Nav1.5 is up-regulated in breast tumours, associating with recurrence, metastasis, and reduced survival [8,9]. Nav1.5 carries a fast inward Na+ current in triple negative (lacking estrogen receptor, progesterone receptor and HER2) MDA-MB-231 cells [9-11]. Pharmacological or genetic ablation of this Na+ current inhibits in vitro cell behaviours associated with the metastatic cascade, including migration, galvanotaxis, and invasion [9-11]. Similar results have been reported in metastatic cell lines from other cancers, suggesting that VGSC expression/activity in cancer may be a general phenomenon [7,12]. Na+ current enhances invasion by promoting cysteine cathepsin activity in caveolae via allosteric regulation of the Na+/H+ exchanger type 1 [13], and Nav1.5 is a key regulator of a gene network that controls invasion [14]. In addition, the widely used VGSC-blocking Class Ib antiarrhythmic agent and antiepileptic drug (AED) phenytoin (5,5-diphenylhydantoin) reduces the migration and invasion of MDA-MB-231 cells in vitro [8]. Furthermore, we have recently shown that the VGSC β1 subunit is also expressed in BCa specimens, and accelerates tumour growth and metastasis in a mouse model [15].
Together, these data highlight the potential for VGSCs as novel molecular targets. However, there remains a paucity of clinically relevant in vivo data exploring their potential therapeutic value. The aim of the present study was to study the effect of phenytoin on tumour growth and metastasis in a mouse model of triple negative BCa. We found that systemic phenytoin treatment reduces cellular proliferation, tumour growth, local invasion and metastasis. This is the first in vivo study demonstrating the potential therapeutic value of pharmacologically targeting VGSCs in BCa using an AED.

Phenytoin reduces tumour growth

Nav1.5 is expressed on cancer cells from breast tumours in clinical specimens, and in MDA-MB-231 cells cultured in vitro [8-11]. Here, we studied VGSC expression in tumours following orthotopic implantation of luciferase-expressing MDA-MB-231 cells into the mammary fat pad of female Rag2 -/- Il2rg -/- mice, a robust model of BCa growth and metastasis [15]. All methods are described in detail in Additional file 1. Nav1.5 expression, detected by immunohistochemistry, was retained in the tumours in vivo (Figure 1Ai). Nav1.7 was also present in the tumours, although expression was weaker (Figure 1Aii). These data agree with previous in vitro studies showing that although Nav1.5 is the predominant VGSC in MDA-MB-231 cells, accounting for >80% of Na+ current, there may be a small contribution from other isotypes, e.g. Nav1.7 [9,11]. We have previously shown that phenytoin inhibits Na+ current and VGSC-dependent migration in MDA-MB-231 cells in culture, suggesting that pharmacological targeting of VGSCs may have therapeutic utility in BCa [8]. In order to test the effect of phenytoin on BCa progression in vivo, we next treated tumour-bearing mice with 60 mg/kg phenytoin or vehicle (by daily intraperitoneal injection) for three weeks, starting one week after orthotopic implantation of MDA-MB-231 cells (Figure 1B). This dosing regimen gave a phenytoin trough level of 9.0 ± 1.0 μg/ml plasma, measured by liquid chromatography-mass spectrometry with single reaction monitoring (LC-SRM-MS) on samples taken 16 h after the last dose, which is within the therapeutic range for epilepsy treatment in rodents (6-23 μg/ml) [16]. We have previously shown that a similar concentration (50 μM) blocks Na+ current in MDA-MB-231 cells by 43% [8]. Importantly, there were no obvious signs of toxicity associated with the phenytoin treatment, and animal weight remained constant throughout the experiment (Figure 1C). Photon flux from tumours increased more slowly in phenytoin-treated animals than control-treated animals, indicating that the drug reduced the rate of tumour growth (Figure 1D,E). We also analysed tumour growth by calliper measurement. As with the bioluminescent data, the volume of tumours increased more slowly in phenytoin-treated animals than in control animals, indicating that phenytoin slowed the rate of tumour growth (Figure 1F).

Phenytoin reduces invasion and proliferation

We next studied the effect of phenytoin treatment on the structure and composition of the primary tumours. At the tumour periphery, there was some local invasion into surrounding skeletal muscle and fibroadipose tissue, and this invasion was moderately reduced (indicated by arrows) in phenytoin-treated animals compared to control (Figure 2A). Various MMPs, e.g. MMP9, are expressed in carcinomas, correlating with local invasion [17]. We found that the density of MMP9-expressing cells was significantly reduced by 51.9% in the tumours of phenytoin-treated animals (P < 0.01; Figure 2B,F). Together, these data suggest that phenytoin reduces local invasion from tumours in vivo, as it does in the same cells cultured in vitro [8].
We found that the prevalence of Ki67-expressing cycling cells was reduced by 62.6% in the tumours of phenytoin-treated animals (P < 0.001; Figure 2C,G). However, the number of apoptotic cells expressing activated caspase-3 was unchanged (Figure 2D,H). Similarly, the phenytoin treatment had no effect on the density of CD31-expressing vascular structures (Figure 2E,I). Together, these data suggest that phenytoin inhibited growth of primary tumours by reducing the number of proliferating cancer cells, rather than by inhibiting angiogenesis or promoting apoptosis. Interestingly, previous studies have indicated that VGSCs do not regulate proliferation of MDA-MB-231 cells in 2D cultures in vitro [9,10]. However, the VGSC blocker tetrodotoxin reduces colony growth in 3D Matrigel matrices [18]. Thus, the contribution of VGSCs to tumour growth in vivo appears complex, and may be dependent on multiple factors, including heterotypic signalling interactions with adjacent cells or the extracellular matrix [15]. In addition, VGSCs may regulate proliferation via reverse Na+/Ca2+ exchange, as has recently been shown in astrocytes after injury [19].

Phenytoin reduces metastasis

When we monitored metastasis 3 weeks after onset of drug treatment, following post mortem resection of the primary tumour (Figure 3A), photon flux was significantly reduced across the whole body, chest and abdomen of phenytoin-treated animals compared to control animals (P < 0.01; Figure 3B). Similarly, there was a notable reduction in photon flux across metastatic sites measured ex vivo (P < 0.01; Figure 3C). In order to further study metastasis to these sites at the cellular level, we next measured the number of GFP-expressing tumour cells within tissue sections. We have previously shown that GFP expression is retained in MDA-MB-231 cells at metastatic sites in this mouse model [15]. The number of GFP-expressing cells was moderately reduced in the liver of phenytoin-treated animals by 35.4% (P < 0.05; Figure 3D,G). Phenytoin treatment caused a more robust reduction in the density of metastatic cells in the lungs and spleen, of 66.3% and 92.4%, respectively (P < 0.001; Figure 3E,F,H,I). In summary, phenytoin treatment reduced BCa metastasis in vivo.

Therapeutic potential

We have previously shown that phenytoin inhibits Na+ current and significantly reduces migration and invasion of BCa cells in vitro [8]. Together with the present data, these findings suggest that targeting VGSC-mediated Na+ current with phenytoin may have therapeutic value. Phenytoin also inhibits migration and secretion in prostate cancer cells [20,21], suggesting that it may have broad utility in other cancers. In support of this, tetrodotoxin has been shown to reduce metastasis in a rat prostate cancer model [22]. In the present study, we provide, for the first time, clinically relevant in vivo data showing that pharmacological targeting of VGSCs with phenytoin significantly reduces tumour growth, local invasion and metastasis in a mouse model of BCa. Indeed, given that the membrane potential (Vm) of cancer cells is relatively depolarized [23], and that phenytoin displays robust use-dependent and tonic channel block at depolarized voltages [8], our data suggest that phenytoin may be a highly effective VGSC blocker in tumours.
We propose that VGSCs may be useful molecular targets for BCa therapy, and that repurposing FDA-approved, VGSC-targeting AEDs and Class I antiarrhythmic agents, e.g. phenytoin, carbamazepine, flecainide, to cancer may therefore improve outcome. It is possible that phenytoin may be effective in combination with existing conventional therapies, e.g. in the adjuvant setting, which would need to be tested in a randomised controlled clinical trial. In support of this notion, application of VGSC-targeting local anaesthetics during radical prostatectomy associates with substantially reduced recurrence and metastasis [24]. In addition, the FDA-approved ALS drug, riluzole, which inhibits both metabotropic glutamate receptors and VGSCs, reduces tumour growth [25]. Furthermore, given that VGSCs favour an invasive/metastatic phenotype [9,13-15], it is possible that the adjuvant prescription of AEDs, which cross the blood-brain-barrier, may reduce and/or delay metastasis formation in patients. This would transform the landscape of cancer treatment considerably, with very little added cost, while leading to healthier patients and huge financial savings.

Acknowledgments

This work was supported by the Medical Research Council [Fellowship G1000508].
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MN and MY carried out the tumour growth and drug studies. AAD and JRT carried out and analysed the LC-SRM-MS. WJB designed the study, performed immunohistochemistry, and drafted the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90.PubMedCrossRef Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90.PubMedCrossRef
2.
4.
Zurück zum Zitat Brackenbury WJ, Calhoun JD, Chen C, Miyazaki H, Nukina N, Oyama F, et al. Functional reciprocity between Na + channel Nav1.6 and β1 subunits in the coordinated regulation of excitability and neurite outgrowth. Proc Natl Acad Sci U S A. 2010;107:2283–8.PubMedCentralPubMedCrossRef Brackenbury WJ, Calhoun JD, Chen C, Miyazaki H, Nukina N, Oyama F, et al. Functional reciprocity between Na + channel Nav1.6 and β1 subunits in the coordinated regulation of excitability and neurite outgrowth. Proc Natl Acad Sci U S A. 2010;107:2283–8.PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Mantegazza M, Curia G, Biagini G, Ragsdale DS, Avoli M. Voltage-gated sodium channels as therapeutic targets in epilepsy and other neurological disorders. Lancet Neurol. 2010;9:413–24.PubMedCrossRef Mantegazza M, Curia G, Biagini G, Ragsdale DS, Avoli M. Voltage-gated sodium channels as therapeutic targets in epilepsy and other neurological disorders. Lancet Neurol. 2010;9:413–24.PubMedCrossRef
6.
Zurück zum Zitat Black JA, Waxman SG. Noncanonical roles of voltage-gated sodium channels. Neuron. 2013;80:280–91.PubMedCrossRef Black JA, Waxman SG. Noncanonical roles of voltage-gated sodium channels. Neuron. 2013;80:280–91.PubMedCrossRef
7.
Zurück zum Zitat Brackenbury WJ. Voltage-gated sodium channels and metastatic disease. Channels (Austin). 2012;6:352–61.CrossRef Brackenbury WJ. Voltage-gated sodium channels and metastatic disease. Channels (Austin). 2012;6:352–61.CrossRef
8.
Zurück zum Zitat Yang M, Kozminski DJ, Wold LA, Modak R, Calhoun JD, Isom LL, et al. Therapeutic potential for phenytoin: targeting Na(v)1.5 sodium channels to reduce migration and invasion in metastatic breast cancer. Breast Cancer Res Treat. 2012;134:603–15.PubMedCentralPubMedCrossRef Yang M, Kozminski DJ, Wold LA, Modak R, Calhoun JD, Isom LL, et al. Therapeutic potential for phenytoin: targeting Na(v)1.5 sodium channels to reduce migration and invasion in metastatic breast cancer. Breast Cancer Res Treat. 2012;134:603–15.PubMedCentralPubMedCrossRef
9.
Zurück zum Zitat Fraser SP, Diss JK, Chioni AM, Mycielska M, Pan H, Yamaci RF, et al. Voltage-gated sodium channel expression and potentiation of human breast cancer metastasis. Clin Cancer Res. 2005;11:5381–9.PubMedCrossRef Fraser SP, Diss JK, Chioni AM, Mycielska M, Pan H, Yamaci RF, et al. Voltage-gated sodium channel expression and potentiation of human breast cancer metastasis. Clin Cancer Res. 2005;11:5381–9.PubMedCrossRef
10.
Zurück zum Zitat Roger S, Besson P, Le Guennec JY. Involvement of a novel fast inward sodium current in the invasion capacity of a breast cancer cell line. Biochim Biophys Acta. 2003;1616:107–11.PubMedCrossRef Roger S, Besson P, Le Guennec JY. Involvement of a novel fast inward sodium current in the invasion capacity of a breast cancer cell line. Biochim Biophys Acta. 2003;1616:107–11.PubMedCrossRef
11.
Zurück zum Zitat Brackenbury WJ, Chioni AM, Diss JK, Djamgoz MB. The neonatal splice variant of Nav1.5 potentiates in vitro metastatic behaviour of MDA-MB-231 human breast cancer cells. Breast Cancer Res Treat. 2007;101:149–60.PubMedCentralPubMedCrossRef Brackenbury WJ, Chioni AM, Diss JK, Djamgoz MB. The neonatal splice variant of Nav1.5 potentiates in vitro metastatic behaviour of MDA-MB-231 human breast cancer cells. Breast Cancer Res Treat. 2007;101:149–60.PubMedCentralPubMedCrossRef
12.
Zurück zum Zitat Brackenbury WJ, Djamgoz MB, Isom LL. An emerging role for voltage-gated Na + channels in cellular migration: regulation of central nervous system development and potentiation of invasive cancers. Neuroscientist. 2008;14:571–83.PubMedCentralPubMedCrossRef Brackenbury WJ, Djamgoz MB, Isom LL. An emerging role for voltage-gated Na + channels in cellular migration: regulation of central nervous system development and potentiation of invasive cancers. Neuroscientist. 2008;14:571–83.PubMedCentralPubMedCrossRef
13.
Zurück zum Zitat Brisson L, Driffort V, Benoist L, Poet M, Counillon L, Antelmi E, et al. NaV1.5 Na(+) channels allosterically regulate the NHE-1 exchanger and promote the activity of breast cancer cell invadopodia. J Cell Sci. 2013;126:4835–42.PubMedCrossRef Brisson L, Driffort V, Benoist L, Poet M, Counillon L, Antelmi E, et al. NaV1.5 Na(+) channels allosterically regulate the NHE-1 exchanger and promote the activity of breast cancer cell invadopodia. J Cell Sci. 2013;126:4835–42.PubMedCrossRef
14.
Zurück zum Zitat House CD, Vaske CJ, Schwartz A, Obias V, Frank B, Luu T, et al. Voltage-gated Na + channel SCN5A is a key regulator of a gene transcriptional network that controls colon cancer invasion. Cancer Res. 2010;70:6957–67.PubMedCentralPubMedCrossRef House CD, Vaske CJ, Schwartz A, Obias V, Frank B, Luu T, et al. Voltage-gated Na + channel SCN5A is a key regulator of a gene transcriptional network that controls colon cancer invasion. Cancer Res. 2010;70:6957–67.PubMedCentralPubMedCrossRef
15.
Zurück zum Zitat Nelson M, Millican-Slater R, Forrest LC, Brackenbury WJ. The sodium channel beta1 subunit mediates outgrowth of neurite-like processes on breast cancer cells and promotes tumour growth and metastasis. Int J Cancer. 2014;135:2338–51.PubMedCentralPubMedCrossRef Nelson M, Millican-Slater R, Forrest LC, Brackenbury WJ. The sodium channel beta1 subunit mediates outgrowth of neurite-like processes on breast cancer cells and promotes tumour growth and metastasis. Int J Cancer. 2014;135:2338–51.PubMedCentralPubMedCrossRef
16.
Zurück zum Zitat Loscher W. The pharmacokinetics of antiepileptic drugs in rats: consequences for maintaining effective drug levels during prolonged drug administration in rat models of epilepsy. Epilepsia. 2007;48:1245–58.PubMedCrossRef Loscher W. The pharmacokinetics of antiepileptic drugs in rats: consequences for maintaining effective drug levels during prolonged drug administration in rat models of epilepsy. Epilepsia. 2007;48:1245–58.PubMedCrossRef
17.
Zurück zum Zitat Borges S, Doppler H, Perez EA, Andorfer CA, Sun Z, Anastasiadis PZ, et al. Pharmacologic reversion of epigenetic silencing of the PRKD1 promoter blocks breast tumor cell invasion and metastasis. Breast Cancer Res. 2013;15:R66.PubMedCentralPubMedCrossRef Borges S, Doppler H, Perez EA, Andorfer CA, Sun Z, Anastasiadis PZ, et al. Pharmacologic reversion of epigenetic silencing of the PRKD1 promoter blocks breast tumor cell invasion and metastasis. Breast Cancer Res. 2013;15:R66.PubMedCentralPubMedCrossRef
18.
Zurück zum Zitat Gillet L, Roger S, Besson P, Lecaille F, Gore J, Bougnoux P, et al. Voltage-gated sodium channel activity promotes cysteine cathepsin-dependent invasiveness and colony growth of human cancer cells. J Biol Chem. 2009;284:8680–91.PubMedCentralPubMedCrossRef Gillet L, Roger S, Besson P, Lecaille F, Gore J, Bougnoux P, et al. Voltage-gated sodium channel activity promotes cysteine cathepsin-dependent invasiveness and colony growth of human cancer cells. J Biol Chem. 2009;284:8680–91.PubMedCentralPubMedCrossRef
19.
Zurück zum Zitat Pappalardo LW, Samad OA, Black JA, Waxman SG. Voltage-gated sodium channel Nav 1.5 contributes to astrogliosis in an in vitro model of glial injury via reverse Na+ /Ca2+ exchange. Glia. 2014;62:1162–75.PubMedCrossRef Pappalardo LW, Samad OA, Black JA, Waxman SG. Voltage-gated sodium channel Nav 1.5 contributes to astrogliosis in an in vitro model of glial injury via reverse Na+ /Ca2+ exchange. Glia. 2014;62:1162–75.PubMedCrossRef
20.
Zurück zum Zitat Abdul M, Hoosein N. Inhibition by anticonvulsants of prostate-specific antigen and interleukin-6 secretion by human prostate cancer cells. Anticancer Res. 2001;21:2045–8.PubMed Abdul M, Hoosein N. Inhibition by anticonvulsants of prostate-specific antigen and interleukin-6 secretion by human prostate cancer cells. Anticancer Res. 2001;21:2045–8.PubMed
21.
Zurück zum Zitat Fraser SP, Salvador V, Manning EA, Mizal J, Altun S, Raza M, et al. Contribution of functional voltage-gated Na+ channel expression to cell behaviors involved in the metastatic cascade in rat prostate cancer: I. lateral motility. J Cell Physiol. 2003;195:479–87.PubMedCrossRef Fraser SP, Salvador V, Manning EA, Mizal J, Altun S, Raza M, et al. Contribution of functional voltage-gated Na+ channel expression to cell behaviors involved in the metastatic cascade in rat prostate cancer: I. lateral motility. J Cell Physiol. 2003;195:479–87.PubMedCrossRef
22.
Zurück zum Zitat Yildirim S, Altun S, Gumushan H, Patel A, Djamgoz MB. Voltage-gated sodium channel activity promotes prostate cancer metastasis in vivo. Cancer Lett. 2012;323:58–61.PubMedCrossRef Yildirim S, Altun S, Gumushan H, Patel A, Djamgoz MB. Voltage-gated sodium channel activity promotes prostate cancer metastasis in vivo. Cancer Lett. 2012;323:58–61.PubMedCrossRef
24.
Zurück zum Zitat Biki B, Mascha E, Moriarty DC, Fitzpatrick JM, Sessler DI, Buggy DJ. Anesthetic technique for radical prostatectomy surgery affects cancer recurrence: a retrospective analysis. Anesthesiology. 2008;109:180–7.PubMedCrossRef Biki B, Mascha E, Moriarty DC, Fitzpatrick JM, Sessler DI, Buggy DJ. Anesthetic technique for radical prostatectomy surgery affects cancer recurrence: a retrospective analysis. Anesthesiology. 2008;109:180–7.PubMedCrossRef
25.
Zurück zum Zitat Speyer CL, Smith JS, Banda M, Devries JA, Mekani T, Gorski DH. Metabotropic glutamate receptor-1: a potential therapeutic target for the treatment of breast cancer. Breast Cancer Res Treat. 2012;132:565–73.PubMedCentralPubMedCrossRef Speyer CL, Smith JS, Banda M, Devries JA, Mekani T, Gorski DH. Metabotropic glutamate receptor-1: a potential therapeutic target for the treatment of breast cancer. Breast Cancer Res Treat. 2012;132:565–73.PubMedCentralPubMedCrossRef
Metadaten
Titel
The sodium channel-blocking antiepileptic drug phenytoin inhibits breast tumour growth and metastasis
verfasst von
Michaela Nelson
Ming Yang
Adam A Dowle
Jerry R Thomas
William J Brackenbury
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2015
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-014-0277-x

Weitere Artikel der Ausgabe 1/2015

Molecular Cancer 1/2015 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.