The role of ion channels in high-grade glioma (HGG)

Panimaya Jeffreena Miranda 1,2*, Caitlyn Richworth 1,3, Natalie Anderson 4,5,6

1 Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia.

2 Division of Paediatrics/Centre for Child Health Research, Medical School, University of Western Australia, Western Australia, Australia.

3 School of Biomedical Sciences, University of Western Australia, Western Australia, Australia.

4 Perioperative Care Program, Perioperative Medicine Team, Telethon Kids Institute, Nedlands, Australia

5 Division of Emergency Medicine, Anaesthesia and Pain Medicine, Medical School, The University of Western Australia, Perth, Australia

6 Institute for Paediatric Perioperative Excellence, The University of Western Australia, Perth, Australia

 

Corresponding Authors: Panimaya Jeffreena Miranda

 * Email: jeffybio@gmail.com

 

Abstract

High-grade glioma (HGG) is an aggressive brain cancer with an overall 5-year survival rate of less than 10% in adults and less than 2% in children. Despite significant research efforts, surgery combined with chemo- and radiotherapy is the only treatment option available for these patients. New targeted therapies such as kinase inhibitors, and combined modalities fail in clinical trials due to the inability of drugs to cross the blood-brain barrier, and HGG pathway rewiring. In vitro studies suggest that ion channels contribute to HGG pathway rewiring and tumor survival. There are several United States Food and Drug Administration-approved neurological drugs that readily cross the blood-brain barrier and target ion channels. These drugs are readily available on the shelf and can be easily repurposed to treat HGG. A systematic understanding of the oncogenic roles of ion channels in patients with HGG will help us to repurpose ion channel drugs to treat HGG. The study of the oncogenic potential and therapeutic targeting of ion channels in HGG is still in the early stage. This review summarises the findings that elucidate the expression and oncogenic potential of ion channels in HGG patients. We have identified the research gaps to translate ion channels as therapeutic targets for HGG. Finally, we highlight the potential to use ion channel drugs as a single agent or as part of combination therapy for the treatment of patients with HGG.

Keywords: High-grade glioma, Ion channels, Ion channel drugs, Tumor resistance, Drug repurposing

Introduction

Glial cells form 50% of the cells in the central nervous system where they support and protect neurons, form myelin, and facilitate cellular homeostasis (1). About a third of all brain cancers originate from glial cells and are known as gliomas. Gliomas are grouped based on their region of origin, grades/pathological features, and aggressiveness. Based on the region of origin, gliomas are subdivided as; diffuse intrinsic pontine glioma (DIPG), astrocytoma, oligodendrogliomas, brain stem glioma, optic nerve glioma, oligoastrocytoma, ependymoma (EPN) and glioblastoma multiforme (GBM) (2). Gliomas are classified as grades based on their pathological features (grade I, II, III and IV). Highly diffusive and invasive gliomas that belong to grades III and IV are collectively known as high-grade gliomas (HGG) and they invade through the extracellular space in the brain and central nervous system (3, 4).

The diffusive nature of HGG makes complete surgical resection impossible (2, 5, 6). Hence, surgery followed by chemo- and radiotherapy is the only treatment option for HGG patients, resulting in a very low 5-year survival rate of <10% (7). As HGGs belong to stage III and IV, most patients die within 18 months of diagnosis despite advances in treatment. Therefore the only way to measure the therapeutic effectiveness of drugs is via the increased survival rate of patients (8). HGG plasticity enables the tumor to rewire and adapt its pathways for tumor growth and drug resistance (8–10) however, the key regulators that drive HGG plasticity are not fully understood. This emphasises the need to identify novel therapeutic targets to prevent HGG plasticity and increase the survival of patients with HGG.

Recent advances in the field of HGG demonstrate a role for ion channels in both HGG proliferation and invasion (11–13). Ion channels are membrane structures that regulate the movement of ions across the cellular membrane to control various physiological functions including brain cell function (14–18). Normal brain cells maintain a high degree of plasticity to cope with continuous brain remodelling during memory formation and to recover from various forms of brain cell injury (19–21). Ion channels are the key regulators of brain cell plasticity that contribute to pathway rewiring in brain cells through their structural and functional alterations (22). Research suggests that cancer cells hijack this dynamic nature of ion channels to support their oncogenicity (23, 24). The role of ion channels in HGG tumors has been demonstrated primarily through retrospective correlational studies of ion channel aberrations and patient survival with genomic and proteomic studies (101, 102). While in vitro studies demonstrate that ion channel gene mutation and abnormal pore formation can drive HGG tumorigenicity (reviewed in (25–28), (12, 29–31), some studies suggest that kinase pathways activates ion channels in HGG (32). However, the functionality and oncogenic potential of many ion channels remain unknown.

Repurposing ion channel drugs for HGG treatment may represent an attractive therapeutic option for patients with HGG, as most clinically approved ion channel drugs are used to treat neurological disorders, and can easily cross the blood-brain barrier (BBB) (reviewed in (33)), (34). While literature does summarize the ability to target ion channels as cancer therapy using laboratory models (reviewed in (35), there are few studies that discuss ion channel expression patterns in HGG clinical/patient samples and correlate them with therapeutic options (36). Therefore, this review will discuss: 1) ion channel biology and its role in cancer, 2) expression and function of ion channels in HGG clinical samples, 3) ion channel therapeutics to treat cancer, including HGG and 4) areas for future research. We also briefly discuss ion channel expression patterns identified in HGG clinical samples and their correlation with patient prognosis and survival with relevance to the various types of ion channels. 

Overview of ion channel biology and its role in cancer

Ion channels are integral membrane proteins that maintain an electrochemical gradient for ion transport across cellular membranes (37). Each ion channel comprises of multiple protein subunits that are assembled together to form a pore-forming structure. This process is known as ion channel biogenesis (38–40). The concentration of different intracellular ions such as calcium, sodium, potassium and chloride play a key role in regulating ion channel biogenesis (Figure 1) (41). Ion channel expression and function is highly cell-type specific (37, 42, 43) (44). Ion channel expression is highly dependent on biological processes including transcription, translation, protein processing, subunit assembly and transportation of ion channel genes. Each cell type may express more than ten different types of ion channels (45).


Figure 1. Regulation of ion channel expression. Ion channel biogenesis is a multistep process and is regulated by intracellular ionic concentration. Extracellular ionic concentration is responsible to keep an ion channel either in the open or closed state (41). 


The function of an ion channel is dependent on its activation state (reviewed in (44)) and ion channels can swing between both active and inactive states (46). The activation state is regulated by multiple factors including gating strategy and the channel orientation across the cell membrane (reviewed in (47–51)). Multiple ion channels share common roles and possess complementary and compensatory functions to effectively maintain the cellular membrane potential (52). The ability of ion channels to constantly change between their activation states make them particularly susceptible to malignant transformation (53). For example, changes in the extracellular ionic concentration results in an acidic tumor microenvironment that is demonstrated to increase cell proliferation and invasion (38, 54), (reviewed in (55–57)). Genomic instability or protein dysfunction of ion channels (58), (reviewed in (56, 59)) is the most common malignant transformation that alters the activation state of ion channels. Their functions include altering key intracellular processes such as transcription, cellular secretion and cell volume to regulate cell proliferation, autophagy and cell cycle (60–62), (reviewed in (63–68) 69–72). Thus, ion channels are dynamic in their function and undergo constant structural, functional, and activation changes to coordinate and maintain an electrochemical gradient. Ligand binding, electrical flux or voltage alterations or a combination of these maintains the activation state of ion channels (73). Collectively, ion channels regulate the functioning of vital organs such as cardiac muscles (reviewed in (74)) and synaptic transmission across neurons (reviewed in (75)). Recent evidence demonstrates a role for ion channels in cancer progression (Table 1) (76, 77) (reviewed in (56, 78, 79)). Cancer cells hijack ion channels for cellular proliferation, invasion, metastasis, and drug resistance (80–82). Whole genome Pan-cancer analysis demonstrated ion channel gene dysregulation in almost all cancer types with particularly high expression of many ion channel genes seen in most cases (83).

In the context of HGG, in vitro proliferation and invasion assays demonstrate a role for ion channels in HGG progression (reviewed in (13)). For instance, calcium (84–86) and calcium-induced potassium channels are involved in HGG proliferation (87–90), (reviewed in (12)); potassium, chloride (91–93) and calcium-activated intermediate (IK) potassium channels regulates HGG migration (4, 94) and sodium and chloride channels play a key role in HGG drug resistance (31, 95, 96).


Table 1. Role of ion channels in cancer progression.

Cancer type

Ion channel type

Genes involved

Oncogenic phenotype

Reference

Nasopharyngeal

Transient Receptor Potential Cation Channel Subfamily M

TRPM7

Cell migration

(202)

Calcium-sensitive chloride channel

CIC-3

Cell proliferation

(175)

Esophageal squamous cell

voltage-dependent potassium channel (Kv)

hERG1

Proliferation, stem cell growth

(203)

Transient Receptor Potential Cation Channel Subfamily V

TRPV2

Proliferation and drug resistance

(204)

Transient Receptor Potential Cation Channel Subfamily C

TRPC6

Poor patient prognosis

(203)

calcium release-activated calcium channel protein 1

Orai1

Cell proliferation

(205)

Thyroid

Transient Receptor Potential Cation Channel Subfamily C

TRPC1

Cell proliferation and migration

(206)

voltage-dependent potassium channel (Kv)

hERG1

Cell migration

(206)

Breast

 

Transient receptor potential melastatin channel

melastatin channels

cellular invasion

(207)

voltage-sensitive calcium-activated chloride channel

TMEM16A

Cell proliferation

(208)

Transient Receptor Potential Cation Channel Subfamily V

TRPV2

Revert drug resistance

(209)

voltage-gated sodium channel

Nav1.5

Tumor Metastasis

(164)

Potassium voltage-gated channel

Kv10.1 (EAG)

Cell proliferation and migration

(210)

small conductance calcium-activated potassium channel

KCa 2.3

Cell migration

(211)

Renal cell

Transient Receptor Potential Cation Channel Subfamily M

TRPM3

Promotes cell growth

(212)

Transient Receptor Potential Cation Channel Subfamily C

TRPC6

Cell proliferation and aggressiveness

(213)

calcium release-activated calcium channel protein 1

Orai1

Cell proliferation and migration

(214)

small conductance calcium-activated potassium channel

KCa3.1

Highly metastatic and reduced progression-free survival

(215)

Colon

Intracellular chloride channel

CLIC1

Cell invasion and migration

(216)

Transient Receptor Potential Cation Channel Subfamily C

TRPC1, Orai1

Cell migration

(203)

voltage-gated sodium channel

Nav1.5

Poor patient prognosis

(217)

small conductance calcium-activated potassium channel

KCa3.1

Proliferation and invasion

(218)

Ovarian

Intracellular chloride channel

CLIC1

Intraperitoneal metastasis

(219)

Transient Receptor Potential Cation Channel Subfamily C

TRPC3

Cell growth

(220)

voltage-gated sodium channel

Nav1.5

Cell metastasis

(221)

Cervical

voltage-dependent potassium channel (Kv)

hERG1

Cell proliferation

(222)

voltage-gated sodium channel

NaV 1.6

Higher invasive potential

(223)

Melanoma

voltage-dependent potassium channel (Kv)

hERG1

Tumor metastasis

(224)

Bladder

Transient Receptor Potential Cation Channel Subfamily M

TRPM7

Proliferation, recurrence, metastasis and invasion

(221,225)

Prostate

Transient Receptor Potential Cation Channel Subfamily M

TRPM4, 8

Enhanced survival, proliferation, cellular invasion

(207)

Intracellular chloride channel

CLIC1

Cell proliferation and metastasis

(226)

Potassium channel subfamily K member 2

TREK-1

Reduced castration resistance-free survival

(227)

voltage-gated sodium channel

Nav1.7

Tumor metastasis

(228)

Intracellular chloride channel

CLIC1

Cell migration and proliferation

(229)

Transient Receptor Potential Cation Channel Subfamily M

TRPM8

Tumor proliferation and growth

(230,231)

Gastric

voltage-dependent potassium channel (Kv)

hERG1

Survival and cell invasion

(203)

Transient Receptor Potential Cation Channel Subfamily M

TRPM7

Cell proliferation

(184)

Intracellular chloride channel

CLIC1

lymphatic invasion, lymph node metastasis and perineural invasion

(232)

calcium release-activated calcium channel protein 1

Orai 1

Cell metabolism, migration and invasion

(233)

voltage-gated sodium channel

Nav1.7

Cell proliferation and invasion

(234)

voltage-dependent potassium channel (Kv)

hERG1B (Kv11.1)

Reduced survival, metastasis

(235)

Hepatocellular

Transient Receptor Potential Cation Channel Subfamily C

TRPC1

Cell proliferation

(236)

Intracellular chloride channel

CLIC1

Poor prognosis

(237)

small conductance calcium-activated potassium channel

KCa3.1

Cell proliferation and migration

(238)

Glioma

small conductance calcium-activated potassium channel

KCa3.1

Tumor invasion

(239)

Transient Receptor Potential Cation Channel Subfamily C

TRPC6

Hypoxia-induced aggressive tumor and angiogenesis

(240)

Transient Receptor Potential Cation Channel Subfamily C

TRPC1,3,5

Cell proliferation

(241)

Intracellular chloride channel

CLIC1

Reduced patient survival

(180)

voltage-dependent calcium channel L-type, alpha 1C subunit

Cav1.2

Cell Proliferation

(242)

P/Q voltage-dependent calcium channel

Cav2.1

Progression of tumor growth

(216)

Voltage-gated potassium channel

Mitochondrial Kv1.3

Cell survival

(243)

ATP driven potassium channel

Katp

Radio resistance

(244)

 


Additionally, in vitro models demonstrate a distinct role for calcium, potassium, sodium, and chloride ion channels in regulating HGG progression and cellular homeostasis compared to glial cells (reviewed in (97, 98)) (Figure 2). Multiple ion channels coordinate together to balance ionic flux, making it complicated to tease apart their mechanism of oncogenesis. For example, Ca2+-activated K+ (BK) channel co-localize with ClC-3 Cl− channels to initiate brain metastasis (4). However, there is limited pre-clinical evidence to demonstrate the oncogenic potential of ion channels and the anti-tumor efficacy of ion channel drugs in HGG tumors (99,100). On the other hand, ion permeating proteins are a common drug target. A pan-cancer analysis identified ion permeating proteins are highly expressed in a group of cancer samples of HGG (83).


Diagram, schematic

Description automatically generated

Figure 2. Expression and function of ion channels in normal brain cells and glioma cells. The pictorial representation shows the expression of some distinct ion channels and their role in glioma progression. Glioma cells have distinct ion channel expression and localization patterns compared to normal glial cells. The key oncogenic channels in glioma include calcium-activated potassium channel that results in increased intracellular calcium which then facilitates cell invasion and proliferation. Cell proliferation is also facilitated by increased calcium influx through the ErB2 receptors that act as temporary ion transporters. Acid-sensing ion channel 2 (ASIC2) is absent in glioma cells which otherwise would inhibit the Acid-sensing ion channel 1 (ASIC1) in the glial cells to regulate sodium conductance. Chemotactic metastasis and drug resistance are driven by the expression of chloride channels on HGG cell membrane.


Expression and function of ion channels in HGG

The success of translational research depends on its ability to mimic clinical conditions in the research environment. Ion channel abnormalities in HGG patient samples have been demonstrated by genomic and proteomic analyses. Genome-wide analysis of HGG tumor samples reveals that up to 90% of all HGG tumors harbour mutations in ion channel genes (29). These findings are in line with evidence from genome-wide analysis of HGG stem-like cell exon sequencing, which also identified distinct ion channel gene mutations in HGG tumor-derived stem-like cells (101). The Repository of Molecular Brain Neoplasia Data (REMBRANDT) is a comprehensive database that has mapped the gene expression and copy number arrays of HGG alongside clinical phenotypic of HGG tumor samples. A dataset study on REMBRANDT revealed a HGG-specific ion channel gene expression signature (56 genes overexpressed out of a panel of 251 genes) which included genes from a range of different families of ion channels (102). This gene signature was validated in an independent in vitro study using pharmacological knock-down or genetic knock-down mouse models. This study demonstrated that the expression of the identified ion channel gene signature enhanced HGG growth in culture (101). In addition, whole-genome HGG tumor analysis has identified a range of ion channel abnormalities including mutations across different families of ion channels in calcium, sodium, potassium, and chloride channels (101). The below section discusses studies that correlate ion channel gene expression with HGG progression.

Calcium channels

Calcium channels were one of the first identified oncogenic ion channels to play a role in the pathogenesis of HGG (59). In vitro studies demonstrate that an increase in intracellular calcium contributes to an increase in HGG proliferation, and inhibition of the T-Type Calcium channel prevents HGG growth and metastasis (103). Cav3.2 is a gene that codes for the T-type calcium channel and increases intracellular calcium levels by opening T-type calcium channels during membrane depolarization. Zhang and colleagues (104) analysed HGG patient data from both The Cancer Genome Atlas (TCGA) and REMBRANDT databases and identified that 11% of patients either had a mutation, or mRNA upregulation or amplification in the Cav3.2 genes (102,104). Suggesting, potential aberrations within Cav3.2 may have driven the oncogenicity of T-Type calcium channel and contributed to poor patient survival in this cohort of patients (104).

Sodium Channels

Joshi et al used tumor genome sequencing information and correlated the presence of sodium channel mutations with a decreased median survival of HGG patients (30). This finding validates other studies that identify a role for activated sodium channels in HGG tumor progression (101). Acid-sensing ion channels (ASIC) are a type of sodium channel that are voltage insensitive in both neuronal and glial cells and can detect ionic alterations in the extracellular environment (105). ASIC expression maintains HGG cells in a depolarised state, making them susceptible to malignancy (reviewed in (106)).

Co-expression of ASIC1 and ASIC2 suppressed sodium influx in normal brain astrocytes.  However, the sodium influx suppression was absent in HGG tumor samples. mRNA expression analysis of the HGG tumor samples showed the absence of  ASIC2 genes led to the increase in sodium influx (107). Similarly, an independent study by Tian and colleagues identified high expression of ASIC1, and ASIC3, in addition to low expression of ASIC2 in the HGG patient tumor dataset from REMBRANDT (108). Further in vitro testing on HGG stem cells demonstrate that the co-expression of ASIC2 and ASIC3 resulted in the suppression of the oncogenic potential of ASIC3 (108). Nav 1.6 is a sodium channel membrane protein mainly concentrated in the sensory and nervous systems (109). Recent drug screening analyses identified oncogenic potential for Nav 1.6 in HGG cells in vitro and targeting these channels have shown effective reduction in cell proliferation. However, further in vivo analysis is needed to validate the oncogenic role of Nav 1.6 (110). Taken together, these studies suggest that increase in sodium influx could be oncogenic in HGG, making sodium channels an appealing target for therapeutic intervention.

Potassium channels

Evidence in exploring the oncogenic role of potassium channels in neurological cancer are starting to emerge (111). Potassium channels can co-assemble with multiple other channels and form the most complex class of ion channels (112). In normal astrocytes, voltage-gated potassium channels, Kv1.5 and Kv1.3, have been shown to have a role in cell proliferation and growth (113,114). This is demonstrated in a 2017 study, where novel Kv1.3 inhibitors (PAPTP or PCARBTP) induced massive cell death in HGG cells. Another study investigating the expression of these channels in HGG samples demonstrated differential expression of Kv1.5 in HGG according to subtype and malignancy grade, while Kv1.3 did not (113). Interestingly, Arvind and colleagues (115) identified overexpression of Kv1.5 protein, a voltage-gated potassium channel, in low-grade gliomas directly correlated with better patient survival. Thus, Kv1.5 protein expression has been identified as a good prognosis marker for low-grade gliomas.

Another potassium channel, Kir4.1, is specifically located on the cell membrane of astrocytes, attaching to blood vessels and forming synapses. A study found that Kir4.1 expression is decreased in HGG and resulted in increased cell invasion, however, the mechanism by which this occurs has not been elucidated (116). It is hypothesized that Kir4.1 expression may favour the assembly of cytoskeletal proteins in the filopodia and hence, increase cell invasion. Kir4.1 also facilitates cellular differentiation in normal astrocytes. These channels are upregulated in terminally differentiated astrocytes and are downregulated in immature proliferative astrocytes ((117) 113). Therefore, it is suggested that the downregulation of these channels in HGG may contribute to unrestrained growth and proliferation. Kir4.1 facilitates cellular differentiation in normal astrocytes, and is not expressed in proliferative astrocytes as this prevents cells from post-mitotic transition (118). In contrast, Kir4.1 channels in HGG cells are localised on the nuclear membrane and are absent in the cell membrane, which may be indicative of tumor cell proliferation. Ca2+-activated K+ (BK) channels co-localize with ClC-3 Cl− channels for the formation of brain metastasis (4). These studies demonstrate an oncogenic role for the potassium channels that are expressed on the internal membrane of brain cells. 

Chloride channels

Chloride transportation is critical for normal cellular functioning (119). Chloride flux contributes to cell proliferation and cell division via cell volume regulation (120). In HGG, chloride channels have been identified to increase proliferation and drug resistance (96). Increased expression of chloride channels in HGG tumor samples correlated with reduced patient survival (96). Wang and colleagues compared tumors from grades I-IV and identified two chloride channel genes (chloride intracellular channel 1 (CLIC1) and chloride intracellular channel 4 (CLIC4)) to be upregulated in grade IV tumors and in paediatric brain cancer (121). This suggests oncogenic roles for CLIC1 and CLIC4 in aggressive HGG and its contribution to radio-resistance (122). Similarly, in an independent study, upregulation of mRNA transcript levels of CLIC1 in HGG patient tumors correlated with poor patient survival (123). Furthermore, the expression of CLIC1 and CLC3 in HGG tumors correlated with chemotactic metastasis and drug resistance (123).

In vitro mechanistic knockdown studies suggest a role for CLIC3 in HGG proliferation and metastasis. Activation of CLC-3 by the Ca2+/calmodulin-dependent protein kinase II enhanced the HGG cellular condensation during cell division, suggesting a role for CLC-3 in HGG proliferation (124). NF-κB is an oncogenic transcription factor that increases the transcription of matrix metalloproteinases MMP-3 and MMP-9 for cell migration in HGG (125).  CLIC3 interacts with Ca2+/calmodulin-dependent protein kinase II to enhance the oncogenic transcriptional activity of NF-κB for cell migration (126). NKCC1 is a form of cotransporter protein that facilitates active transport of chloride, sodium, and potassium ions. In HGG, NKCC1 accumulates high concentrations of intracellular Cl− which is known to be utilised by HGG cells during invasion (127) suggesting a role for chloride channels and transporters in regulating HGG cell volume for oncogenesis (128).

Miscellaneous channels

Besides the four major ion channels, there are other minor channels including transient receptor potential melastatin (TRPM) (129), transient receptor potential canonical (TRPC), purinergic receptors (PRX) and human ether-a-go-go related gene (hERG). These minor channels have distinct expression patterns in HGG patient cohorts which correlate with patient survival. For example, a recent study concluded that overexpression of TRPM3 and P2RX4 directly correlated with reduced survival of patients with HGG (101). In contrast, Alptekin and colleagues (130) analyzed the survival of 33 patients with HGG and identified that patients who survived longer than 12 months expressed high transcripts of TRP channel genes (TRPC1, TRPC6, TRPM2, TRPM3, TRPM7, TRPM8, TRPV1, and TRPV2). This suggests that the expression of some of the transient channels in patients with HGG may be indicative of a good prognosis (130). Similarly, P2X7R, an ATP-gated cation-permeable receptor, was highly expressed in a subset of HGG patient tumor biopsies. The expression of these receptors were correlated with disease-free overall patient survival, and a positive response to radiotherapy (131). In addition, over-expression of gamma-aminobutyric acid type A receptor subunit gamma (GABRG3) in HGG tumors is suggestive of an increased mean patient survival (101). Overall, these findings suggest that ion channels can also be used as biomarkers for positive prognoses (increased patient survival). Knockdown studies suggest a role for mechanosensitive ion channel Piezo2 in glioma chemo resistance (132). Piezo2 is also identified to be overexpressed in patient fatality caused by peritumoral brain edema (133). Table 2 summarizes the different ion channel families, their roles in HGG, along with potential targeting strategies.


Table 2. Clinically approved ion channel drugs.

Ion channel targeting

Drug

Treatment

Reference

Sodium channel blockers

Tetrodotoxin  (TTX)

Pain relief

(304)

voltage-dependent sodium channel

Lidocaine,

lignocaine and Novocaine

Anaesthetics

(305,306)

voltage-dependent sodium channel

Lidocaine, Flecainide, Propafenone

Antiarrhythmic drugs

(307)

voltage-dependent sodium channel

Phenytoin, fosphenytoin, Lacosamide, Valproate, Zonisamide

Antiepileptic

(308)

voltage-dependent sodium channel

Lacosamide

Anticonvulsant

(309)

Sodium potassium channel blockers

phenytoin, topiramate, lamotrigine and carbamazepine

Antiepileptic

(310–312)

Voltage-gated L-type calcium channel blockers

verapamil, diltiazem, nimodipine, nifedipine and amlodipine

Angina

(313)

Potassium ATP channel agonist

vasodilator, and nicorandil

Angina

(314)

sodium and potassium channels

Amiodarone

Antiarrhythmic

(315,316)

Voltage-gated calcium channel blockers

verapamil, amlodipine and nifedipine

Hyertension

(317)

potassium channel activator

diazoxide

Vasodilation during hypertension

(314)

Regulator of intracellular calcium levels

Nicotinic acid (NA)

Atherosclerosis.

(318)

K ATP channels blocker

sulphonyl urea drugs – glibenclamide

Diabetes

(319)

selective inhibitor of Kv1.3

Margatoxin (MgTX),

Non-small cell lung cancer cell

(320)

antagonist of TRPM8

cannabigerol (CBG)

Colon cancer cells

(321)

VGSC-blocking drug

phenytoin

Breast cancer in vitro

(164)

potassium channel blocker

Oxaliplatin

Anti-proliferative effects on HGG, colorectal cancer in vitro

(322)

Kv channels blocker

PAPTP and PCARBTP

Glioma cells in vitro

(243)

InaP blockers

ranolazine and riluzole

Metastatic breast and prostate cancer

(197,233,23)

Multi-channel blocker, Potassium channel modulator

Hydroquinidine

Currently used to treat short-QT and Brugada arrhythmia syndromes

Significant antiproliferative and pro-apoptotic effect on TMZ-sensitive and -resistant HGG cells

(325)

Sodium channel blocker

Oxcarbazepine

FDA-approved antiepileptic drug

Anti-proliferative and pro-apoptotic in IDH mutant glioma stem cells

(326)

EAG2-Kvβ2 complex blocker

Designer peptide against EAG2-Kvβ2 complex

Anti-proliferative in patient-derived xenograft and syngeneic mouse models

(327)

SK2 channel blocker

P01 scorpion toxin

 

Anti-proliferative in U87 glioma cells

(328)

hERG channel opener and potassium modulator

NS1643 – small molecule inhibitor

Combinations of Pantoprazole with TMZ, retigabine and NS1643  had anti-proliferative in U87 cells

(182)

Potassium channel opener

Retigabine - FDA-approved epilepsy drug

proton-pump inhibitor

Pantoprazole - FDA-approved proton-pump inhibitor

Calcium channel blocker

Flunarizine  - Not FDA-approved. Used to treat migraines

Anti proliferative effect and inhibit invadopodium

(329)

Calcium channel blocker

Econazole nitrate - Anti -fungal drug

Potassium channel blocker

Quinine hydrochloride  - Anti-malarial drug

Potassium channel Kv1.3 blocker

PAPTP/PCARBTP

Anti-proliferative in HGG cells

(243)

T-type/L-type calcium channel blocker

Mibefradil was used to treat hypertension. Currently withdrawn from market

Suppressed HGG growth and stemness

(104)

Potent TRPC antagonist (TRP3-7)

Compound 15g

Anti-proliferative effects in HGG

(254)

hERG antagonist/ligand

Doxazosin - Treatment for benign prostatic hyperplasia

Anti-proliferative effects in HGG

(269)

hERG antagonist/ligand

letrozole - Phase I clinical trial

Anti-proliferative effects in HGG

(270)

AMPAR (calcium permeable channels)

Fluoxetine - FDA approved antidepressant

Anti-proliferative effects in HGG

(238)

Sodium/potassium/chloride co-transporter isoform 1 (NKCC1)

STS66 Bumetamide derivative

Anti-proliferative, reduced cell growth in HGG

(330)

TRPV4 antagonist

Cannabidiol

Anti-proliferative in HGG

(331)

 


Collectively, there is evidence demonstrating ion channel aberrations in HGG tumors. Some ion channels play an oncogenic role while others may be tumor suppressive and each of the ion channel family members can contribute to a particular hallmark of cancer. For example, inhibiting sodium channels can prevent tumor cell invasion while inhibiting calcium channels may trigger an anti-proliferative effect. However, ion channel aberrations have only been retrospectively correlated with the survival of patients with HGG and an in-depth mechanistic understanding of their oncogenic roles in HGG is lacking. The current research gap between pre-clinical testing and the clinical understanding of HGG tumors has limited the effective translation of research findings for HGG treatment and will be discussed below.

Overview of ion channels therapeutics in cancer

Every cell has ion channels and ion transporters along their plasma membrane to transport ions across the cells. Each type of cell possesses specific ion selectivity and permeability making their ionic composition different from one another and from their microenvironment (134). This ionic difference creates a membrane potential (Vm) to the cell membrane (135). The change in Vm in turn regulates the movements and function of the cellular proteins to drive cellular activities such as proliferation, invasion and differentiation. The ionic exchange is necessary to regulate cellular volume, a critical factor in cell cycle and cell invasion (136). Cardiovascular, anaesthetic and many psychiatric drugs target ion channels (74,137–139). A faster approach to translational medicine is to repurpose drugs. Drug repurposing is a process that has been largely used for neurological disorders. Ion channel drugs have been used to treat various neurological disorders at different treatment regimens (140). The recent developments in high-throughput assays and technologies such as flux-based assays, fluorescence-based assays and automated electrophysiological assays have drastically changed the outlook of using ion channel drugs to treat “channelopathies” such as Bartter Syndrome, seizures and cystic fibrosis through ion-channel-specific assays (47,141,141–146). There are many clinically approved ion channel drugs to treat neurological disorders (147), neuronal damage (148), cardiovascular damage, cardiac disease, hypertension, diabetes, epilepsy, spinocerebellar ataxia type-13, infectious diseases (149), and brain defects (150). These drugs can effectively cross the blood-brain barrier and the majority are classified as anticonvulsant drugs or calcium and sodium channel blockers (33,151). As a result, ion channel drugs are being tested to treat a range of diseases, disorders and cancers, including HGG (Table 3).


Table 3. Role of ion channels in HGG and potential targeting strategies.

Ion channel name

Ion channel type

Genes involved

Oncogenic Phenotype

Potential targeting strategy

Reference

Cav3.1

Voltage-gated calcium channels T-type, alpha 1G subunit

CACNA1G

Increased proliferation and regulation in autophagy

Cardiac arrhythmia, epilepsy, hypertension drugs can be used

(245)

Cav3.2

Voltage-gated calcium channels T-type, alpha 1G subunit

CACNA1H

Increased proliferation through AKT/mTOR pathways

Pain, and epilepsy drugs can be tested

(104,246,247)

Cav2.1

Voltage-dependent P/Q type

CACNA1A

Increased proliferation

Epilepsy, and migraine drugs can be used to target

(241)

Cav1.2

Voltage-dependent L-type

CACNA1C

Increased proliferation

Cardiovascular drugs

(248)

TRPC1, 5

Transient receptor potential cation channel, subfamily M, member 1 and 5

TRPC1, TRPC5

Increased proliferation through regulation of calcium signalling and increased migration

Riluzole TRPC5 agonist approved drug for the treatment of amyotrophic lateral sclerosis can be used in HGG

(249–251)

TRPM8

Transient receptor potential cation channel, subfamily M, member 8

TRPM8

Increased proliferation, and migration mediated by BK activation

Anti‐inflammatory drugs targeting TRPM8 can be used to target HGG

(252,253)

TRPM7

Transient receptor potential cation channel, subfamily M, member 7

TRPM7

Increased, proliferation, migration and invasion

(253–255)

TRPC6

Transient receptor potential cation channel subfamily C member 6

TRPC6

Increased proliferation, regulation of cell cycle, hypoxic migration

(203,256)

CLIC1

Chloride intracellular channel protein 1

CLIC1

Increased proliferation

Biotin conjugated analog of MTI-101 can target CLIC1 to reduce HGG proliferation

(180,257)

CLC3

Chloride voltage-gated channel 3

CLCN3

Increased migration and invasion

Inducible gene deletion strategies can help target CLCN3 to reduce HGG migration and invasion

(124,126,258)

TRPML2

Mucolipin-2 transient receptor potential cation channel

MCOLN2

Increased proliferation and decreased apoptosis

TRP channel inhibitors such as Capsaicin, SKF-96365 and 2-APB can target MCOLN2 in HGG

(259–262)

TRPV4

Transient receptor potential vanilloid 4

TRPV4

Increased migration and invasion, cytoskeletal remodelling

Orally active pulmonary edema drug targeting TRPV4 can reduce HGG migration

(263–265)

Kv1.3, Kv1.5

Voltage-gated potassium channel

KCNA3, KCNA5

Increased proliferation

Kv1.3 specific toxins such as charybdotoxin and margatoxin can be used to prevent HGG proliferation

(266–268)

Kv2.1

Voltage-gated potassium channel

KCNB1

Increased proliferation, regulation of autophagy

Antiarrhythmic drugs such as Quinidine, 4-Aminopyridine (4-AP) and potassium channel blocker,  Tetraethylammonium, are specific blockers of Kv2.1

(70,113,126)

Kv11.1 (hERG)

Ether-a-go-go potassium channel

KCNH2

Increased proliferation

Dofetilide an anti-arrhythmic drug and E-4031 a well known hERG channel blocker may reduce proliferation in HGG

(224,269–271)

VRAC

Volume-regulated anion channel

LRRC8A

Hypoxia-related proliferation and survival

Small molecule inhibitors such as DCPIB or LRRC8A specific antibodies are good therapeutic opportunities for HGG

(27,272–274)

Nav1.6

Voltage-gated sodium channel

SCN8A

Increased proliferation, migration and invasion

Small molecules like PF-06372865 and GS967 have been shown to selectively inhibit SCN8A.

(110,275)

Nav1.5

Voltage-gated sodium channel

SCN5A

Increased invasion with NHE1

Multiple antiarrhythmic and antianginal drugs such as Amiodarone, Ranolazine,  Quinidine, Mexiletine, Lidocaine, Flecainide can target SCN5A in HGG

(51,276,277)

AMPAR

Calcium permeable channels

GLUA1 and GLUA4

Increased proliferation and invasion

Selective GLUA1 inhibitor - LY3130481 and GLUA4 inhibitor such as CX614 and Cyclothiazide

(238,278–280)

ANO1, TMEM16A

Anoctamin 1 voltage-gated calcium-activated anion channel

TMEM16A

Increased proliferation, supports maintenance of stemness in GSCs

Anoctamin 1 inhibitors such as T16Ainh-A01 and CaCCinh-A01 can reduce GSC stemness

(281–283)

ASIC1 and ENaC

Acid-sensing ion channel 1a

ASIC1

Increased migration and lamellipodium expansion

Amiloride is a well-known diuretic that blocks epithelial sodium channels

(284–286)

Kir4.1

ATP-sensitive inward rectifier potassium channel 10

KCNJ10

Decreased proliferation

Baicalein is a flavonoid compound that effectively inhibit KCNJ10 can be used to treat HGG

(118,287,288)

KATP, Kir6.2

ATP-sensitive potassium channels

KCNJ11

Increased proliferation

Glyburide, a KATP channels blocker currently in the clinic to treat neonatal diabetes caused by KCNJ11 mutations can be used to reduce HGG proliferation

(289–291)

KCa1.1, BK

Calcium-activated potassium channel subunit alpha-1

KCNMA1

Increased proliferation and migration

Clinically available epileptic drug such as Paxilline blocks KCNMA1

(292–294)

KCa3.1

Intermediate-conductance calcium-activated potassium channel

KCNN4

Increased migration and invasion

Senicapoc (ICA-17043) is a KCa3.1 channel blocker, that reduces hemolysis and improving red blood cell survival in patients with sickle cell anemia. This drug may reduce HGG migration by blocking KCa3.1 with minimal side effects

 

(97,292,295)

PIEZO1

Mechanosensitive  non-specific cation channel

PIEZO1

Increased proliferation, regulation of tissue stiffness and aggression

There are no clinically approved drugs to treat PIEZO1 channel. GsMTx4 is a peptide toxin derived from tarantula venom and is potent to inhibit PIEZO1.

(296–298)

NHE5

Na+/H+ exchanger 5

SLC9A5

increased proliferation, metastasis, cell adhesion and invasion

There are no selective inhibitors for NHE5. NHE1 inhibitors such as Amiloride and its derivatives can be used to reduce HGG growth and metastasis.

(57,299)

NHE9

Endosomal pH regulator

SLC9A9

Increased proliferation, stemness

There are no selective inhibitors for NHE5. NHE1 inhibitors such as Amiloride and its derivatives can be used to reduce HGG growth and metastasis.

(57,300,301)

NKCC1

Na+K+/Cl- cotransporter isoform 1

SLC12A2

Increased proliferation, migration, invasion

Bumetanide and Furosemide are loop diuretic that inhibits NKCC1 and is used primarily to treat edema and hypertension. This can be tested on HGG for anti-cancer effects.

(302,303)

 


Electrophysiological analysis of cancer cells demonstrate depolarization of the cancer cell membrane which contributes to the stemness of cancer cells and attribute to the cellular proliferation, invasion and migration (109, 135, 152). Studies using hepatoma and adenocarcinoma demonstrates increased intracellular sodium concentration compared to normal cells, suggesting a role for cancer cell depolarisation (153, 154). More recently, ion channels have been identified as a target for cancer therapy with a few reviews summarising their role as a therapeutic target for cancer (55,155,156). Ion channel drugs modulate ion channels by either blocking or opening them, altering the ionic concentration and in turn, reducing the function of oncogenic proteins across cancer cells (157,158). However, the drugs may have different modes of action in different types of cancer due to their diverse oncogenic function and their complex interactions between other ion channels and oncogenes (159,160). Once the altered channels are identified in any given cancer, treating the alteration using modulators will recalibrate the ionic balance and alter the Vm to reduce the oncogenic potential (161). Some of these drugs have been used in pre-clinical models to demonstrate their anti-cancer effects (162–164). Based on drug repository screening in silico, a study screened over 100 United States Food and Drug Administration (FDA) approved drugs against small cell lung cancer (SCLC) (165). Of the top 100 drugs identified, many were ion channel modulators such as imipramine, promethazine and verapamil, that significantly inhibited the growth of SCLC both as a single agent (165) and in combination (166). However, these drugs were specific to SCLC and did not add any survival benefit in a phase III study in patients with multidrug-resistant multiple myeloma (167). This suggests that ion channels may interact with other pathways specific to the cellular context and hence, studying ion channel drugs in the presence of other oncogenic drivers may be a greater determinant of drug efficacy. Further, the clinical availability of multiple ion channel drugs means a shorter translation time and a higher rate of clinical success, underlining the need for additional research to identify roles of ion channels in HGG tumors. However, there is limited literature exploring the opportunity of repurposing ion channel drugs for cancer therapy (36).

Therapeutic targeting of ion channels in HGG

HGGs are commonly treated with alkylating agents (such as temozolomide) (168,169) and ionizing radiation (168,170) that causes DNA damage and induce apoptotic cell death (168). However, HGG tumors develop resistance to standard treatments due to pathway rewiring, resulting in mortality. Chemo- and radiotherapy-induced DNA damage triggers a series of tumor-cell survival mechanisms including alterations in calcium-activated potassium channels and calcium-permeable non-selective cation channels (95). Ion channels are active in excitable cells such as neurons and possess cell-specific functions (133). In vitro analyses demonstrate that activated ion channels induce both intrinsic and acquired resistance to radiotherapy in HGG stem-like cells (95) (171). Current combination treatment with alkylating agents and ionising agents are ineffective and additional research is required to urgently identify alternate therapies (172). Apart from conventional drug treatments, the use of alternative electric fields known as Tumor Treating Fields has been recently used to treat paediatric HGG by targeting ion channels (173). Currently, this technique is being used in combination with ion channel blockers to induce cell death, demonstrating oncogenic role for ion channels in HGG (122). Based on the functions of ion channels in brain cells, it is proposed that targeting aquaporin (water channels) together with ion channels can limit HGG invasiveness, however this is yet to be tested in vivo (174).

Ion channel drugs as single-agent therapy in HGG

CLC-3, a voltage-gated chloride channel, is present in the plasma and intracellular membrane of HGG cells (175) and is predicted to drive drug resistance (96). Chlorotoxin (Cltx) preferentially inhibits HGG cell invasion by binding to CLC-3/matrix metallopeptidase 2 (MMP2) membrane complex and inhibits the enzymatic activity of MMP2 to reduce the cell surface expression levels of MMP2 to inhibit cell invasion. Cltx has recently entered phase I and II clinical trials for patients with HGG (176). Similarly, I-TM-601, a synthetically labelled Cltx, has also entered phase I and II clinical trials for patients with HGG tumors (177). Besides its therapeutic potential, radiolabelled I-TM-601 has also been used as a diagnostic marker in identifying tumor burden in patients with HGG. Radiolabelled I-TM-601 can be effectively detected using whole-brain single-photon emission computed tomography scans (178) and immunohistochemistry of brain tissue (179). Another study tested the efficacy of a therapeutic antibody against CLIC1 in a pre-clinical study using HGG derived progenitor cells where the antibody significantly increased survival in mouse models (180).

Ion channel drugs as part of combination therapy in HGG

Multiple independent oncogenic pathways combine to trigger the hallmarks of cancer. Combination therapy is, therefore, the gold standard for cancer treatment (181). Ion channel drugs that failed as a single-agent treatment have been shown to increase survival benefits when used as combination therapy in HGG clinical trials (182). Amiodarone, an anti-arrhythmic drug, targets multiple ion channels and increases the risk of certain types of cancers when administered for neurological disorders (183). However, in combination with a therapeutic recombinant human protein, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), amiodarone impeded HGG growth in vitro through the induction of calcium influx associated apoptosis (184). This apoptotic effect was selectively in HGG cells and did not affect normal astrocytes, highlighting the potential utility of amiodarone as part of combination therapy for patients with HGG with minimal side effects. This evidence together demonstrates that the pharmacodynamics of ion channel drugs could be completely different in a cancer setting as opposed to neurological disorders. As a result, ion channel drugs should be treated as novel compounds for cancer treatment and stresses the importance of rigorous pre-clinical and clinical testing. Bepridil and cibenzoline are two calcium channel drugs that were previously used only as anti-arrhythmic agents. These two drugs impeded HGG cell growth as part of combination therapy with TRAIL (185). The dual calcium and G-protein coupled receptor inhibitor, pimozide, has been shown to reverse resistance to the chemotherapeutic drug, temozolomide and sensitize HGG cells to radiation treatment (186). Furthermore, nifedipine, a calcium channel blocker, sensitized resistant HGG cells to cisplatin and inhibited tumor growth both in vitro and in pre-clinical mouse models (187). In vitro and in vivo studies demonstrate that mibefradil, a nonspecific calcium channel blocker sensitizes HGG stem-like cells (GSC) to temozolomide by inhibiting the AKT/mTOR pathways and reversing stemness. Mibefradil sensitized HGG to temozolomide and stimulated apoptotic proteins such as survivin and BAX (103). Similarly, Krouse and colleagues repurposed mibefradil to sensitize HGG tumors to conventional chemotherapy in a phase 1b clinical trial (188). The results from the clinical trial demonstrated a positive outcome in patients with HGG (188–190). However, despite the positive outcome in patients with HGG, the trial could not progress further as mibefradil was withdrawn from the market following multiple reports of drug-drug interactions (191).

Although these studies are preliminary and not all drugs have been tested in pre-clinical animal models, the findings highlight the potential in repurposing ion channel drugs as a treatment for patients with HGG. However, to date, studies investigating the role of ion channels in HGG survival have been associational, not mechanistic and further studies are necessary to identify the mechanistic role for ion channels as a potential therapeutic target for HGG. Pre-clinical testing of ion channel drugs on clinically relevant HGG models will expedite the clinical translation of FDA-approved ion channel drugs as both single and in combination with conventional therapies, in both adult and paediatric patients with HGGs, which may otherwise cause severe neurological side effects (192).

Research gaps in repurposing ion channel drugs for cancer therapeutics – preclinical model development

Historically, limitations of both in vivo animal studies and 2D in vitro models have delayed progress in this field (193). 2D immortalized monolayer cell culture have been used due to their cost effectiveness and reproducibility. However, they do not accurately reflect the tumor environment, the highly heterogeneous nature of HGG nature. Since the announcement by the United States Food and Drug Administration in 2022 to abolish the mandate to test on animals, (through the FDA Modernization Act 2.0) in vitro methods and models are rapidly advancing in this area (193–195). Non-animal models include 2D cell culture, 3D spheres (or neurospheres), organoids, bio printing, tissue-slice cultures and tumor-on-chip methods, all which have advantages and disadvantages (194,195). To account for the highly heterogeneous nature of HGG and for in vitro studies to be reflective of clinical outcomes, it is important to use patient-derived cell lines. Development of 3D in vitro models that can incorporate vasculature and immune cell components will advance this field (194). Tumor-on-chip is both dynamic and can incorporate vasculature by joining multiple organ (organ-on-chip) systems. Alternatively, culturing 3D spheroids in a chip environment provides a more dynamic and physiologically accurate model, which has already been used to test certain FDA approved drugs for repurposing (196). More complex models have been developed since, integrating biosensor enhanced on-chip models (197). Complex scaffolding models utilising biomaterials have been developed to overcome some of the limitations of current models (195). Bio-banking will be essential to advance these preclinical models. The use of preclinical models using patient-derived cell lines can help investigate the mechanism of action and thereby, improve the translation of research findings. Further improvements must focus on the accurate recapitulation of the tumor microenvironment (including electrical stimulation) and the range and plasticity of cellular states of HGG among others (198). Combining the knowledge gleaned from computational simulation of ion channel biology can further improve in vitro models, particularly for tissue engineering and microfluidic (tumor-on-chip) approaches (193,198). It is evident that ion channels play an oncogenic role in HGG and additional research in clinically relevant models is required for the development of effective treatments.

Future directions

Compelling evidence on the prevalence of ion channel aberrations in patients with HGG is starting to accumulate. However, while ion channels are emerging as promising oncogenic targets across many different cancers, very few ion channel inhibitors have been tested in patients with HGG (177,199).

Clinical failure is largely due to an oversight on the interaction of ion channels with other oncogenes such as tyrosine kinases and other ion channel family members. However, these complex interactions may be key contributors to the cellular and pharmacological response to ion channel drugs. For example, the same ion channel drug may induce completely different outcomes in a patient with a kinase mutation as compared to a patient with a transcription factor mutation. Thus, ion channels need to be studied under the influence of other oncogenic genes in patients with HGG and administered as personalized medicine based on the tumor pathology. Additionally, ion channels compensate for each other’s function (52). This may explain why some drugs can effectively impede cancer growth while others do not. Hence, it is critical to study ionic alterations as an orchestra of multiple cellular ion channels that maintains an electric gradient on the cell membrane to regulate cellular processes. To achieve this, appropriate in vitro models are necessary (194,200).  Testing ion channel drugs in patient derived HGG neurospheres (201) recapitulates the clinical phenotype and hence, will significantly increase the success of ion channel drugs in the clinic. Patient-derived cell line mouse models will help us understand the drug’s mechanism of action in the presence of the tumor microenvironment to bridge the knowledge gap between the clinic and laboratory research of HGG. Personalized approaches are essential for treating HGG due to their significant heterogeneity. These tumors exhibit vast genetic, epigenetic, and phenotypic diversity between both patients and within the same tumor. This variability impacts treatment responses and outcomes, making standardized therapies less effective. By tailoring personalized treatment strategies to the unique characteristics of each patient's tumor, we can improve the efficacy and outcomes of patients with HGG. With many ion channel drugs on the shelf, molecular profiling of tumor biopsy samples to identify ion channel aberrations, can address the complex nature of HGG more effectively with ion channel drugs.

It is important to understand the level of deviation between the effects of ion channel drugs in vitro and in vivo compared to the effects they will have in clinic. Furthermore, it is necessary to undertake studies in clinically relevant models through multi-omic approaches to better understand the distinct oncogenic roles of ion channels in HGG. These studies will enrich our knowledge on the molecular mechanism of ion channel drugs on HGG tumors and help us understand drug induced resistance. These findings will facilitate identifying a targeted therapy to treat patients with HGG with minimal side effects.

Conclusion

The highly plastic, diffusive, and heterogeneous nature of HGG tumors results in very low patient survival, which has been improved only minimally with current therapeutic options due to the development of drug resistance. The current landscape of HGG treatment is characterized by a combination of surgery, radiotherapy, and chemotherapy, yet these approaches often fail to extend the patient survival. Ion channels have been identified as a promising target for HGG therapeutics, due to strong evidence of ion channel pathologies (abnormalities/mutations) in HGG proliferation, and drug resistance. The distinct patterns of ion channel expression observed in HGG compared to normal brain tissues suggest that these channels play a crucial role in HGG progression. Notably, the upregulation of certain ion channels, such as those involved in calcium, potassium, and chloride transport, underscores their potential as biomarkers for HGG prognosis and as targets for therapeutic intervention. The same ion channels identified as therapeutic targets could also be used as biomarkers for disease or disease progression. Ion channel drugs have been repurposed for a host of neurological conditions, and although these drugs cross the blood-brain barrier, only a few have been tested for their anti-cancer effects. In addition, their complex mode of action is not well understood. There is growing evidence of the potential to use ion channel drugs for cancer therapy, particularly in combination with conventional therapy, but differences in the pharmacodynamics of ion channel drugs need further mechanistic investigation. The integration of ion channel-targeting drugs into clinical protocols offers a promising avenue to enhance therapeutic efficacy. Preclinical studies have already demonstrated the potential of ion channel modulators in reducing HGG growth and sensitizing tumors to conventional treatments. For instance, drugs targeting specific potassium and chloride channels have shown encouraging results in pre-clinical models by impairing HGG growth and inducing apoptosis.

Future research should focus on elucidating the precise roles of various ion channels in HGG pathophysiology and identifying ion channel-targeting compounds for clinical use. Personalized medicine approaches, leveraging the unique ion channel expression profiles of individual tumors, could further refine treatment strategies, ensuring maximal therapeutic benefit while minimizing adverse effects. New 3D in vitro models using patient-derived cells, tissue engineering and microfluidic approaches are improving the accurate recapitulation of the tumor environment and the range and plasticity of cellular states of HGG, showing promise to elucidate oncogenic mechanisms of HGG. Targeting ion channels represents a novel and promising therapeutic strategy in the fight against HGG, with the potential to significantly improve patient outcomes and advance the current standard of care. Clinically relevant and physiologically accurate models will improve testing of ion channel drugs, offering a personalized medicine approach that can be combined with multi-omics to improve our understanding of HGG, HGG drug resistance and create enhanced therapeutics with minimal side effects. 

Acknowledgment

We thank the Pirate Ship Foundation, The Cure starts Now, Perth Children’s hospital Foundation, The Brain Tumor Charity, and the Robert Connor Dawes Foundation for supporting the research work related to the review. A special thanks to senior postdoctoral researchers within the institute who mentored us in completing this review.

Author contribution

PJM conceptualizing, drafting, editing and reference collection. NA conceptualizing and overall editing, CR drafting and editing.

Conflict of interest

The authors report no conflict of interest.

Funding

There is no funding agency involved in this research.

References

 

1.     Jäkel S, Dimou L. Glial Cells and Their Function in the Adult Brain: A Journey through the History of Their Ablation. Front Cell Neurosci. 2017;11:24–24.

2.     Zong H, Verhaak RGW, Canoll P. The cellular origin for malignant glioma and prospects for clinical advancements. Expert Rev Mol Diagn. 2012 May;12(4):383–94.

3.     Forsyth PA, Wong H, Laing TD, Rewcastle NB, Morris DG, Muzik H, et al. Gelatinase-A (MMP-2), gelatinase-B (MMP-9) and membrane type matrix metalloproteinase-1 (MT1-MMP) are involved in different aspects of the pathophysiology of malignant gliomas. Br J Cancer. 1999 Apr;79(11–12):1828–35.

4.     Turner KL, Sontheimer H. Cl- and K+ channels and their role in primary brain tumour biology. Philos Trans R Soc Lond B Biol Sci. 2014 Mar;369(1638):20130095–20130095.

5.     Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol (Berl). 2016 Jun;131(6):803–20.

6.     Valk PE, Dillon WP. Radiation injury of the brain. AJNR Am J Neuroradiol. 1991 Jan;12(1):45–62.

7.     Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJB, Janzer RC, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 2009 May;10(5):459–66.

8.     Pillay V, Allaf L, Wilding AL, Donoghue JF, Court NW, Greenall SA, et al. The plasticity of oncogene addiction: implications for targeted therapies directed to receptor tyrosine kinases. Neoplasia N Y N. 2009 May;11(5):448–58, 2 p following 458.

9.     Mateos-Aparicio P, Rodríguez-Moreno A. The Impact of Studying Brain Plasticity. Front Cell Neurosci. 2019;13:66–66.

10.   Abou-Antoun TJ, Hale JS, Lathia JD, Dombrowski SM. Brain Cancer Stem Cells in Adults and Children: Cell Biology and Therapeutic Implications. Neurother J Am Soc Exp Neurother. 2017 Apr;14(2):372–84.

11.   Simon OJ, Müntefering T, Grauer OM, Meuth SG. The role of ion channels in malignant brain tumors. J Neurooncol. 2015 Nov;125(2):225–35.

12.   Molenaar RJ. Ion channels in glioblastoma. ISRN Neurol. 2011;2011:590249–590249.

13.   Griffin M, Khan R, Basu S, Smith S. Ion Channels as Therapeutic Targets in High Grade Gliomas. Cancers. 2020 Oct;12(10).

14.   Kirmiz M, Palacio S, Thapa P, King AN, Sack JT, Trimmer JS. Remodeling neuronal ER-PM junctions is a conserved nonconducting function of Kv2 plasma membrane ion channels. Mol Biol Cell. 2018 Oct;29(20):2410–32.

15.   Chittajallu R, Chen Y, Wang H, Yuan X, Ghiani CA, Heckman T, et al. Regulation of Kv1 subunit expression in oligodendrocyte progenitor cells and their role in G1/S phase progression of the cell cycle. Proc Natl Acad Sci U S A. 2002 Feb;99(4):2350–5.

16.   Ghiani CA, Yuan X, Eisen AM, Knutson PL, DePinho RA, McBain CJ, et al. Voltage-activated K+ channels and membrane depolarization regulate accumulation of the cyclin-dependent kinase inhibitors p27(Kip1) and p21(CIP1) in glial progenitor cells. J Neurosci Off J Soc Neurosci. 1999 Jul;19(13):5380–92.

17.   Becchetti A. Ion channels and transporters in cancer. 1. Ion channels and cell proliferation in cancer. Am J Physiol Cell Physiol. 2011 Aug;301(2):C255-65.

18.   Bortner CD, Hughes FM, Cidlowski JA. A primary role for K+ and Na+ efflux in the activation of apoptosis. J Biol Chem. 1997 Dec;272(51):32436–42.

19.   Daoudal G, Debanne D. Long-term plasticity of intrinsic excitability: learning rules and mechanisms. Learn Mem Cold Spring Harb N. 2003;10(6):456–65.

20.   Naoki H, Sakumura Y, Ishii S. Local signaling with molecular diffusion as a decoder of Ca2+ signals in synaptic plasticity. Mol Syst Biol. 2005;1:2005.0027-2005.0027.

21.   Dityatev A, Rusakov DA. Molecular signals of plasticity at the tetrapartite synapse. Curr Opin Neurobiol. 2011 Apr;21(2):353–9.

22.   Zhao X, Han C, Zeng Z, Liu L, Wang H, Xu J, et al. Glutamate Attenuates the Survival Property of IGFR through NR2B Containing N-Methyl-D-aspartate Receptors in Cortical Neurons. Oxid Med Cell Longev. 2020;2020:5173184–5173184.

23.   Ryland KE, Hawkins AG, Weisenberger DJ, Punj V, Borinstein SC, Laird PW, et al. Promoter Methylation Analysis Reveals That KCNA5 Ion Channel Silencing Supports Ewing Sarcoma Cell Proliferation. Mol Cancer Res MCR. 2016 Jan;14(1):26–34.

24.   Liu ZB, Liu C, Zeng B, Huang LP, Yao LH. Modulation Effects of Cordycepin on Voltage-Gated Sodium Channels in Rat Hippocampal CA1 Pyramidal Neurons in the Presence/Absence of Oxygen. Neural Plast. 2017;2017:2459053–2459053.

25.   HODGKIN AL, HUXLEY AF. A quantitative description of membrane current and its application to conduction and excitation in nerve. J Physiol. 1952 Aug;117(4):500–44.

26.   Catacuzzeno L, Sforna L, Esposito V, Limatola C, Franciolini F. Ion Channels in Glioma Malignancy. Rev Physiol Biochem Pharmacol. 2021;181:223–67.

27.   Liu J, Qu C, Han C, Chen MM, An LJ, Zou W. Potassium channels and their role in glioma: A mini review. Mol Membr Biol. 2019 Dec;35(1):76–85.

28.   Varricchio A, Ramesh SA, Yool AJ. Novel Ion Channel Targets and Drug Delivery Tools for Controlling Glioblastoma Cell Invasiveness. Int J Mol Sci. 2021 Nov;22(21).

29.   Parsons DW, Jones S, Zhang X, Lin JCH, Leary RJ, Angenendt P, et al. An integrated genomic analysis of human glioblastoma multiforme. Science. 2008 Sep;321(5897):1807–12.

30.   Joshi AD, Parsons DW, Velculescu VE, Riggins GJ. Sodium ion channel mutations in glioblastoma patients correlate with shorter survival. Mol Cancer. 2011 Feb;10:17–17.

31.   Sheykhzadeh S, Luo M, Peng B, White J, Abdalla Y, Tang T, et al. Transferrin-targeted porous silicon nanoparticles reduce glioblastoma cell migration across tight extracellular space. Sci Rep. 2020 Feb;10(1):2320–2320.

32.   Olsen ML, Weaver AK, Ritch PS, Sontheimer H. Modulation of glioma BK channels via erbB2. J Neurosci Res. 2005 Jul;81(2):179–89.

33.   Abbott NJ, Khan EU, Rollinson CMS, Reichel A, Janigro D, Dombrowski SM, et al. Drug resistance in epilepsy: the role of the blood-brain barrier. Novartis Found Symp. 2002;243:38–5.

34.   Elias AF, Lin BC, Piggott BJ. Ion Channels in Gliomas-From Molecular Basis to Treatment. Int J Mol Sci. 2023 Jan;24(3).

35.   Keluth S, Jitte S, Bhushan R, Ranjan OP, Murti K, Ravichandiran V, et al. Targeting Ion Channels for the Treatment of Glioma. Mini-Rev Med Chem. 2023 Jul;23(12):1298–318.

36.   Kirtonia A, Gala K, Fernandes SG, Pandya G, Pandey AK, Sethi G, et al. Repurposing of drugs: An attractive pharmacological strategy for cancer therapeutics. Semin Cancer Biol. 2021 Jan;68:258–78.

37.   Gadsby DC. Ion channels versus ion pumps: the principal difference, in principle. Nat Rev Mol Cell Biol. 2009 May;10(5):344–52.

38.   Anderson KJ, Cormier RT, Scott PM. Role of ion channels in gastrointestinal cancer. World J Gastroenterol. 2019 Oct;25(38):5732–72.

39.   Green WN. Ion channel assembly: creating structures that function. J Gen Physiol. 1999 Feb;113(2):163–70.

40.   Unwin N. The structure of ion channels in membranes of excitable cells. Neuron. 1989 Dec;3(6):665–76.

41.   Isacoff EY, Jan LY, Minor DL. Conduits of life’s spark: a perspective on ion channel research since the birth of neuron. Neuron. 2013 Oct;80(3):658–74.

42.   Kulbacka J, Choromańska A, Rossowska J, Weżgowiec J, Saczko J, Rols MP. Cell Membrane Transport Mechanisms: Ion Channels and Electrical Properties of Cell Membranes. Adv Anat Embryol Cell Biol. 2017;227:39–58.

43.   Fohlmeister JF. Voltage gating by molecular subunits of Na+ and K+ ion channels: higher-dimensional cubic kinetics, rate constants, and temperature. J Neurophysiol. 2015 Jun;113(10):3759–77.

44.   Abdul Kadir L, Stacey M, Barrett-Jolley R. Emerging Roles of the Membrane Potential: Action Beyond the Action Potential. Front Physiol. 2018;9:1661–1661.

45.   Shah S, Chu Y, Cegielski V, Chu XP. Acid-Sensing Ion Channel 1 Contributes to Weak Acid-Induced Migration of Human Malignant Glioma Cells. Front Physiol. 2021;12:734418–734418.

46.   Armstrong CM. Na channel inactivation from open and closed states. Proc Natl Acad Sci. 2006 Nov;103(47):17991–6.

47.   Waszkielewicz AM, Gunia A, Szkaradek N, Słoczyńska K, Krupińska S, Marona H. Ion channels as drug targets in central nervous system disorders. Curr Med Chem. 2013;20(10):1241–85.

48.   Nelson MT, Todorovic SM, Perez-Reyes E. The role of T-type calcium channels in epilepsy and pain. Curr Pharm Des. 2006;12(18):2189–97.

49.   Smart TG, Paoletti P. Synaptic neurotransmitter-gated receptors. Cold Spring Harb Perspect Biol. 2012 Mar;4(3).

50.   Cheng KT, Ong HL, Liu X, Ambudkar IS. Contribution and regulation of TRPC channels in store-operated Ca2+ entry. Curr Top Membr. 2013;71:149–79.

51.   Catterall WA. Voltage-gated calcium channels. Cold Spring Harb Perspect Biol. 2011 Aug;3(8):a003947–a003947.

52.   Rosati B, McKinnon D. Regulation of ion channel expression. Circ Res. 2004 Apr;94(7):874–83.

53.   Soriani O, Rapetti-Mauss R. Sigma 1 Receptor and Ion Channel Dynamics in Cancer. Adv Exp Med Biol. 2017;964:63–77.

54.   Haworth AS, Brackenbury WJ. Emerging roles for multifunctional ion channel auxiliary subunits in cancer. Cell Calcium. 2019 Jun;80:125–40.

55.   Capatina AL, Lagos D, Brackenbury WJ. Targeting Ion Channels for Cancer Treatment: Current Progress and Future Challenges. Rev Physiol Biochem Pharmacol. 2022;183:1–43.

56.   Litan A, Langhans SA. Cancer as a channelopathy: ion channels and pumps in tumor development and progression. Front Cell Neurosci. 2015;9:86–86.

57.   Pedersen SF, Stock C. Ion channels and transporters in cancer: pathophysiology, regulation, and clinical potential. Cancer Res. 2013 Mar;73(6):1658–61.

58.   Farfariello V, Prevarskaya N, Gkika D. Ion Channel Profiling in Prostate Cancer: Toward Cell Population-Specific Screening. Rev Physiol Biochem Pharmacol. 2021;181:39–56.

59.   Prevarskaya N, Skryma R, Shuba Y. Ion Channels in Cancer: Are Cancer Hallmarks Oncochannelopathies? Physiol Rev. 2018 Apr;98(2):559–621.

60.   Shuttleworth TJ. Intracellular Ca2+ signalling in secretory cells. J Exp Biol. 1997 Jan;200(Pt 2):303–14.

61.   Rizzuto R, De Stefani D, Raffaello A, Mammucari C. Mitochondria as sensors and regulators of calcium signalling. Nat Rev Mol Cell Biol. 2012 Sep;13(9):566–78.

62.   Kondratskyi A, Yassine M, Kondratska K, Skryma R, Slomianny C, Prevarskaya N. Calcium-permeable ion channels in control of autophagy and cancer. Front Physiol. 2013;4:272–272.

63.   Braun M, Ramracheya R, Bengtsson M, Zhang Q, Karanauskaite J, Partridge C, et al. Voltage-gated ion channels in human pancreatic beta-cells: electrophysiological characterization and role in insulin secretion. Diabetes. 2008 Jun;57(6):1618–28.

64.   Roux B. Ion channels and ion selectivity. Essays Biochem. 2017 May;61(2):201–9.

65.   Talvenheimo JA. The purification of ion channels from excitable cells. J Membr Biol. 1985;87(2):77–91.

66.   Gusovsky F, Daly JW. Formation of second messengers in response to activation of ion channels in excitable cells. Cell Mol Neurobiol. 1988 Jun;8(2):157–69.

67.   Bertrán GC, D’Alessio C, Kotsias BA. [Ion channels in non excitable cells]. Medicina (Mex). 1995;55(5 Pt 1):449–56.

68.   Kramer RH, Chambers JJ, Trauner D. Photochemical tools for remote control of ion channels in excitable cells. Nat Chem Biol. 2005 Dec;1(7):360–5.

69.   Deng XL, Lau CP, Lai K, Cheung KF, Lau GK, Li GR. Cell cycle-dependent expression of potassium channels and cell proliferation in rat mesenchymal stem cells from bone marrow. Cell Prolif. 2007 Oct;40(5):656–70.

70.   Urrego D, Tomczak AP, Zahed F, Stühmer W, Pardo LA. Potassium channels in cell cycle and cell proliferation. Philos Trans R Soc Lond B Biol Sci. 2014 Mar;369(1638):20130094–20130094.

71.   Ekhterae D, Platoshyn O, Krick S, Yu Y, McDaniel SS, Yuan JX. Bcl-2 decreases voltage-gated K+ channel activity and enhances survival in vascular smooth muscle cells. Am J Physiol Cell Physiol. 2001 Jul;281(1):C157-65.

72.   Williams SS, French JN, Gilbert M, Rangaswami AA, Walleczek J, Knox SJ. Bcl-2 overexpression results in enhanced capacitative calcium entry and resistance to SKF-96365-induced apoptosis. Cancer Res. 2000 Aug;60(16):4358–61.

73.   Roden DM, Kupershmidt S. From genes to channels: normal mechanisms. Cardiovasc Res. 1999 May;42(2):318–26.

74.   Wickenden A, Priest B, Erdemli G. Ion channel drug discovery: challenges and future directions. Future Med Chem. 2012 Apr;4(5):661–79.

75.   Chen H, Ikeda SR. Modulation of ion channels and synaptic transmission by a human sensory neuron-specific G-protein-coupled receptor, SNSR4/mrgX1, heterologously expressed in cultured rat neurons. J Neurosci Off J Soc Neurosci. 2004 May;24(21):5044–53.

76.   Anderson AJ, Harvey AL. Effects of the potassium channel blocking dendrotoxins on acetylcholine release and motor nerve terminal activity. Br J Pharmacol. 1988 Jan;93(1):215–21.

77.   Ge L, Hoa NT, Wilson Z, Arismendi-Morillo G, Kong XT, Tajhya RB, et al. Big Potassium (BK) ion channels in biology, disease and possible targets for cancer immunotherapy. Int Immunopharmacol. 2014 Oct;22(2):427–43.

78.   Leanza L, Biasutto L, Managò A, Gulbins E, Zoratti M, Szabò I. Intracellular ion channels and cancer. Front Physiol. 2013 Sep;4:227–227.

79.   Peruzzo R, Biasutto L, Szabò I, Leanza L. Impact of intracellular ion channels on cancer development and progression. Eur Biophys J EBJ. 2016 Oct;45(7):685–707.

80.   Lev S, Moreno H, Martinez R, Canoll P, Peles E, Musacchio JM, et al. Protein tyrosine kinase PYK2 involved in Ca(2+)-induced regulation of ion channel and MAP kinase functions. Nature. 1995 Aug;376(6543):737–45.

81.   Cheng W, Zhu Y, Wang H. The MAPK pathway is involved in the regulation of rapid pacing-induced ionic channel remodeling in rat atrial myocytes. Mol Med Rep. 2016 Mar;13(3):2677–82.

82.   Melnikova IN, Gardner PD. The signal transduction pathway underlying ion channel gene regulation by SP1-C-Jun interactions. J Biol Chem. 2001 Jun;276(22):19040–5.

83.   Bahcheli AT, Min HK, Bayati M, Zhao H, Fortuna A, Dong W, et al. Pan-cancer ion transport signature reveals functional regulators of glioblastoma aggression. EMBO J. 2024 Jan;43(2):196–224.

84.   Hoffmann EK, Lambert IH. Ion channels and transporters in the development of drug resistance in cancer cells. Philos Trans R Soc Lond B Biol Sci. 2014 Mar;369(1638):20130109–20130109.

85.   Szado T, Vanderheyden V, Parys JB, De Smedt H, Rietdorf K, Kotelevets L, et al. Phosphorylation of inositol 1,4,5-trisphosphate receptors by protein kinase B/Akt inhibits Ca2+ release and apoptosis. Proc Natl Acad Sci U S A. 2008 Feb;105(7):2427–32.

86.   Liu H, Hughes JD, Rollins S, Chen B, Perkins E. Calcium entry via ORAI1 regulates glioblastoma cell proliferation and apoptosis. Exp Mol Pathol. 2011 Dec;91(3):753–60.

87.   McFerrin MB, Turner KL, Cuddapah VA, Sontheimer H. Differential role of IK and BK potassium channels as mediators of intrinsic and extrinsic apoptotic cell death. Am J Physiol Cell Physiol. 2012 Nov;303(10):C1070-8.

88.   Ransom CB, Sontheimer H. BK channels in human glioma cells. J Neurophysiol. 2001 Feb;85(2):790–803.

89.   Ransom CB, Liu X, Sontheimer H. BK channels in human glioma cells have enhanced calcium sensitivity. Glia. 2002 Jun;38(4):281–91.

90.   Liu X, Chang Y, Reinhart PH, Sontheimer H, Chang Y. Cloning and characterization of glioma BK, a novel BK channel isoform highly expressed in human glioma cells. J Neurosci Off J Soc Neurosci. 2002 Mar;22(5):1840–9.

91.   Olsen ML, Schade S, Lyons SA, Amaral MD, Sontheimer H. Expression of voltage-gated chloride channels in human glioma cells. J Neurosci Off J Soc Neurosci. 2003 Jul;23(13):5572–82.

92.   Sontheimer H. Ion channels and amino acid transporters support the growth and invasion of primary brain tumors. Mol Neurobiol. 2004 Feb;29(1):61–71.

93.   McFerrin MB, Sontheimer H. A role for ion channels in glioma cell invasion. Neuron Glia Biol. 2006 Feb;2(1):39–49.

94.   Ruggieri P, Mangino G, Fioretti B, Catacuzzeno L, Puca R, Ponti D, et al. The inhibition of KCa3.1 channels activity reduces cell motility in glioblastoma derived cancer stem cells. PloS One. 2012;7(10):e47825–e47825.

95.   Huber SM, Butz L, Stegen B, Klumpp L, Klumpp D, Eckert F. Role of ion channels in ionizing radiation-induced cell death. Biochim Biophys Acta. 2015 Oct;1848(10 Pt B):2657–64.

96.   Habela CW, Olsen ML, Sontheimer H. ClC3 is a critical regulator of the cell cycle in normal and malignant glial cells. J Neurosci Off J Soc Neurosci. 2008 Sep;28(37):9205–17.

97.   Turner KL, Honasoge A, Robert SM, McFerrin MM, Sontheimer H. A proinvasive role for the Ca(2+) -activated K(+) channel KCa3.1 in malignant glioma. Glia. 2014 Jun;62(6):971–81.

98.   Barbieri F, Verduci I, Carlini V, Zona G, Pagano A, Mazzanti M, et al. Repurposed Biguanide Drugs in Glioblastoma Exert Antiproliferative Effects via the Inhibition of Intracellular Chloride Channel 1 Activity. Front Oncol. 2019;9:135–135.

99.   García-Santisteban I, Peters GJ, Giovannetti E, Rodríguez JA. USP1 deubiquitinase: cellular functions, regulatory mechanisms and emerging potential as target in cancer therapy. Mol Cancer. 2013 Aug;12:91–91.

100. Ranjan A, Srivastava SK. Penfluridol suppresses glioblastoma tumor growth by Akt-mediated inhibition of GLI1. Oncotarget. 2017 May;8(20):32960–76.

101. Pollak J, Rai KG, Funk CC, Arora S, Lee E, Zhu J, et al. Ion channel expression patterns in glioblastoma stem cells with functional and therapeutic implications for malignancy. PloS One. 2017;12(3):e0172884–e0172884.

102. Madhavan S, Zenklusen JC, Kotliarov Y, Sahni H, Fine HA, Buetow K. Rembrandt: helping personalized medicine become a reality through integrative translational research. Mol Cancer Res MCR. 2009 Feb;7(2):157–67.

103. Zhang Y, Zhang J, Jiang D, Zhang D, Qian Z, Liu C, et al. Inhibition of T-type Ca2+ channels by endostatin attenuates human glioblastoma cell proliferation and migration. Br J Pharmacol. 2012 Jun;166(4):1247–60.

104. Zhang Y, Cruickshanks N, Yuan F, Wang B, Pahuski M, Wulfkuhle J, et al. Targetable T-type Calcium Channels Drive Glioblastoma. Cancer Res. 2017 Jul;77(13):3479–90.

105. Cheng YR, Jiang BY, Chen CC. Acid-sensing ion channels: dual function proteins for chemo-sensing and mechano-sensing. J Biomed Sci. 2018 May;25(1):46–46.

106. Sivils A, Yang F, Wang JQ, Chu XP. Acid-Sensing Ion Channel 2: Function and Modulation. Membranes. 2022 Jan;12(2).

107. Berdiev BK, Xia J, McLean LA, Markert JM, Gillespie GY, Mapstone TB, et al. Acid-sensing ion channels in malignant gliomas. J Biol Chem. 2003 Apr;278(17):15023–34.

108. Tian Y, Bresenitz P, Reska A, El Moussaoui L, Beier CP, Gründer S. Glioblastoma cancer stem cell lines express functional acid sensing ion channels ASIC1a and ASIC3. Sci Rep. 2017 Oct;7(1):13674–13674.

109. Caldwell JH, Schaller KL, Lasher RS, Peles E, Levinson SR. Sodium channel Nav1. 6 is localized at nodes of Ranvier, dendrites, and synapses. Proc Natl Acad Sci. 2000;97(10):5616–20.

110. Ai Y, Zhang X, Hu X, Gao J, Liu J, Ou S, et al. Role of the voltage‑gated sodium channel Nav1.6 in glioma and candidate drugs screening. Int J Mol Med. 2023 Jun;51(6).

111. Boyle Y, Johns TG, Fletcher EV. Potassium Ion Channels in Malignant Central Nervous System Cancers. Cancers. 2022 Sep;14(19).

112. Berkefeld H, Sailer CA, Bildl W, Rohde V, Thumfart JO, Eble S, et al. BKCa-Cav channel complexes mediate rapid and localized Ca2+-activated K+ signaling. Science. 2006 Oct;314(5799):615–20.

113. Comes N, Bielanska J, Vallejo-Gracia A, Serrano-Albarrás A, Marruecos L, Gómez D, et al. The voltage-dependent K+ channels Kv1. 3 and Kv1. 5 in human cancer. Front Physiol. 2013;4:283.

114. Bielanska J, Hernandez-Losa J, Perez-Verdaguer M, Moline T, Somoza R, Cajal S, et al. Voltage-dependent potassium channels Kv1. 3 and Kv1. 5 in human cancer. Curr Cancer Drug Targets. 2009;9(8):904–14.

115. Arvind S, Arivazhagan A, Santosh V, Chandramouli BA. Differential expression of a novel voltage gated potassium channel--Kv 1.5 in astrocytomas and its impact on prognosis in glioblastoma. Br J Neurosurg. 2012 Feb;26(1):16–20.

116. Meizeng L, Tian P, Qing Zhao, Xialin Ma, Yunxiang Zhang, Li. Potassium channels: Novel targets for tumor diagnosis and chemoresistance. Front Oncol. 2023;(12):1074469.

117. Ohno Y, Kunisawa N, Shimizu S. Emerging roles of astrocyte Kir4. 1 channels in the pathogenesis and treatment of brain diseases. Int J Mol Sci. 2021;22(19):10236.

118. Higashimori H, Sontheimer H. Role of Kir4.1 channels in growth control of glia. Glia. 2007 Dec;55(16):1668–79.

119. Franciolini F, Petris A. Chloride channels of biological membranes. Biochim Biophys Acta BBA-Rev Biomembr. 1990;1031(2):247–59.

120. Nilius B, Droogmans G. Amazing chloride channels: an overview. Acta Physiol Scand. 2003;177(2):119–47.

121. Wang CY, Lai MD, Phan NN, Sun Z, Lin YC. Meta-Analysis of Public Microarray Datasets Reveals Voltage-Gated Calcium Gene Signatures in Clinical Cancer Patients. PloS One. 2015;10(7):e0125766–e0125766.

122. Griffin M, Basu S, Khan R, Smith S. CLIC1 and CLIC4 Ion Channel Deficiency Confers Increased Sensitivity to Tumour Treating Fields and Improved Survival in Paediatric Glioblastoma. Neuro-Oncol. 2022 Oct;24(Supplement_4):iv4–iv4.

123. Sciaccaluga M, Fioretti B, Catacuzzeno L, Pagani F, Bertollini C, Rosito M, et al. CXCL12-induced glioblastoma cell migration requires intermediate conductance Ca2+-activated K+ channel activity. Am J Physiol Cell Physiol. 2010 Jul;299(1):C175-84.

124. Cuddapah VA, Sontheimer H. Molecular interaction and functional regulation of ClC-3 by Ca2+/calmodulin-dependent protein kinase II (CaMKII) in human malignant glioma. J Biol Chem. 2010;285(15):11188–96.

125. Sun P, Mu Y, Zhang S. A novel NF-κB/MMP-3 signal pathway involves in the aggressivity of glioma promoted by Bmi-1. Tumor Biol. 2014;35:12721–7.

126. Wang B, Xie J, He HY, Huang EW, Cao QH, Luo L, et al. Suppression of CLC-3 chloride channel reduces the aggressiveness of glioma through inhibiting nuclear factor-κB pathway. Oncotarget. 2017;8(38):63788.

127. Sontheimer H. An unexpected role for ion channels in brain tumor metastasis. Exp Biol Med Maywood NJ. 2008 Jul;233(7):779–91.

128. Ernest NJ, Weaver AK, Van Duyn LB, Sontheimer HW. Relative contribution of chloride channels and transporters to regulatory volume decrease in human glioma cells. Am J Physiol-Cell Physiol. 2005;288(6):C1451–60.

129. Chinigò G, Castel H, Chever O, Gkika D. TRP Channels in Brain Tumors. Front Cell Dev Biol. 2021;9:617801–617801.

130. Alptekin M, Eroglu S, Tutar E, Sencan S, Geyik MA, Ulasli M, et al. Gene expressions of TRP channels in glioblastoma multiforme and relation with survival. Tumour Biol J Int Soc Oncodevelopmental Biol Med. 2015 Dec;36(12):9209–13.

131. Gehring MP, Kipper F, Nicoletti NF, Sperotto ND, Zanin R, Tamajusuku ASK, et al. P2X7 receptor as predictor gene for glioma radiosensitivity and median survival. Int J Biochem Cell Biol. 2015 Nov;68:92–100.

132. Chen X, Momin A, Wanggou S, Wang X, Min HK, Dou W, et al. Mechanosensitive brain tumor cells construct blood-tumor barrier to mask chemosensitivity. Neuron. 2023 Jan;111(1):30-48.e14.

133. Qu S, Hu T, Qiu O, Su Y, Gu J, Xia Z. Effect of Piezo1 Overexpression on Peritumoral Brain Edema in Glioblastomas. AJNR Am J Neuroradiol. 2020 Aug;41(8):1423–9.

134. Goldman DE. Potential, impedance, and rectification in membranes. J Gen Physiol. 1943;27(1):37–60.

135. Levin M. Large-scale biophysics: ion flows and regeneration. Trends Cell Biol. 2007;17(6):261–70.

136. Hodgkin AL, Katz B. The effect of sodium ions on the electrical activity of the giant axon of the squid. J Physiol. 1949;108(1):37.

137. Imbrici P, Liantonio A, Camerino GM, De Bellis M, Camerino C, Mele A, et al. Therapeutic Approaches to Genetic Ion Channelopathies and Perspectives in Drug Discovery. Front Pharmacol. 2016;7:121–121.

138. Markman JD, Dworkin RH. Ion channel targets and treatment efficacy in neuropathic pain. J Pain. 2006 Jan;7(1 Suppl 1):S38-47.

139. Grigoriadis DE, Hoare SRJ, Lechner SM, Slee DH, Williams JA. Drugability of extracellular targets: discovery of small molecule drugs targeting allosteric, functional, and subunit-selective sites on GPCRs and ion channels. Neuropsychopharmacol Off Publ Am Coll Neuropsychopharmacol. 2009 Jan;34(1):106–25.

140. Caban A, Pisarczyk K, Kopacz K, Kapuśniak A, Toumi M, Rémuzat C, et al. Filling the gap in CNS drug development: evaluation of the role of drug repurposing. J Mark Access Health Policy. 2017;5(1):1299833.

141. McManus OB, Garcia ML, Weaver D, Bryant M, Titus S, Herrington JB. Ion Channel Screening, in Assay Guidance Manual. 2012.

142. Gubič Š, Hendrickx LA, Toplak Ž, Sterle M, Peigneur S, Tomašič T, et al. Discovery of KV 1.3 ion channel inhibitors: Medicinal chemistry approaches and challenges. Med Res Rev. 2021 Jul;41(4):2423–73.

143. Yu H bo, Li M, Wang W ping, Wang X liang. High throughput screening technologies for ion channels. Acta Pharmacol Sin. 2016 Jan;37(1):34–43.

144. Kwok TCY, Ricker N, Fraser R, Chan AW, Burns A, Stanley EF, et al. A small-molecule screen in C. elegans yields a new calcium channel antagonist. Nature. 2006 May;441(7089):91–5.

145. Burns AR, Wallace IM, Wildenhain J, Tyers M, Giaever G, Bader GD, et al. A predictive model for drug bioaccumulation and bioactivity in Caenorhabditis elegans. Nat Chem Biol. 2010 Jul;6(7):549–57.

146. Jiang Q, Li K, Lu WJ, Li S, Chen X, Liu XJ, et al. Identification of small-molecule ion channel modulators in C. elegans channelopathy models. Nat Commun. 2018 Sep;9(1):3941–3941.

147. Clare JJ. Targeting ion channels for drug discovery. Discov Med. 2010 Mar;9(46):253–60.

148. Hammarstrom AK, Gage PW. Inhibition of oxidative metabolism increases persistent sodium current in rat CA1 hippocampal neurons. J Physiol. 1998 Aug;510 ( Pt 3)(Pt 3):735–41.

149. Issa FA, Mock AF, Sagasti A, Papazian DM. Spinocerebellar ataxia type 13 mutation that is associated with disease onset in infancy disrupts axonal pathfinding during neuronal development. Dis Model Mech. 2012 Nov;5(6):921–9.

150. Cooper EC, Jan LY. Ion channel genes and human neurological disease: recent progress, prospects, and challenges. Proc Natl Acad Sci U S A. 1999 Apr;96(9):4759–66.

151. Lipkind GM, Fozzard HA. Molecular model of anticonvulsant drug binding to the voltage-gated sodium channel inner pore. Mol Pharmacol. 2010 Oct;78(4):631–8.

152. Mycielska ME, Palmer CP, Brackenbury WJ, Djamgoz MB. Expression of Na+‐dependent citrate transport in a strongly metastatic human prostate cancer PC‐3M cell line: regulation by voltage‐gated Na+ channel activity. J Physiol. 2005;563(2):393–408.

153. Cameron IL, Smith NK, Pool T, Sparks R. Intracellular concentration of sodium and other elements as related to mitogenesis and oncogenesis in vivo. Cancer Res. 1980;40(5):1493–500.

154. 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.

155. Szabo I, Zoratti M, Biasutto L. Targeting mitochondrial ion channels for cancer therapy. Redox Biol. 2021 Jun;42:101846–101846.

156. Yang J, Yu G, Sessler JL, Shin I, Gale PA, Huang F. Artificial transmembrane ion transporters as potential therapeutics. Chem. 2021 Dec;7(12):3256–91.

157. Prevarskaya N, Skryma R, Shuba Y. Ion channels and the hallmarks of cancer. Trends Mol Med. 2010;16(3):107–21.

158. Arcangeli A, Becchetti A. Integrin structure and functional relation with ion channels. Integrins Ion Channels Mol Complexes Signal. 2010;1–7.

159. Djamgoz MB, Mycielska M, Madeja Z, Fraser SP, Korohoda W. Directional movement of rat prostate cancer cells in direct-current electric field: involvement of voltagegated Na+ channel activity. J Cell Sci. 2001;114(14):2697–705.

160. K K. Ion channels and cancer. J Membr Biol. 2005;(205):159–73.

161. Lang F, Stournaras C. Ion channels in cancer: future perspectives and clinical potential. Philos Trans R Soc Lond B Biol Sci. 2014 Mar;369(1638):20130108–20130108.

162. Dai X, Wang D, Zhang J. Programmed cell death, redox imbalance, and cancer therapeutics. Apoptosis Int J Program Cell Death. 2021 Aug;26(7–8):385–414.

163. Azimi I, Robitaille M, Armitage K, So CL, Milevskiy MJG, Northwood K, et al. Activation of the Ion Channel TRPV4 Induces Epithelial to Mesenchymal Transition in Breast Cancer Cells. Int J Mol Sci. 2020 Dec;21(24).

164. Nelson M, Yang M, Dowle AA, Thomas JR, Brackenbury WJ. The sodium channel-blocking antiepileptic drug phenytoin inhibits breast tumour growth and metastasis. Mol Cancer. 2015 Jan;14(1):13–13.

165. Jahchan NS, Dudley JT, Mazur PK, Flores N, Yang D, Palmerton A, et al. A drug repositioning approach identifies tricyclic antidepressants as inhibitors of small cell lung cancer and other neuroendocrine tumors. Cancer Discov. 2013 Dec;3(12):1364–77.

166. Millward MJ, Cantwell BM, Munro NC, Robinson A, Corris PA, Harris AL. Oral verapamil with chemotherapy for advanced non-small cell lung cancer: a randomised study. Br J Cancer. 1993 May;67(5):1031–5.

167. Dalton WS, Crowley JJ, Salmon SS, Grogan TM, Laufman LR, Weiss GR, et al. 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. 1995 Feb;75(3):815–20.

168. Sarkaria JN, Kitange GJ, James CD, Plummer R, Calvert H, Weller M, et al. Mechanisms of chemoresistance to alkylating agents in malignant glioma. Clin Cancer Res Off J Am Assoc Cancer Res. 2008 May;14(10):2900–8.

169. Friedman HS, McLendon RE, Kerby T, Dugan M, Bigner SH, Henry AJ, et al. DNA mismatch repair and O6-alkylguanine-DNA alkyltransferase analysis and response to Temodal in newly diagnosed malignant glioma. J Clin Oncol Off J Am Soc Clin Oncol. 1998 Dec;16(12):3851–7.

170. Simonetti G, Gaviani P, Botturi A, Innocenti A, Lamperti E, Silvani A. Clinical management of grade III oligodendroglioma. Cancer Manag Res. 2015;7:213–23.

171. Jamal M, Rath BH, Tsang PS, Camphausen K, Tofilon PJ. The brain microenvironment preferentially enhances the radioresistance of CD133(+) glioblastoma stem-like cells. Neoplasia N Y N. 2012 Feb;14(2):150–8.

172. Blaney SM, Poplack DG. Pharmacologic strategies for the treatment of meningeal malignancy. Invest New Drugs. 1996;14:69–85.

173. Rominiyi O, Vanderlinden A, Clenton SJ, Bridgewater C, Al-Tamimi Y, Collis SJ. Tumour treating fields therapy for glioblastoma: current advances and future directions. Br J Cancer. 2021;124(4):697–709.

174. Varricchio A, Yool AJ. Aquaporins and Ion Channels as Dual Targets in the Design of Novel Glioblastoma Therapeutics to Limit Invasiveness. Cancers. 2023 Jan;15(3).

175. Xu B, Mao J, Wang L, Zhu L, Li H, Wang W, et al. ClC-3 chloride channels are essential for cell proliferation and cell cycle progression in nasopharyngeal carcinoma cells. Acta Biochim Biophys Sin. 2010 Jun;42(6):370–80.

176. Kasai T, Nakamura K, Vaidyanath A, Chen L, Sekhar S, El-Ghlban S, et al. Chlorotoxin Fused to IgG-Fc Inhibits Glioblastoma Cell Motility via Receptor-Mediated Endocytosis. J Drug Deliv. 2012;2012:975763–975763.

177. Mamelak AN, Jacoby DB. Targeted delivery of antitumoral therapy to glioma and other malignancies with synthetic chlorotoxin (TM-601). Expert Opin Drug Deliv. 2007 Mar;4(2):175–86.

178. Hockaday DC, Shen S, Fiveash J, Raubitschek A, Colcher D, Liu A, et al. Imaging glioma extent with 131I-TM-601. J Nucl Med Off Publ Soc Nucl Med. 2005 Apr;46(4):580–6.

179. Lyons SA, O’Neal J, Sontheimer H. Chlorotoxin, a scorpion-derived peptide, specifically binds to gliomas and tumors of neuroectodermal origin. Glia. 2002 Aug;39(2):162–73.

180. Setti M, Savalli N, Osti D, Richichi C, Angelini M, Brescia P, et al. Functional role of CLIC1 ion channel in glioblastoma-derived stem/progenitor cells. J Natl Cancer Inst. 2013 Nov;105(21):1644–55.

181. Bayat Mokhtari R, Homayouni TS, Baluch N, Morgatskaya E, Kumar S, Das B, et al. Combination therapy in combating cancer. Oncotarget. 2017 Jun;8(23):38022–43.

182. Mathews J, Kuchling F, Baez-Nieto D, Diberardinis M, Pan JQ, Levin M. Ion Channel Drugs Suppress Cancer Phenotype in NG108-15 and U87 Cells: Toward Novel Electroceuticals for Glioblastoma. Cancers. 2022 Mar;14(6).

183. Su VYF, Hu YW, Chou KT, Ou SM, Lee YC, Lin EYH, et al. Amiodarone and the risk of cancer: a nationwide population-based study. Cancer. 2013 May;119(9):1699–705.

184. Kim BJ, Kim SY, Lee S, Jeon JH, Matsui H, Kwon YK, et al. The role of transient receptor potential channel blockers in human gastric cancer cell viability. Can J Physiol Pharmacol. 2012 Feb;90(2):175–86.

185. Kim IY, Kang YJ, Yoon MJ, Kim EH, Kim SU, Kwon TK, et al. Amiodarone sensitizes human glioma cells but not astrocytes to TRAIL-induced apoptosis via CHOP-mediated DR5 upregulation. Neuro-Oncol. 2011 Mar;13(3):267–79.

186. Rath BH, Camphausen K, Tofilon PJ. Glioblastoma radiosensitization by pimozide. Transl Cancer Res. 2016 Nov;5(Suppl 6):S1029–32.

187. Kondo S, Yin D, Morimura T, Kubo H, Nakatsu S, Takeuchi J. Combination therapy with cisplatin and nifedipine induces apoptosis in cisplatin-sensitive and cisplatin-resistant human glioblastoma cells. Br J Cancer. 1995 Feb;71(2):282–9.

188. Krouse AJ, Gray L, Macdonald T, McCray J. Repurposing and Rescuing of Mibefradil, an Antihypertensive, for Cancer: A Case Study. Assay Drug Dev Technol. 2015 Dec;13(10):650–3.

189. Holdhoff M, Ye X, Supko JG, Nabors LB, Desai AS, Walbert T, et al. Timed sequential therapy of the selective T-type calcium channel blocker mibefradil and temozolomide in patients with recurrent high-grade gliomas. Neuro-Oncol. 2017 Jun;19(6):845–52.

190. Keir ST, Friedman HS, Reardon DA, Bigner DD, Gray LA. Mibefradil, a novel therapy for glioblastoma multiforme: cell cycle synchronization and interlaced therapy in a murine model. J Neurooncol. 2013 Jan;111(2):97–102.

191. Mullins ME, Horowitz BZ, Linden DH, Smith GW, Norton RL, Stump J. Life-threatening interaction of mibefradil and beta-blockers with dihydropyridine calcium channel blockers. JAMA. 1998 Jul;280(2):157–8.

192. Papin JA, Hunter T, Palsson BO, Subramaniam S. Reconstruction of cellular signalling networks and analysis of their properties. Nat Rev Mol Cell Biol. 2005 Feb;6(2):99–111.

193. Higginbottom SL, Tomaskovic-Crook E, Crook JM. Considerations for modelling diffuse high-grade gliomas and developing clinically relevant therapies. Cancer Metastasis Rev. 2023;42(2):507–41.

194. Zottel A, Jovčevska I, Šamec N. Non-animal glioblastoma models for personalized treatment. Heliyon. 2023;

195. Ahmed T. Biomaterial-based in vitro 3D modeling of glioblastoma multiforme. Cancer Pathog Ther. 2023;1(03):177–94.

196. Fan Y, Nguyen DT, Akay Y, Xu F, Akay M. Engineering a brain cancer chip for high-throughput drug screening. Sci Rep. 2016;6(1):25062.

197. Thenuwara G, Javed B, Singh B, Tian F. Biosensor-Enhanced Organ-on-a-Chip Models for Investigating Glioblastoma Tumor Microenvironment Dynamics. Sensors. 2024;24(9):2865.

198. Maffeo C, Bhattacharya S, Yoo J, Wells D, Aksimentiev A. Modeling and simulation of ion channels. Chem Rev. 2012;112(12):6250–84.

199. Mamelak AN, Rosenfeld S, Bucholz R, Raubitschek A, Nabors LB, Fiveash JB, et al. Phase I single-dose study of intracavitary-administered iodine-131-TM-601 in adults with recurrent high-grade glioma. J Clin Oncol Off J Am Soc Clin Oncol. 2006 Aug;24(22):3644–50.

200. FOLGIERO V, ROMANIA P, ROSSI F, CAFORIO M, NIC M, DIBUSZ K, et al. Advanced Non-animal Models in Biomedical Research. 2020;

201. Stringer BW, Day BW, D’Souza RCJ, Jamieson PR, Ensbey KS, Bruce ZC, et al. A reference collection of patient-derived cell line and xenograft models of proneural, classical and mesenchymal glioblastoma. Sci Rep. 2019 Mar;9(1):4902–4902.

202. Chen JP, Luan Y, You CX, Chen XH, Luo RC, Li R. TRPM7 regulates the migration of human nasopharyngeal carcinoma cell by mediating Ca(2+) influx. Cell Calcium. 2010 May;47(5):425–32.

203. Ding X, He Z, Shi Y, Wang Q, Wang Y. Targeting TRPC6 channels in oesophageal carcinoma growth. Expert Opin Ther Targets. 2010 May;14(5):513–27.

204. Shiozaki A, Kudou M, Ichikawa D, Fujiwara H, Shimizu H, Ishimoto T, et al. Esophageal cancer stem cells are suppressed by tranilast, a TRPV2 channel inhibitor. J Gastroenterol. 2018 Feb;53(2):197–207.

205. Choi S, Cui C, Luo Y, Kim SH, Ko JK, Huo X, et al. Selective inhibitory effects of zinc on cell proliferation in esophageal squamous cell carcinoma through Orai1. FASEB J Off Publ Fed Am Soc Exp Biol. 2018 Jan;32(1):404–16.

206. Asghar MY, Magnusson M, Kemppainen K, Sukumaran P, Löf C, Pulli I, et al. Transient Receptor Potential Canonical 1 (TRPC1) Channels as Regulators of Sphingolipid and VEGF Receptor Expression: IMPLICATIONS FOR THYROID CANCER CELL MIGRATION AND PROLIFERATION. J Biol Chem. 2015 Jun;290(26):16116–31.

207. Shapovalov G, Ritaine A, Skryma R, Prevarskaya N. Role of TRP ion channels in cancer and tumorigenesis. Semin Immunopathol. 2016 May;38(3):357–69.

208. Wu H, Wang H, Guan S, Zhang J, Chen Q, Wang X, et al. Cell-specific regulation of proliferation by Ano1/TMEM16A in breast cancer with different ER, PR, and HER2 status. Oncotarget. 2017 Oct;8(49):84996–5013.

209. Elbaz M, Ahirwar D, Xiaoli Z, Zhou X, Lustberg M, Nasser MW, et al. TRPV2 is a novel biomarker and therapeutic target in triple negative breast cancer. Oncotarget. 2018 Sep;9(71):33459–70.

210. Badaoui M, Mimsy-Julienne C, Saby C, Van Gulick L, Peretti M, Jeannesson P, et al. Collagen type 1 promotes survival of human breast cancer cells by overexpressing Kv10.1 potassium and Orai1 calcium channels through DDR1-dependent pathway. Oncotarget. 2018 May;9(37):24653–71.

211. Potier M, Chantome A, Joulin V, Girault A, Roger S, Besson P, et al. The SK3/K(Ca)2.3 potassium channel is a new cellular target for edelfosine. Br J Pharmacol. 2011 Jan;162(2):464–79.

212. Hall DP, Cost NG, Hegde S, Kellner E, Mikhaylova O, Stratton Y, et al. TRPM3 and miR-204 establish a regulatory circuit that controls oncogenic autophagy in clear cell renal cell carcinoma. Cancer Cell. 2014 Nov;26(5):738–53.

213. Song J, Wang Y, Li X, Shen Y, Yin M, Guo Y, et al. Critical role of TRPC6 channels in the development of human renal cell carcinoma. Mol Biol Rep. 2013 Aug;40(8):5115–22.

214. Kim JH, Lkhagvadorj S, Lee MR, Hwang KH, Chung HC, Jung JH, et al. Orai1 and STIM1 are critical for cell migration and proliferation of clear cell renal cell carcinoma. Biochem Biophys Res Commun. 2014 May;448(1):76–82.

215. Rabjerg M, Oliván-Viguera A, Hansen LK, Jensen L, Sevelsted-Møller L, Walter S, et al. High expression of KCa3.1 in patients with clear cell renal carcinoma predicts high metastatic risk and poor survival. PloS One. 2015;10(4):e0122992–e0122992.

216. Wang R, Gurguis CI, Gu W, Ko EA, Lim I, Bang H, et al. Ion channel gene expression predicts survival in glioma patients. Sci Rep. 2015 Aug;5:11593–11593.

217. Peng J, Ou Q, Wu X, Zhang R, Zhao Q, Jiang W, et al. Expression of voltage-gated sodium channel Nav1.5 in non-metastatic colon cancer and its associations with estrogen receptor (ER)-β expression and clinical outcomes. Chin J Cancer. 2017 Nov;36(1):89–89.

218. Song P, Du Y, Song W, Chen H, Xuan Z, Zhao L, et al. KCa3.1 as an Effective Target for Inhibition of Growth and Progression of Intrahepatic Cholangiocarcinoma. J Cancer. 2017;8(9):1568–78.

219. Ye Y, Yin M, Huang B, Wang Y, Li X, Lou G. CLIC1 a novel biomarker of intraperitoneal metastasis in serous epithelial ovarian cancer. Tumour Biol J Int Soc Oncodevelopmental Biol Med. 2015 Jun;36(6):4175–9.

220. Yang SL, Cao Q, Zhou KC, Feng YJ, Wang YZ. Transient receptor potential channel C3 contributes to the progression of human ovarian cancer. Oncogene. 2009 Mar;28(10):1320–8.

221. Gao R, Shen Y, Cai J, Lei M, Wang Z. Expression of voltage-gated sodium channel alpha subunit in human ovarian cancer. Oncol Rep. 2010 May;23(5):1293–9.

222. Farias LMB, Ocaña DB, Díaz L, Larrea F, Avila-Chávez E, Cadena A, et al. Ether a go-go potassium channels as human cervical cancer markers. Cancer Res. 2004 Oct;64(19):6996–7001.

223. Hernandez-Plata E, Ortiz CS, Marquina-Castillo B, Medina-Martinez I, Alfaro A, Berumen J, et al. Overexpression of NaV 1.6 channels is associated with the invasion capacity of human cervical cancer. Int J Cancer. 2012 May;130(9):2013–23.

224. Arcangeli A, Romoli MR, Boni L, Gerlini G, Tofani L, Urso C, et al. High hERG1 expression in advanced melanoma. Melanoma Res. 2013 Jun;23(3):185–90.

225. Cao R, Meng Z, Liu T, Wang G, Qian G, Cao T, et al. Decreased TRPM7 inhibits activities and induces apoptosis of bladder cancer cells via ERK1/2 pathway. Oncotarget. 2016 Nov;7(45):72941–60.

226. Tian Y, Guan Y, Jia Y, Meng Q, Yang J. Chloride intracellular channel 1 regulates prostate cancer cell proliferation and migration through the MAPK/ERK pathway. Cancer Biother Radiopharm. 2014 Oct;29(8):339–44.

227. Zhang GM, Wan FN, Qin XJ, Cao DL, Zhang HL, Zhu Y, et al. Prognostic significance of the TREK-1 K2P potassium channels in prostate cancer. Oncotarget. 2015 Jul;6(21):18460–8.

228. Diss JKJ, Stewart D, Pani F, Foster CS, Walker MM, Patel A, et al. A potential novel marker for human prostate cancer: voltage-gated sodium channel expression in vivo. Prostate Cancer Prostatic Dis. 2005;8(3):266–73.

229. Lu J, Dong Q, Zhang B, Wang X, Ye B, Zhang F, et al. Chloride intracellular channel 1 (CLIC1) is activated and functions as an oncogene in pancreatic cancer. Med Oncol Northwood Lond Engl. 2015 Jun;32(6):616–616.

230. Yee NS, Chan AS, Yee JD, Yee RK. TRPM7 and TRPM8 Ion Channels in Pancreatic Adenocarcinoma: Potential Roles as Cancer Biomarkers and Targets. Scientifica. 2012;2012:415158–415158.

231. Yee NS, Brown RD, Lee MS, Zhou W, Jensen C, Gerke H, et al. TRPM8 ion channel is aberrantly expressed and required for preventing replicative senescence in pancreatic adenocarcinoma: potential role of TRPM8 as a biomarker and target. Cancer Biol Ther. 2012 Jun;13(8):592–9.

232. Chen CD, Wang CS, Huang YH, Chien KY, Liang Y, Chen WJ, et al. Overexpression of CLIC1 in human gastric carcinoma and its clinicopathological significance. Proteomics. 2007 Jan;7(1):155–67.

233. Xia J, Wang H, Huang H, Sun L, Dong S, Huang N, et al. Elevated Orai1 and STIM1 expressions upregulate MACC1 expression to promote tumor cell proliferation, metabolism, migration, and invasion in human gastric cancer. Cancer Lett. 2016 Oct;381(1):31–40.

234. Xia J, Huang N, Huang H, Sun L, Dong S, Su J, et al. Voltage-gated sodium channel Nav 1.7 promotes gastric cancer progression through MACC1-mediated upregulation of NHE1. Int J Cancer. 2016 Dec;139(11):2553–69.

235. Ding XW, Yang WB, Gao S, Wang W, Li Z, Hu WM, et al. Prognostic significance of hERG1 expression in gastric cancer. Dig Dis Sci. 2010 Apr;55(4):1004–10.

236. Selli C, Erac Y, Kosova B, Erdal ES, Tosun M. Silencing of TRPC1 regulates store-operated calcium entry and proliferation in Huh7 hepatocellular carcinoma cells. Biomed Pharmacother Biomedecine Pharmacother. 2015 Apr;71:194–200.

237. Zhang S, Wang XM, Yin ZY, Zhao WX, Zhou JY, Zhao BX, et al. Chloride intracellular channel 1 is overexpression in hepatic tumor and correlates with a poor prognosis. APMIS Acta Pathol Microbiol Immunol Scand. 2013 Nov;121(11):1047–53.

238. Liu Y, Zhao L, Ma W, Cao X, Chen H, Feng D, et al. The Blockage of KCa3.1 Channel Inhibited Proliferation, Migration and Promoted Apoptosis of Human Hepatocellular Carcinoma Cells. J Cancer. 2015;6(7):643–51.

239. D’Alessandro G, Catalano M, Sciaccaluga M, Chece G, Cipriani R, Rosito M, et al. KCa3.1 channels are involved in the infiltrative behavior of glioblastoma in vivo. Cell Death Dis. 2013 Aug;4(8):e773–e773.

240. Chigurupati S, Venkataraman R, Barrera D, Naganathan A, Madan M, Paul L, et al. Receptor channel TRPC6 is a key mediator of Notch-driven glioblastoma growth and invasiveness. Cancer Res. 2010 Jan;70(1):418–27.

241. Bomben VC, Sontheimer HW. Inhibition of transient receptor potential canonical channels impairs cytokinesis in human malignant gliomas. Cell Prolif. 2008 Feb;41(1):98–121.

242. Lee YS, Sayeed MM, Wurster RD. Inhibition of cell growth and intracellular Ca2+ mobilization in human brain tumor cells by Ca2+ channel antagonists. Mol Chem Neuropathol. 1994 Jun;22(2):81–95.

243. Venturini E, Leanza L, Azzolini M, Kadow S, Mattarei A, Weller M, et al. Targeting the Potassium Channel Kv1.3 Kills Glioblastoma Cells. Neurosignals. 2017;25(1):26–38.

244. Huang L, Li B, Tang S, Guo H, Li W, Huang X, et al. Mitochondrial KATP Channels Control Glioma Radioresistance by Regulating ROS-Induced ERK Activation. Mol Neurobiol. 2015 Aug;52(1):626–37.

245. Visa A, Sallán MC, Maiques O, Alza L, Talavera E, López-Ortega R, et al. T-Type Cav3. 1 channels mediate progression and chemotherapeutic resistance in glioblastoma. Cancer Res. 2019;79(8):1857–68.

246. Liu S, Ba Y, Li C, Xu G. Inactivation of CACNA1H induces cell apoptosis by initiating endoplasmic reticulum stress in glioma. Transl Neurosci. 2023;14(1):20220285.

247. Valerie NC, Dziegielewska B, Hosing AS, Augustin E, Gray LS, Brautigan DL, et al. Inhibition of T-type calcium channels disrupts Akt signaling and promotes apoptosis in glioblastoma cells. Biochem Pharmacol. 2013;85(7):888–97.

248. Mertens-Walker I, Bolitho C, Baxter RC, Marsh DJ. Gonadotropin-induced ovarian cancer cell migration and proliferation require extracellular signal-regulated kinase 1/2 activation regulated by calcium and protein kinase Cδ. Endocr Relat Cancer. 2010;17(2):335.

249. Bomben VC, Turner KL, Barclay TC, Sontheimer H. Transient receptor potential canonical channels are essential for chemotactic migration of human malignant gliomas. J Cell Physiol. 2011;226(7):1879–88.

250. Cuddapah VA, Turner KL, Sontheimer H. Calcium entry via TRPC1 channels activates chloride currents in human glioma cells. Cell Calcium. 2013;53(3):187–94.

251. Richter JM, Schaefer M, Hill K. Riluzole activates TRPC5 channels independently of PLC activity. Br J Pharmacol. 2014;171(1):158–70.

252. Zeng J, Wu Y, Zhuang S, Qin L, Hua S, Mungur R, et al. Identification of the role of TRPM8 in glioblastoma and its effect on proliferation, apoptosis and invasion of the U251 human glioblastoma cell line. Oncol Rep. 2019;42(4):1517–26.

253. Caceres AI, Liu B, Jabba SV, Achanta S, Morris JB, Jordt S. Transient receptor potential cation channel subfamily M member 8 channels mediate the anti‐inflammatory effects of eucalyptol. Br J Pharmacol. 2017;174(9):867–79.

254. Wang S, Li X, Hu Y, Wang L, Lv G, Feng Y, et al. Discovery of N-alkyl-N-benzyl thiazoles as novel TRPC antagonists for the treatment of glioblastoma multiforme. Eur J Med Chem. 2024;265:116066.

255. Wan J, Guo AA, King P, Guo S, Saafir T, Jiang Y, et al. TRPM7 induces tumorigenesis and stemness through notch activation in glioma. Front Pharmacol. 2020;11:590723.

256. Yin H, Cheng H, Li P, Yang Z. TRPC6 interacted with KCa1. 1 channels to regulate the proliferation and apoptosis of glioma cells. Arch Biochem Biophys. 2022;725:109268.

257. Freeman S, Elzamzamy O, Geldenhuys W, Hazlehurst L. CLIC1 is an Attractive Candidate for the Initial Binding Target of the Novel Peptide MTI‐101. FASEB J. 2020;34(S1):1–1.

258. Xiong D, Heyman NS, Airey J, Zhang M, Singer CA, Rawat S, et al. Cardiac-specific, inducible ClC-3 gene deletion eliminates native volume-sensitive chloride channels and produces myocardial hypertrophy in adult mice. J Mol Cell Cardiol. 2010;48(1):211–9.

259. Morelli MB, Nabissi M, Amantini C, Maggi F, Ricci-Vitiani L, Pallini R, et al. TRPML2 Mucolipin Channels Drive the Response of Glioma Stem Cells to Temozolomide and Affect the Overall Survival in Glioblastoma Patients. Int J Mol Sci. 2022;23(23):15356.

260. Santoni G, Maggi F, Amantini C, Arcella A, Marinelli O, Nabissi M, et al. Coexpression of TRPML1 and TRPML2 Mucolipin Channels Affects the Survival of Glioblastoma Patients. Int J Mol Sci. 2022;23(14):7741.

261. Mori A, Lehmann S, O’Kelly J, Kumagai T, Desmond JC, Pervan M, et al. Capsaicin, a component of red peppers, inhibits the growth of androgen-independent, p53 mutant prostate cancer cells. Cancer Res. 2006;66(6):3222–9.

262. Prevarskaya N, Zhang L, Barritt G. TRP channels in cancer. Biochim Biophys Acta BBA-Mol Basis Dis. 2007;1772(8):937–46.

263. Huang T, Xu T, Wang Y, Zhou Y, Yu D, Wang Z, et al. Cannabidiol inhibits human glioma by induction of lethal mitophagy through activating TRPV4. Autophagy. 2021;17(11):3592–606.

264. Yang W, Wu P fei, Ma J xing, Liao M jun, Xu L shan, Yi L. TRPV4 activates the Cdc42/N-wasp pathway to promote glioblastoma invasion by altering cellular protrusions. Sci Rep. 2020;10(1):14151.

265. Thorneloe KS, Cheung M, Bao W, Alsaid H, Lenhard S, Jian MY, et al. An orally active TRPV4 channel blocker prevents and resolves pulmonary edema induced by heart failure. Sci Transl Med. 2012;4(159):159ra148-159ra148.

266. Venturini E, Leanza L, Azzolini M, Kadow S, Mattarei A, Weller M, et al. Targeting the potassium channel Kv1. 3 kills glioblastoma cells. Neurosignals. 2018;25(1):26–38.

267. Grimaldi A, D’Alessandro G, Di Castro MA, Lauro C, Singh V, Pagani F, et al. Kv1. 3 activity perturbs the homeostatic properties of astrocytes in glioma. Sci Rep. 2018;8(1):7654.

268. Leonard RJ, Garcia ML, Slaughter RS, Reuben JP. Selective blockers of voltage-gated K+ channels depolarize human T lymphocytes: mechanism of the antiproliferative effect of charybdotoxin. Proc Natl Acad Sci. 1992;89(21):10094–8.

269. Staudacher I, Jehle J, Staudacher K, Pledl HW, Lemke D, Schweizer PA, et al. HERG K+ channel-dependent apoptosis and cell cycle arrest in human glioblastoma cells. PloS One. 2014;9(2):e88164.

270. Shugg T, Dave N, Amarh E, Assiri AA, Pollok KE, Overholser BR. Letrozole targets the human ether‐a‐go‐go–related gene potassium current in glioblastoma. Basic Clin Pharmacol Toxicol. 2021;128(3):357–65.

271. Cherubini A, Taddei G, Crociani O, Paglierani M, Buccoliero A, Fontana L, et al. HERG potassium channels are more frequently expressed in human endometrial cancer as compared to non-cancerous endometrium. Br J Cancer. 2000;83(12):1722–9.

272. Caramia M, Sforna L, Franciolini F, Catacuzzeno L. The volume-regulated anion channel in glioblastoma. Cancers. 2019;11(3):307.

273. Lv J, Liang Y, Zhang S, Lan Q, Xu Z, Wu X, et al. DCPIB, an inhibitor of volume-regulated anion channels, distinctly modulates K2P channels. ACS Chem Neurosci. 2019;10(6):2786–93.

274. Voss FK, Ullrich F, Münch J, Lazarow K, Lutter D, Mah N, et al. Identification of LRRC8 heteromers as an essential component of the volume-regulated anion channel VRAC. Science. 2014;344(6184):634–8.

275. Baker EM, Thompson CH, Hawkins NA, Wagnon JL, Wengert ER, Patel MK, et al. The novel sodium channel modulator GS‐458967 (GS 967) is an effective treatment in a mouse model of SCN 8A encephalopathy. Epilepsia. 2018;59(6):1166–76.

276. Xing D, Wang J, Ou S, Wang Y, Qiu B, Ding D, et al. Expression of neonatal Nav1. 5 in human brain astrocytoma and its effect on proliferation, invasion and apoptosis of astrocytoma cells. Oncol Rep. 2014;31(6):2692–700.

277. O’Hara T, Rudy Y. Quantitative comparison of cardiac ventricular myocyte electrophysiology and response to drugs in human and nonhuman species. Am J Physiol-Heart Circ Physiol. 2012;302(5):H1023–30.

278. Ramaswamy P, Dalavaikodihalli Nanjaiah N, Prasad C, Goswami K. Transcriptional modulation of calcium‐permeable AMPA receptor subunits in glioblastoma by MEK–ERK1/2 inhibitors and their role in invasion. Cell Biol Int. 2020;44(3):830–7.

279. Arai A, Kessler M. Pharmacology of ampakine modulators: from AMPA receptors to synapses and behavior. Curr Drug Targets. 2007;8(5):583–602.

280. Partin KM, Fleck MW, Mayer ML. AMPA receptor flip/flop mutants affecting deactivation, desensitization, and modulation by cyclothiazide, aniracetam, and thiocyanate. J Neurosci. 1996;16(21):6634–47.

281. Kim HJ, Kim JY, Jung CW, Lee YS, An JY, Kim EH, et al. ANO1 regulates the maintenance of stemness in glioblastoma stem cells by stabilizing EGFRvIII. Oncogene. 2021;40(8):1490–502.

282. Xuan Z bo, Wang Y ji, Xie J. ANO6 promotes cell proliferation and invasion in glioma through regulating the ERK signaling pathway. OncoTargets Ther. 2019;6721–31.

283. Britschgi A, Bill A, Brinkhaus H, Rothwell C, Clay I, Duss S, et al. Calcium-activated chloride channel ANO1 promotes breast cancer progression by activating EGFR and CAMK signaling. Proc Natl Acad Sci. 2013;110(11):E1026–34.

284. Sheng Y, Wu B, Leng T, Zhu L, Xiong Z. Acid-sensing ion channel 1 (ASIC1) mediates weak acid-induced migration of human malignant glioma cells. Am J Cancer Res. 2021;11(3):997.

285. King P, Wan J, Guo AA, Guo S, Jiang Y, Liu M. Regulation of gliomagenesis and stemness through acid sensor ASIC1a. Int J Oncol. 2021;59(4):1–15.

286. Leng T, Si H, Li J, Yang T, Zhu M, Wang B, et al. Amiloride Analogs as ASIC 1a Inhibitors. CNS Neurosci Ther. 2016;22(6):468–76.

287. Kapoor N, Lee W, Clark E, Bartoszewski R, McNicholas CM, Latham CB, et al. Interaction of ASIC1 and ENaC subunits in human glioma cells and rat astrocytes. Am J Physiol-Cell Physiol. 2011;300(6):C1246–59.

288. Chen WC, Kuo TH, Tzeng YS, Tsai YC. Baicalin induces apoptosis in SW620 human colorectal carcinoma cells in vitro and suppresses tumor growth in vivo. Molecules. 2012;17(4):3844–57.

289. Huang L, Li B, Li W, Guo H, Zou F. ATP-sensitive potassium channels control glioma cells proliferation by regulating ERK activity. Carcinogenesis. 2009;30(5):737–44.

290. Hattersley AT, Ashcroft FM. Activating mutations in Kir6. 2 and neonatal diabetes: new clinical syndromes, new scientific insights, and new therapy. Diabetes. 2005;54(9):2503–13.

291. Rafiq M, Flanagan SE, Patch AM, Shields BM, Ellard S, Hattersley AT, et al. Effective treatment with oral sulfonylureas in patients with diabetes due to sulfonylurea receptor 1 (SUR1) mutations. Diabetes Care. 2008;31(2):204–9.

292. D’Alessandro G, Limatola C, Catalano M. Functional roles of the Ca2+-activated K+ channel, KCa3. 1, in brain tumors. Curr Neuropharmacol. 2018;16(5):636–43.

293. Gribkoff VK, Starrett Jr JE, Dworetzky SI. The Pharmacology and Molecular Biology of Large-Conductance Calcium= Activated (BK) Potassium Channels. Adv Pharmacol. 1996;37:319–48.

294. Zhou Y, Lingle CJ. Paxilline inhibits BK channels by an almost exclusively closed-channel block mechanism. J Gen Physiol. 2014;144(5):415–40.

295. Ataga KI, Smith WR, De Castro LM, Swerdlow P, Saunthararajah Y, Castro O, et al. Efficacy and safety of the Gardos channel blocker, senicapoc (ICA-17043), in patients with sickle cell anemia. Blood J Am Soc Hematol. 2008;111(8):3991–7.

296. Zhou W, Liu X, van Wijnbergen JWM, Yuan L, Liu Y, Zhang C, et al. Identification of PIEZO1 as a potential prognostic marker in gliomas. Sci Rep. 2020;10(1):16121.

297. Chen X, Wanggou S, Bodalia A, Zhu M, Dong W, Fan JJ, et al. A feedforward mechanism mediated by mechanosensitive ion channel PIEZO1 and tissue mechanics promotes glioma aggression. Neuron. 2018;100(4):799–815.

298. Bae C, Sachs F, Gottlieb PA. The mechanosensitive ion channel Piezo1 is inhibited by the peptide GsMTx4. Biochemistry. 2011;50(29):6295–300.

299. Kurata T, Rajendran V, Fan S, Ohta T, Numata M, Fushida S. NHE5 regulates growth factor signaling, integrin trafficking, and degradation in glioma cells. Clin Exp Metastasis. 2019;36:527–38.

300. Ko M, Makena MR, Schiapparelli P, Suarez-Meade P, Mekile AX, Lal B, et al. The endosomal pH regulator NHE9 is a driver of stemness in glioblastoma. PNAS Nexus. 2022;1(1):pgac013.

301. Pall AE, Juratli L, Guntur D, Bandyopadhyay K, Kondapalli KC. A gain of function paradox: Targeted therapy for glioblastoma associated with abnormal NHE9 expression. J Cell Mol Med. 2019;23(11):7859–72.

302. Luo L, Wang J, Ding D, Hasan MN, Yang SS, Lin SH, et al. Role of NKCC1 activity in glioma K+ homeostasis and cell growth: New insights with the bumetanide-derivative STS66. Front Physiol. 2020;11:911.

303. Blaesse P, Airaksinen MS, Rivera C, Kaila K. Cation-chloride cotransporters and neuronal function. Neuron. 2009;61(6):820–38.

304. Nieto FR, Cobos EJ, Tejada MÁ, Sánchez-Fernández C, González-Cano R, Cendán CM. Tetrodotoxin (TTX) as a therapeutic agent for pain. Mar Drugs. 2012 Feb;10(2):281–305.

305. Challapalli V, Tremont-Lukats IW, McNicol ED, Lau J, Carr DB. Systemic administration of local anesthetic agents to relieve neuropathic pain. Cochrane Database Syst Rev. 2019 Oct;2019(10).

306. Brinkrolf P, Hahnenkamp K. Systemic lidocaine in surgical procedures: effects beyond sodium channel blockade. Curr Opin Anaesthesiol. 2014 Aug;27(4):420–5.

307. McCann G. Pharmacological treatment of significant cardiac arrhythmias. Br J Sports Med. 2000 Oct;34(5):401–2.

308. Fairhurst C, Martin F, Watt I, Doran T, Bland M, Brackenbury WJ. Sodium channel-inhibiting drugs and cancer survival: protocol for a cohort study using the CPRD primary care database. BMJ Open. 2016 Sep;6(9):e011661–e011661.

309. Doty P, Rudd GD, Stoehr T, Thomas D. Lacosamide. Neurother J Am Soc Exp Neurother. 2007 Jan;4(1):145–8.

310. 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 Apr;9(4):413–24.

311. Black JA, Waxman SG. Phenytoin protects central axons in experimental autoimmune encephalomyelitis. J Neurol Sci. 2008 Nov;274(1–2):57–63.

312. Perucca E. A pharmacological and clinical review on topiramate, a new antiepileptic drug. Pharmacol Res. 1997 Apr;35(4):241–56.

313. Silke B, Goldhammer E, Sharma SK, Verma SP, Taylor SH. An exercise hemodynamic comparison of verapamil, diltiazem, and amlodipine in coronary artery disease. Cardiovasc Drugs Ther. 1990 Apr;4(2):457–63.

314. Rubaiy HN. The therapeutic agents that target ATP-sensitive potassium channels. Acta Pharm Zagreb Croat. 2016 Mar;66(1):23–34.

315. Sanguinetti MC, Bennett PB. Antiarrhythmic drug target choices and screening. Circ Res. 2003 Sep;93(6):491–9.

316. Ravens U, Poulet C, Wettwer E, Knaut M. Atrial selectivity of antiarrhythmic drugs. J Physiol. 2013 Sep;591(17):4087–97.

317. Michiels CF, Van Hove CE, Martinet W, De Meyer GRY, Fransen P. L-type Ca2+ channel blockers inhibit the window contraction of mouse aorta segments with high affinity. Eur J Pharmacol. 2014 Sep;738:170–8.

318. Lukasova M, Malaval C, Gille A, Kero J, Offermanns S. Nicotinic acid inhibits progression of atherosclerosis in mice through its receptor GPR109A expressed by immune cells. J Clin Invest. 2011 Mar;121(3):1163–73.

319. Ashcroft SJ, Ashcroft FM. Properties and functions of ATP-sensitive K-channels. Cell Signal. 1990;2(3):197–214.

320. Jang SH, Choi SY, Ryu PD, Lee SY. Anti-proliferative effect of Kv1.3 blockers in A549 human lung adenocarcinoma in vitro and in vivo. Eur J Pharmacol. 2011 Jan;651(1–3):26–32.

321. Borrelli F, Pagano E, Romano B, Panzera S, Maiello F, Coppola D, et al. Colon carcinogenesis is inhibited by the TRPM8 antagonist cannabigerol, a Cannabis-derived non-psychotropic cannabinoid. Carcinogenesis. 2014 Dec;35(12):2787–97.

322. Huang MH, Huang YM, Wu SN. The Inhibition by Oxaliplatin, a Platinum-Based Anti-Neoplastic Agent, of the Activity of Intermediate-Conductance Ca2+-Activated K+ Channels in Human Glioma Cells. Cell Physiol Biochem Int J Exp Cell Physiol Biochem Pharmacol. 2015;37(4):1390–406.

323. Djamgoz MBA, Onkal R. Persistent current blockers of voltage-gated sodium channels: a clinical opportunity for controlling metastatic disease. Recent Patents Anticancer Drug Discov. 2013 Jan;8(1):66–84.

324. Brackenbury WJ, Chioni AM, Diss JKJ, Djamgoz MBA. The neonatal splice variant of Nav1.5 potentiates in vitro invasive behaviour of MDA-MB-231 human breast cancer cells. Breast Cancer Res Treat. 2007 Jan;101(2):149–60.

325. Yavuz M, Şahin B, Baykal AT, Demircan T. Hydroquinidine displays a significant anti-carcinogenic activity in breast and ovarian cancer cells via inhibiting cell-cycle and stimulating apoptosis. Turk J Biol. 2023;47(1):44–60.

326. Dao Trong P, Jungwirth G, Unterberg A, Herold-Mende C, Warta R. The Antiepileptic Drug Oxcarbazepine Inhibits the Growth of Patient-Derived Isocitrate Dehydrogenase Mutant Glioma Stem-like Cells. Cells. 2023;12(8):1200.

327. Dong W, Fekete A, Chen X, Liu H, Beilhartz GL, Chen X, et al. A designer peptide against the EAG2–Kvβ2 potassium channel targets the interaction of cancer cells and neurons to treat glioblastoma. Nat Cancer. 2023;4(10):1418–36.

328. Mlayah-Bellalouna S, Aissaoui-Zid D, Chantome A, Jebali J, Souid S, Ayedi E, et al. Insights into the mechanisms governing P01 scorpion toxin effect against U87 glioblastoma cells oncogenesis. Front Pharmacol. 2023;14:1203247.

329. Dinevska M, Gazibegovic N, Morokoff AP, Kaye AH, Drummond KJ, Mantamadiotis T, et al. Inhibition of radiation and temozolomide-induced glioblastoma invadopodia activity using ion channel drugs. Cancers. 2020;12(10):2888.

330. Luo L, Wang J, Ding D, Hasan MN, Yang SS, Lin SH, et al. Role of NKCC1 activity in glioma K+ homeostasis and cell growth: New insights with the bumetanide-derivative STS66. Front Physiol. 2020;11:911.

331. Huang T, Xu T, Wang Y, Zhou Y, Yu D, Wang Z, et al. Cannabidiol inhibits human glioma by induction of lethal mitophagy through activating TRPV4. Autophagy. 2021;17(11):3592–606.