Open Access

Repurposing the antipsychotic drug penfluridol for cancer treatment (Review)

  • Authors:
    • Asma Ali Ibrahim Mze
    • Amirah Abdul Rahman
  • View Affiliations

  • Published online on: October 25, 2024     https://doi.org/10.3892/or.2024.8833
  • Article Number: 174
  • Copyright: © Ali Ibrahim Mze et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer is one of the most prevalent diseases and the leading cause of death worldwide. Despite the improved survival rates of cancer in recent years, the current available treatments often face resistance and side effects. Drug repurposing represents a cost‑effective and efficient alternative to cancer treatment. Recent studies revealed that penfluridol (PF), an antipsychotic drug, is a promising anticancer agent. In the present study, a scoping review was conducted to ascertain the anticancer properties of PF. For this, a literature search was performed using the Scopus, PubMed and Web of Science databases with the search string ‘penfluridol’ AND ‘cancer’. A total of 23 original articles with in vivo and/or in vitro studies on the effect of PF on cancer were included in the scoping review. The outcome of the analysis demonstrated the anticancer potential of PF. PF significantly inhibited cell proliferation, metastasis and invasion while inducing apoptosis and autophagy in vivo and across a spectrum of cancer cell lines, including breast, lung, pancreatic, glioblastoma, gallbladder, bladder, oesophageal, leukaemia and renal cancers. However, research on PF derivatives with high anticancer activities and reduced neurological side effects may be necessary.

Introduction

Cancer is one of the most prevalent diseases and the leading cause of death worldwide. The global cancer incidence and mortality were estimated at nearly 19 million new cases and 10 million deaths in 2020, with lung, colorectal, breast and prostate cancer accounting for the majority of the diagnosed cancers worldwide (1,2). Despite the increasing survival rates of cancer patients observed in the past decades due to improved and game-changing therapies, the global incidence of cancer keeps increasing and is estimated to reach 28 million per year in 2040, leading to a physical, emotional and financial burden to the patients and their families, and a strain on resources of the public health system (2,3). The cancer burden has been attributed to the inequitable access to cancer prevention, early detection, screening and treatment of the population, particularly in low- and middle-income countries. In addition, an increase in the price of cancer drugs has been recorded in recent years. Cancer is predicted to cost $25.2 trillion to the world economy from 2020 to 2050, bringing severe financial distress to the economy, affected patients and families, and the public health system (4,5). Furthermore, highly effective treatments, including radiotherapy, chemotherapy, surgery, targeted therapy and hormonal treatments, which significantly contribute to the overall reduction of cancer mortality, recurrence and spread and increase in survival rate, have limitations in curing cancer, particularly in patients with delayed diagnoses and in patients with cancer therapy resistance, resulting in a worse survival outcome (6,7).

Drug repositioning, an approach to using approved drugs to treat diseases outside their original indication scope, has gained significant attention in recent years (8). Repurposing existing drugs reduces the exorbitant cost of developing new drugs, estimated to be between $314 million and $2.8 billion per medication in 2018. It shortens the time taken for the drug approval process, which can otherwise take 10 to 15 years to reach the market (8,9). Furthermore, repurposed drugs have a decreased likelihood of clinical failure due to adverse effects, as their safety and dosing have already received clinical approval (10). Consequently, repurposing drugs for cancer treatment represents a cost-effective method for cancer therapy.

Various antipsychotic drugs including penfluridol (PF), phenothiazine, pimozide, chlorpromazine and thioridazine have been discovered to have anticancer properties and have been identified as a promising alternative cancer treatment (11). Several studies have reported that patients taking antipsychotic drugs have a lower cancer incidence compared to the general population (12). PF is a well-established first-generation diphenylbutylpiperidine (DPBP) antipsychotic drug used to treat chronic schizophrenia and other psychotic disorders, and has been found to inhibit cancer (11,13). Indeed, PF inhibits cancer cell proliferation and induces apoptosis and autophagy in various cancer cell lines (14). PF has a half-life of 70 h, can cross the blood-brain barrier (BBB) and block the dopamine receptor (DRD2) binding sites, making it a potent drug for the treatment of brain cancers, as well as cancers with high brain metastasis ability (13,15,16). This scoping review aimed to briefly investigate the anticancer properties of PF, focusing on in vivo and in vitro studies and providing a concise overview of PF's anticancer mechanism of action.

Methodology

Protocol

This scoping review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) checklist and PRISMA-ScR Tip sheets (17). The review was conducted in five stages according to Mak and Thomas's (18) steps for conducting a scoping review: i) Identify the research question; ii) identify relevant studies; iii) select the studies to be included in the review; iv) charting data; and v) collate, summarize and report results.

Identification of the research question

This review aimed to investigate the effect of PF on cancer. Following the standard recommendation for a broader research question (19), a primary research question was developed: What are the effects of PF on cancer? This research question provided sufficient literature to warrant the scoping review and envelop the studies on PF as a treatment for cancer. Hence, no further narrowing of the research question was necessary.

Identification of relevant studies

A literature search was conducted using the Scopus (https://www-scopus-com.uitm.idm.oclc.org), PubMed (https://pubmed.ncbi.nlm.nih.gov) and Web of Science (WoS; http://www-webofscience-com.uitm.idm.oclc.org) databases with the search string ‘penfluridol’ AND ‘cancer’. The search, conducted in November 2023, included no additional filters.

Study selection

Following the search, the identified records were assessed in two phases. In the first phase, one author (AAIM) evaluated the obtained articles by their titles and excluded duplicate articles, review papers and retracted papers. The same author screened the remaining journal papers based on their titles and abstracts. Any articles with no relation to the roles of PF in treating cancers according to their titles and abstracts and any conference abstracts were excluded. In the second phase, one author (AAIM) retrieved and assessed the remaining articles' full text. In the end, original research articles on the roles, functions, importance and mechanisms of PF in treating cancer written in English were retained and included in the scoping review. The second author (AAR) was consulted for their opinion on the obtained data throughout the study selection stages.

Mendeley Desktop version 1.19.8 (Elsevier) and Microsoft Excel version 16.89.1 (Microsoft Corporation) were used to sort out the literature and identify the articles' duplication by independently filtering out the articles' DOIs, titles and abstracts.

Charting the data

Microsoft Excel version 16.89.1 (Microsoft Corporation) was used to organize the data from the selected articles. Data extraction was performed following the PRISMA guidelines by (AAIM). The extracted data were categorised into author names, publication year, type of cancer, dose of PF, study design, significant findings, key findings and limitations.

Collating, summarizing and reporting of the results

The extracted data were tabulated in Microsoft Excel version 16.89.1 (Microsoft Corporation) with rows relating to the articles and columns that classify the variables and contain the relevant information. This table was used to classify and report the information collected.

Results

Article selection

The primary search yielded 144 articles, 34 from PubMed, 62 from Scopus and 48 from WoS. A total of 59 duplicates were removed, resulting in 85 records retained and screened further. Subsequently, a total of 24 articles were eliminated due to being reviews (n=22) and retracted papers (n=2). The remaining 61 articles were screened based on title and abstract and 24 articles with no relation to the roles of PF in treating cancers and poster abstracts (n=9) were excluded. A total of 28 records were identified as eligible for further full-text assessment. Of the 28 articles, five were excluded due to lack of availability (n=2), focusing on PF derivatives (n=2) and commentaries (n=1). Finally, 23 original research studies in English were retained for this scoping review. The PRISMA flowchart on the paper selection is presented in Fig. 1.

Study characteristics

The main findings are summarised and presented in Table I. Out of the 23 original articles included in this review, 4 evaluated the anticancer effect of PF on breast cancer, 4 on lung cancer, 4 on pancreatic cancer, 3 on glioblastoma, 1 on bladder cancer, 1 on gallbladder cancer, 1 on oesophageal cancer, 1 on leukaemia cancer and 1 on renal cancer. The remaining 3 articles included in this review studied the effect of PF on various types of cancer. All of the studies in this review included an in vitro model of a cell line supplemented with 0 to 40 µM PF, while most studies used an in vivo model treated with PF at 1 to 10 mg/kg.

Table I.

The effect of penfluridol on cancer.

Table I.

The effect of penfluridol on cancer.

Type of cancerAuthor(s), yearStudy modelDosesTreatment durationFindings(Refs.)
BreastHedrickSKBR3 &0.7–7.5 µM24 hPF inhibited cell growth and cell migration and(20)
et al 2017MDA-MB- induced cell apoptosis. PF induced ROS in breast
231 cells cancer cells by downregulating the expression of
Sp transcript factor (Sp1, Sp2 and Sp4) and cMyc
(miR-27a and miR-20/miR-17). PF repressed the
expression of α5-, α6-, β1 and β4-integrin.
Female10 mg/kg/19 daysPF decreased the tumour volume, inhibited tumour
athymicday growth and induced apoptosis. PF ROS-dependently
nude mice reduced the expression of Sp1, Sp3, Sp4 and Sp-
regulated genes (α6-, α5-, β1- and β4-integrins).
GuptaMCF-7,0-20 µM24-72 hPF treatment inhibited the cell growth and colony(21)
et al 2019MCF-7HH, formation of PXT-sensitive and PXT-resistant cells
MCF-7PR, in a concentration- and time-dependent manner. PF
4T1 & 4T1PR downregulated the expression of HER2, β-catenin,
cells c-Myc, TCF-1, TCF-4, p-GSK3β and cyclin D1. PF
induced apoptosis in breast cancer cells. A
combination of PXT and PF significantly reduced
the cancer cells' viability, suppressing the
chemoresistance markers while enhancing apoptosis.
Female10 mg/kg/23 daysPF significantly suppressed the growth of paclitaxel-
Balb/c miceday resistant cells alone or in combination with 5 mg/kg/3
days PXT. PF alone and PF combined with PXT
inhibited HER2 and β-catenin expression and
increased apoptosis.
RanjanMDA-MB-0-20 µmol/l24-72 hPF suppressed the proliferation, migration and(22)
et al 2016231, 4T1 & invasion of breast cancer. PF treatment inhibited
HCC1806 cells' survival and motility by reducing the
cells expression of integrin α6, integrin β4, FAK, paxillin,
Rac1/2/3 and ROCK1 and inducing apoptosis.
Female Balb/10 mg/kg27 daysPF suppressed tumour growth by inhibiting
c mice β4-integrin and enhancing apoptosis in mice.
Intracardiac or intracranial injection of breast cancer
cells revealed similar results.
SrivastavaHUVECs &0-5 µM24-48 hLow doses of PF blocked the angiogenesis-(23)
et al 2020MDA-MB-23 suppressed VEGF-induced primary endothelial cell
cells migration and tube formation of the cancer cells. PF
inhibited the Src and Akt signalling pathways.
C57B/L61 mg/kg11 daysLow dose of PF blocked VEGF- and FGF-induced
mice angiogenesis.
LungLai et alA549 &0-7.5 µM24 hPF inhibited lung tumour growth by reducing(24)
2022HCC827 mitochondrial ATP production and downregulating
cells NADH levels. PF mediated the suppression of
mitochondrial biogenesis, impeded the level
of PGC-1α protein and mRNA, SIRT1 and p53, and
induces AMPK activation.
NOD-SCID5 m/kg28 daysPF inhibited the growth and metastasis of lung
mice cancer in mice. These results were enhanced in
combination with 2DG. PF decreased the expression
of PGC-1α and SIRT1.
Xue et alA549, H446,0-20 µM24-72 hPF inhibited lung cancer cell growth and viability(26)
2020H1993, with G0/G1 cell cycle phase arrest by enhancing the
SPC-A1 & expression level of p21/p27 and decreasing the
LL2 cells expression levels of the cyclin-CDK complex. PF
induced apoptosis via the mitochondria-mediated
intrinsic apoptosis pathway and inhibited the
migration and invasion of lung cancer cells.
Female10 mg/kg26 daysPF inhibited tumour growth in an A549 cell
Balb/c xenograft mouse model. PF blocked the proliferation
nude mice and metastasis of lung cancer in mice by
regulating the AKT and MMP signalling pathways
while inducing apoptosis.
HungNSCLC,7.5–10 µM24-72 hPF suppressed cell proliferation by inducing ER(25)
et al 2019A549, stress- mediated autophagosome accumulation to
HCC827 & deplete ATP energy. PF inhibited cell migration.
BEAS-2B PF induced nonapoptotic cell death by blocking
cells autophagic flux and autophagosome formation
of LC3B-II protein in lung cancer cells.
NOD-SCID5-10 mg/kg35 daysPF inhibited the growth and the metastasis of lung
mice cancer in mice by inducing an accumulation of
autophagosome-related protein.
HungA549, H23,0-20 µM24-72 hPF reduced the migration, invasion and adhesion(27)
et al 2021HCC827, of LADC cells. PF inhibited MMP-12 by
PC9, H1975, downregulating the uPA/uPAR/TGF-β/Akt axis to
HMEC-1 & modulate the motility and adhesion of LADC cells.
BEAS-2B PF inhibited EMT byupregulating E-cadherin and
downregulating N-cadherin.
Male NOD-2.5 µM5 weeksPF suppressed lung metastasis and colony formation.
SCID mice PF increased the expression of E-cadherin levels and
decreased N-cadherin and MMP-12 levels in tumour
tissues.
PancreaticRanjanAsPC-1,0-10 µM24-72 hPF induced ER stress in pancreatic cancer cells(28)
et al 2017BxPC-3 & characterised by the upregulation of ER stress
Panc-1 cells markers of BIP, CHOP and IRE1α. PF-induced ER
stress led to autophagy.
Female10 mg/kg3 weeksPF-treated mice had a high level of BIP, CHOP
athymic and IRE1α expression in the tumour lysates and a
nude mice reduction of tumour mass.
Ranjan andBxPC-3 &0-20 µM24-72 hPF induced apoptosis and inhibited the growth of(29)
SrivastavaAsPC-1 pancreatic cancer cells. PF enhanced autophagy in
2016 pancreatic cancer cells mediated by apoptosis. PF
impeded the formation of lysosomes.
Female10 mg/kg27 & 59PF reduced the growth of BxPC-3 tumour xenografts
athymic daysand the development of orthotopically implanted
nude mice pancreatic tumours by 80% by inducing autophagy-
mediated apoptosis in the tumours.
DandawateAsPC-1,0-40 µM24-72 hPF highly bound to the JAK2 domain PRLR. PF(30)
et al 2020BxPC-3, suppresses pancreatic cancer cells' proliferation and
Panc-1, colony and spheroid formation. PF enhanced PDAC
MiaPaCa-2, & autophagy. The PF mechanism of inhibition of
UNKC-6141 PDAC cell growth did not proceed through DRD2.
Male C57BL/65 mg/kg28 & 35PF decreased the weight of the orthotopic tumours.
daysPF significantly inhibited the tumour weight and
mice & PDX- volume in xenograft tumours. PF reduced PDAC
carrying tumour growth by inducing autophagy.
NGS mice
ChienPanc0403,0-10 µM0-36 hPF inhibited pancreatic cancer cell proliferation and(31)
et al 2015SU8686, growth and induced apoptosis. PF decreased the
MiaPaCa2, phosphorylation levels of SRC, AKT and p70S6K.
Panc1, PF induced the activation of PP2A protein
Panc0504, phosphatase leading to pancreatic cancer cell death.
AsPc1,
Panc1005,
Panc0203,
Panc0327,
BxPc3 &
HPDE
GlioblastomaRanjan andGBM43,0-20 µM24-72 hPF reduced the survival rate and induced apoptosis(32)
SrivastavaGBM10, in glioblastoma cell lines. PF treatment suppressed
2017GBM44, the phosphorylation of Akt at Ser473 and decreased
GBM28, the expression of GLI1, OCT4, Nanog and Sox2.
GBM14 &
U251MG
Athymic nude10 mg/39-54PF treatment inhibited the growth and reduced the
micekg/day volume of days in vivo glioblastoma tumour models.
PF reduced pAkt, GL1 and OCT4 and enhanced
apoptosis.
Kim et alCSC2, X01,0-20 µM24-72 hPF suppressed the growth of GSCs in a dose- and(33)
20190315, 528NS, time- dependent manner. PF suppressed the stemness
83NS, U87MG of GSCs and inhibited their sphere-forming ability
& T98G cells and invasiveness. PF inhibited the EMT potential
in human GSCs.
Female Balb/c0.860 daysPF decreased invasion and tumour volume in
nude micemg/kg/ orthotopic xenografts by reducing GLI1, uPAR,
week SOX2 and vimentin expression. With a
combination of PF and TMZ, the tumour
disappeared in the mice.
RanjanFemale10 mg/kg40-48PF treatment suppressed glioblastoma tumour(34)
et al 2017athymic daysgrowth by reducing murine myeloid-derived
nude mice & suppressor cells (MDSC). PF enhanced splenic cell
female SCID- proliferation, increased M1 macrophages and
NOD mice suppressed T-regulatory cells and inflammation in
the tumour.
GallbladderHu et alEH-GB1,0-10 µM24-72 hPF inhibited the proliferation and migration of(35)
2022GBC-SD & gallbladder cancer (GBC) cells and enhanced
SGC-996 apoptosis. PF treatment caused the activation
cells of AMPK/PFKB3-mediated glycolysis in GBCs,
and a combination of PF and 2-DG or AMPK
inhibitor CC improved the anticancer effect of PF.
Old female10 mg/kg15 daysPF suppressed lung tumour growth. PF promoted
nude mice apoptosis and the activation of AMPK/PFKFB3
signaling.
Bladdervan derUCB &0-100 µM0-40 hPF inhibited the cell viability and clonogenicity(36)
Horst et alUM-UC- of the UCB cell line. PF induced lysosomal
20203luc2 cells leakage and destabilized lysosomal structures in
human UCB cells, resulting in the redistribution
of phosphatidylserine from the internal to the
external membrane surface, an early indicator
of apoptosis.
Female Balb/c130 µg/kg29 daysPF significantly decreased tumour growth,
nude miceper week invasion and metastasis in mice.
OesophagealZhengKYSE30,0-10 µmol/l24-72 hPF inhibited proliferation and colony formation(37)
et al 2020KYSE150 & and enhanced the apoptosis of the esophageal
KYDEE270 squamous cell carcinoma (ESCC) cells. The
inhibition of the cells was mediated by
AMPK/FOXO3a/BIM signalling. PF
inhibited glycolysis.
Female Balb/c9-18 mg/kg42 daysPF decreased tumour volume and cell
nude mice proliferation and induced apoptosis in tumour
xenograft mice. PF inhibited glycolysis.
LeukaemiaWu et alHL-60, U9377.5–20 µM24-72 hPF inhibited the proliferation of AML cells.(38)
2019& MV4-11 PF induced apoptosis by activating PP2A to
cell lines suppress Akt and MAPK activities. PF
with FLT3-WT triggered autophagic responses by inducing
or FLT3-ITD the elevation of intracellular ROS levels.
RenalTung786-O,0-25 µM24-72 hPF decreased the proliferation and colony(39)
et al 2022Caki-1, A498 formation of RCC cells. PF induced autophagy
& ACHN cells through the ER stress-mediated UPR, leading
to apoptosis. PF reduced stemness and sphere
formation in RCC cells.
NOD-scid5 µM5 weeksPF reduced the tumour volume and growth by
IL2Rγ suppressing the GLI1 and proliferation
(NSG) mice index Ki-67.
DiverseWu et alB16, LL/2,0-9 µmol/l24-72 hPF inhibited the proliferation of melanoma,(40)
2014CT26 & lung carcinoma, colon carcinoma and breast
4T1 cells cancer cells in a time-dependent manner. PF
increased the accumulation of unesterified
cholesterol in the cancer cells.
Female0.06–0.12 mg/45 daysPF suppressed lung, colon, melanoma and
C57BL/6week breast tumour growth, and tumour weight of
mice & the mice. PF decreased the total cholesterol
Balb/c mice in the tumour tissue of the mice.
VaraldaHCT116,10-160 µmol/l24-72 hPF inhibited the proliferation of colorectal(14)
et al 2020SW620, and breast cancer and glioblastoma cells.
MCF7, MDA- PF reduced MCF7 and HCT116 cell motility
MB-231, and migration. PF induced mitochondrial
U87 & U251 and lysosomal membrane alteration and
cells phospholipid aggregation in cancer cells. PF
suppressed the mTOR pathway, induced
by AMPK activation and autophagy.
Du et alHeLa5 µmol/l24 hPF has high radiosensitivity activity. PF(41)
2018 inhibited the repair of DSBs post-IR in HeLa
cells by reducing NHEJ repair and preventing
the activation of DNA-PKcs.

[i] AML, acute myeloid leukaemia; AMPK, AMP-activated protein kinase; AKT, serine/threonine-specific protein kinase 1; ATP, adenosine triphosphate; BAX, BCL-2-associated X; BCL-2, B-cell leukemia/lymphoma 2; BIM, BCL-2-interacting mediator; BIP, binding protein; CC, compound C; CCL4, C-C motif chemokine ligand 4; CHOP, C/EBP homologous protein; CNS, central nervous system; DPBP, diphenylbutylpiperidine; EMT, epithelial-mesenchymal transition; ER, endoplasmic reticulum; ESCC, esophageal squamous cell carcinoma; FAK, focal adhesion kinase; GBC, gallbladder cancer; GLI1, glioma-associated oncogene homolog 1; GSC, glioma-sphere forming cells; GSH, glutathione; GSK, glycogen synthase kinase; IR, ionizing radiation; IRE1α, inositol requiring 1α; KRT, keratine; LADC, lung adenocarcinoma; LC3, light chain; LLC, Lewis lung carcinoma; MAPK, mitogen-activated protein kinase; MDSC, myeloid-derived suppressor cells; miR, microRNA; MMP, matrix metalloproteinase; mTOR, mammalian target of rapamycin; NHEJ, non-homologous end joining; NSCLC, non-small-cell lung cancer; OCTA, optical coherence tomography angiography; OXPHOS, oxidative phosphorylation; PARP, poly-ADP ribose polymerase; PDAC, pancreatic ductal adenocarcinoma; PF, penfluridol; PFKFB3, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3; PP2A, protein phosphatase 2A; PRLR, prolactin receptor; PTX, paclitaxel; PUMA, p53-upregulated modulator of apoptosis; RCC, renal cell carcinoma; ROCK1, Rho-associated coiled-coil containing protein kinase 1; ROS, reactive oxygen species; Sp, specificity protein; TCF, transcription factor; UCB, urothelial carcinoma of the bladder; UPR, unfolded protein response; uPAR, urokinase plasminogen activator receptors; VEGF, vascular endothelial growth factor; 2-DG, 2-deoxy-D-glucose.

Synthesis of the results
Breast cancer

PF was found to exert its anticancer properties on breast cancer through multiple pathways. PF inhibits cell proliferation in breast cancer cells (2023). For instance, in the relevant studies, PF successfully reduced the tumour volume and weight in the in vivo model of breast cancer. Furthermore, PF was found to reduce cell migration through reactive oxygen species (ROS) (20), the integrin pathway (22) and the VEGF pathway (23). In addition, PF was capable of inducing apoptosis in breast cancer cells (2022). While Hedrick et al (20) imparted that the anticancer properties of PF in breast cancer are ROS-dependent, Gupta et al (21) revealed that PF downregulates the expression of chemoresistance markers in paclitaxel (PXT)-resistant breast cancer, leading to apoptosis. In fact, cotreatment of PF with glutathione attenuated the effect of PF on breast cancer (20), and the combination of PF and PXT decreased the expression of human EGFR 2 (HER2), β-catenin and cyclin D, simultaneously enhancing apoptosis (21). It is worth mentioning that chronic administration of PF failed to elicit any significant toxic or behavioural side effects in mice (22) and that low concentrations of PF can block angiogenesis in vitro and in vivo (23).

Lung cancer

Studies on PF in the treatment of lung cancer revealed that PF inhibits lung tumour growth by inducing mitochondrial ATP energy loss in vivo and in vitro (24). Similar results were obtained pertaining to autophagy (25). PF inhibited lung cancer proliferation, migration and metastasis in vivo and in vitro by regulating the AKT and MMP signalling pathway (26,27). PF suppressed MMPs and epithelial-mesenchymal transition (EMT) required for cancer cell motility and adhesion (27). Induced G0/G1 phase arrest, ROS and endoplasmic reticulum (ER) stress, as well as loss of the mitochondrial membrane potential (ΔΨm), were noted in PF-treated lung cancer (25,26). On the other hand, PF in combination with glycolysis inhibitor 2-deoxy-D-glucose synergistically enhanced the inhibitory effect of PF on lung cancer cell growth and the total amount of mitochondria (24). No side effects were noted in the mice used in these studies.

Pancreatic cancer

In pancreatic cancer, PF was reported to induce autophagy (2830) and apoptosis (29,31) in in vitro and in vivo models. PF inhibited the proliferation and colony formation of pancreatic cancer cells (2831) through the suppression of prolactin receptor (PRLR) signalling (30) and the activation of protein phosphatase 2A (PP2A) protein phosphatase (31). A study by Ranjan & Srivastava (29) revealed that PF induced autophagy-mediated apoptosis in pancreatic cancer. However, while PF exerts its antipsychotic activity by blocking the DRD2 receptors, the antiproliferative activity of PF in pancreatic ductal adenocarcinoma is not related to DRD2 and does not trigger protein degradation (30).

Glioblastoma

In glioblastoma, PF appeared to reduce the growth of glioblastoma cells and a tumour model through the apoptosis pathway (32). In addition, GLA1 was downregulated in a glioblastoma cancer model treated with PF (32,33). On the other hand, Ranjan et al (34) showed that PF treatment suppresses glioblastoma growth through immune regulation, while Kim et al (33) denoted that it is by reducing the sphere formation, stemness and invasiveness of the cells. Of note, a combination of PF and temozolomide (TMZ) produced maximal antiproliferative and antitumor effects in in vivo and in vitro models (33).

Other cancers

PF inhibited the cell proliferation and colony formation of esophageal squamous cell carcinoma (ESCC) cells (37), renal cell carcinoma (RCC) cell lines (39), acute myeloid leukaemia (AML) cells (38), gallbladder cancer (35), bladder cancer (36), colorectal and breast cancer, glioblastomas, lung cancer and melanoma cells (14,40). For instance, PF reduces glycolysis and promotes cell apoptosis (35,36). Apoptosis was induced in cancer cells through repressed glycolysis (37) and activation of PP2A (38) in ESCC and AML cells, respectively. Combining 2-deoxy-D-glucose (2-DG) or AMP-activated protein kinase (AMPK) inhibitor Compound C (CC) significantly improved PF's anticancer effect in gallbladder cancer (35). Furthermore, PF enhanced autophagy in cancer cells by upregulating ER stress and ROS (38,39). It is worth highlighting that the anticancer activities of PF on RCC partially occurred through DRD2 (39) and that PF suppresses the mTOR pathway in various cell lines (14). Besides that, in vivo studies uncovered that PF effectively prolongs the survival rate of mice bearing lung tumours and enhances the decrease of total cholesterol in tumour tissues with no significant difference in the serum cholesterol levels of the PF-treated mice (40). No side effects to the vital organs of the PF-treated mice were noted despite the decrease in tumour growth and volume in vivo (37,39). In addition, PF-treated urothelial carcinoma of the bladder (UCB) orthotopic xenograft mice exhibited normal murine urothelium, epithelial tissue integrity, proliferation index (proliferating cell nuclear antigen), viable epithelial cell numbers and fragmented keratin (36). In the meantime, extensive research by Du et al (41) unveiled that PF has high radiosensitizing activity and can suppress the activity of non-homologous end joining (NHEJ).

Discussion

PF

PF {4-(4-chloro-α,α,α,-trifluoro-m-tolyl)-1-[4,4-bis-(p-fluorophenyl)-butyl]-4-piperidinol} is a DPBP, first-generation antipsychotic drug used to treat chronic schizophrenia, Tourette's syndrome, acute psychosis and other psychotic disorders discovered by Janssen Pharmaceutical in 1968 (15,42,43). PF is a long-lasting neuroleptics drug orally administered once a week with a half-life of 66 h and a terminal plasma half-life of 199 h (44). Indeed, PF is highly lipophilic and is deposited in the adipose tissue after absorption in the gastrointestinal tract, which heightens its prolonged duration of action, since PF is gradually released from these reservoirs (15,44). PF is excreted via urine and faeces (45). The maximal weekly dose of PF treatment for patients with chronic schizophrenia varies between 60 and 140 mg (46,47). According to Andrade (48), long half-life antipsychotic drugs significantly reduce the withdrawal or discontinuation syndrome of the drug but can be disadvantageous in case of toxicity or side effects. For instance, PF treatment fosters various adverse effects, including depression, agitation, drowsiness, tachycardia, hypotension, insomnia and neuroleptic malignant syndrome (49). Therefore, a low dose of PF treatment is recommended.

PF is capable of penetrating the BBB, making it a promising option for the treatment of brain-metastasized cancers (33). Airoldi et al (43) revealed that despite the distribution of PF in the adipose tissue being higher, PF remains concentrated in the brain with the highest concentration recorded in the hemisphere part of the brain 1 h after the injection and the mesencephalon 24 h after the injection of 0.5 mg/kg PF in rats. PF acts by blocking the dopamine receptor, particularly DRD2, where it binds with a high affinity, leading to the inhibition of dopaminergic neurotransmission (13,15,16). A former study by Enyeart et al (50) revealed that the PF also blocks the T-type and L-type calcium channels at low concentrations. Additionally, Vlachos et al (12) have reported that patients taking antipsychotic drugs have a lower cancer incidence compared to the general population (12). Henceforth, examining the anticancer properties of PF may offer valuable insight. Fig. 2 summarises the anticancer activities of PF.

The anticancer properties of PF

PF was found to inhibit cell proliferation in a dose- and time-dependent manner in all the cancer cells reported in this review (14,2041). For instance, <10 µmol/l of PF induced 50% cell death in various cancer cell lines (14), while <2.5 µM of PF inhibited the proliferation and colony formation of lung and renal cancer cells (25,39). Meanwhile, 0.12 mg/week of PF remarkably prolonged the survival time in the LL/2 in vivo cancer model (40), indicating that a low concentration of PF can provide anticancer properties. The antiproliferative effects of PF are not mediated by major neuroreceptor systems (14), but instead by interfering with the mechanism involved in cancer growth. Indeed, PF induced cell death by downregulating the cell growth proteins (cyclin D and MYC) and expression of the G2-M phase of cell cycle arrest protein cyclin B1 and p21 in pancreatic cells (31). The regulatory protein of the transition between the G2 to M phase has a significant role in the proliferation of the tumour and inhibition of these proteins led to cell-cycle arrest and eventually apoptosis (51,52). In addition, PF activates PP2A, mediating apoptosis and cell death (31,38). PP2A is a positive regulator of apoptosis and activation of PP2A dephosphosphorylates and inactivates anti-apoptotic Bcl-2, inducing apoptosis (53).

On the other hand, PF increased ROS levels in lung cancer cells, leading to cell death (26). ROS functions as a double-edged sword in cancer cells and can act as a tumour-promoting or suppressing agent (54,55). Cancer cells possess elevated levels of ROS in comparison to normal cells due to their fast proliferation and alteration in cellular metabolism, particularly the reprogrammed metabolism to aerobic glycolysis (56,57). Moderate levels of ROS are necessary for cancer cell proliferation, invasion, migration and angiogenesis, while excessively elevated ROS increase cancer cell damage and death (58,59). Indeed, PF induced ROS production triggering autophagy in AML cells (38). Furthermore, Hedrick et al (20) revealed that PF enhances ROS production, leading to the downregulation of cMyc expression and decreased expression of Sp1, Sp3, Sp4 and Sp-regulated genes in vitro and in an in vivo model of breast cancer, hence inhibiting cell viability and inducing apoptosis. The specificity proteins Sp and the multifaceted oncogene c-Myc are essential regulators of cellular proliferation, metabolism, metastasis and apoptosis, which, when upregulated, conduce to the progression of cancer (6063). Furthermore, Sp regulates the activation or repression of integrins, transmembrane receptors which partake in cancer development, progression, invasion, metastasis and resistance to therapy (6467). Ranjan et al (22) showed that PF reduced the expression of α6, α4, αv, β4, β1 and β3 integrins, as well as the downstream effector molecules of integrin signalling, including focal adhesion kinase (FAK), phosphorylated paxillin and paxillin in breast cancer (22). Desgrosellier & Cheresh (68) revealed that integrin signalling plays a significant role in cancer migration, proliferation and invasion and inhibition of integrins lead to the repression of tumour invasion and growth.

Apart from that, PF enhances energy loss. For instance, according to Lai et al (24), PF inhibits non-small cell lung cancer (NSCLC) cell growth by increasing ATP energy loss through the suppression of mitochondrial ATP production activity and biogenesis. Similarly, suppression of ATP increased the formation and accumulation of autophagosomes in NSCLC, leading to death (25). Indeed, cancer cells require a considerable energy supply for their metabolism (69). This phenomenon known as the Warburg effect is marked by an elevated glycolysis in the cancer cells, including aerobic glycolysis to meet the energy needs of the cells to grow (70). Several studies have shown that targeting the metabolism of cancer cells impacts the growth of the cancer and inhibition of the glycolysis reduced the biosynthesis of ATP, a constituent of DNA, RNA and phospholipids (71), thus suppressing cancer cell growth. PF inhibited ESCC cell proliferation by repressing glycolysis through AMPK signalling (37). The activation of AMPK/forkhead box (FOX)O3a/BCL-2-interacting mediator (BIM) signalling in an oesophageal cell line induced apoptosis (37). In addition, Hu et al (35) reported that the combination of glycolytic inhibitor 2-DG or AMPK inhibitor CC with PF treatment significantly inhibited the proliferation of gallbladder cancer. Furthermore, PF dysregulated cholesterol homeostasis by enhancing the accumulation of unesterified cholesterol in LL/2, 4T1, B16 and CT26 cancer cells and decreased the total cholesterol in tumour tissues of PF-treated mice (40). Cholesterol plays a significant role in tumour cell growth (72). Henceforth, PF has potential anti-metabolic properties for the treatment of cancer.

Apoptosis and autophagy, programmed cell death pathways crucial in maintaining cellular and organismal homeostasis, are the main mechanisms researched in cancer (73,74). PF was found to enhance apoptosis by inducing nuclear fragmentation, poly-ADP ribose polymerase cleavage, caspase-3 activation and condensed nuclear chromatin (21,29,38). Similarly, PF upregulated the expression levels of the proapoptotic proteins BIM, BAX and p53-upregulated modulator of apoptosis (PUMA) and downregulated the antiapoptotic gene Bcl-2 in pancreatic cancer (31). Then again, PF induced G0/G1 phase arrest and, vehemently, cell apoptosis in lung cancer via the mitochondrial apoptotic pathway (26). For instance, Xue et al (26) reported that PF increased the loss of ΔΨm. Thus, PF induces tumour suppression through apoptosis. Despite that, Hung et al (25) remarked that PF's antiproliferative effect on NSCLC cells is independent of apoptosis. In fact, instead of the apoptotic characteristics, formation of autophagosome protein light chain (LC)3B-II and expression of autophagosome markers autophagy-related protein 5, Beclin-1 and p62 were recorded in PF-treated NSCLC cells, implying that PF enhances cell death through autophagy (25,73,75,76). Indeed, PF promoted an increase in LC3-II and p62 protein expression, phospholipid aggregates and lysosomal membrane damage and consequently cell death in various cell lines (14,29,30). In addition, in in vivo and in vitro studies of PF-treated lung and pancreatic cancer, an increase in ER stress was observed, which stimulated autophagy, noticeable by the overexpression of binding protein, C/EBP homologous protein (CHOP) and inositol-requiring 1α, increased unfolded protein response (UPR) signals and activation of p38/MAPK (25,28). Pre-treatment of the pancreatic cells with pharmacological inhibitors such as sodium phenylbutyrate and mithramycin or silencing of CHOP, followed by PF treatment, considerably suppressed autophagy, revealing that PF-induced enhancement of ER stress leads to autophagy in pancreatic cancer (28). Of note, blocking autophagy with pharmacological inhibitors significantly increases PF-induced apoptosis in AML cells (38). However, treatment of BxPC-3 and AsPC-1 with autophagy inhibitors and LC3B silencing followed by PF treatment resulted in a significant reduction of apoptosis, suggesting that PF induces autophagy-mediated apoptosis in pancreatic cancer (29). Similar results were obtained in RCC cell lines, where PF induced autophagy-mediated apoptosis through the upregulation of ER stress-mediated UPR (39). Therefore, PF enhances autophagy and autophagy-mediated apoptosis in cancer.

In addition, PF constrained migration, adhesion, invasion and metastasis in cancer cells (27,36). Metastasis, a hallmark of cancer-cell migration from their initial site, is a crucial factor of cancer therapy failure associated with an unfavourable prognosis and cancer-associated death (77). PF treatment significantly reduced cell migration and invasion by reducing the expression of phosphorylated Rac family small GTPase 1 (Rac1), Rac1/2/3 and Rho-associated coiled-coil containing protein kinase 1 proteins in triple-negative breast cancer cells (22). Furthermore, Kim et al (33) revealed that PF inhibited stemness by reducing the expression of SOX2, Nestin and optical coherence tomography angiography and invasiveness by decreasing the expression of integrin α6 and urokinase plasminogen activator receptor in glioma-sphere forming cells. In addition, PF reduced the potential for EMT, associated with cancer migration, invasion, stemness and metastasis, of glioblastoma cancer models by downregulating the expression of EMT markers (vimentin, Zeb 1, N-cadherin, Snail and Slug) (33,78,79). Besides that, PF reduced the expression of glioma-associated oncogene homolog 1 and OCT4 in several glioblastoma cell lines (32) and reduced lung cancer migration in vivo and in vitro by blocking the FAK-related migration signalling pathway and regulating the AKT and MMP signalling pathway (26). Despite PF's ability to constrain the mobility of bladder cancer cells, no notable effects were registered in the urothelium and epithelial tissue of UCB orthotopic xenograft mice (36). Therefore, PF exhibits anti-migration and anti-metastasis properties without any side effects.

PF significantly inhibits angiogenesis, a mechanism by which tumours acquire new nutrients for cell metabolism, leading to cancer growth (80). Research by Srivastava et al (23) revealed that PF inhibits angiogenesis in in vitro and in vivo models of triple-negative breast cancer by suppressing VEGF-induced Src and Akt activation to prevent VEGF activity (23). VEGF is a major angiogenic agent in tumours that can initiate the growth and metastasis of tumours (81). Furthermore, PF increased the immune surveillance in glioblastoma cancer (33). Indeed, PF reduced the number of regulatory T cells (Treg), suppressed the expression of FoxP3 and CD4 by Tregs and increased M1 macrophages (increase of CD86 and IL-12 expression), but also inhibited tumour inflammation markers [C-C motif chemokine ligand 4 (CCL4) and IFNγ] responsible for tumour progression, as observed in PF-treated mouse glioblastoma cancer model (33,34). CCL4 has been identified as a marker of tumour proliferation in RCC and is associated with immune checkpoint genes, including lymphocyte activating 3, cytotoxic T-lymphocyte-associated protein 4 and programmed cell death protein 1 in RCC (82). Hence, the reduction of CCL4 in glioblastoma by PF indicates that PF suppresses the progression and immunity of the cancer cells. Therefore, PF can impede angiogenesis in tumours and modulate the immune system.

Lastly, PF considerably downregulated the expression of the chemoresistance markers HER2, β-catenin, cyclin D1 and c-Myc in PXT-resistant breast cancer (21). Indeed, chemoresistance promoted cancer relapses and metastasis, thus leading to a more aggressive form of cancer (83,84). Gupta et al (21) revealed that PF reduced the tumour volume in PXT-resistant breast cancer female Balb/c mice by 40%, but the combination of PF and PXT induced apoptosis and decreased the expression of HER2, β-catenin and cyclin D1. Moreover, Du et al (41) unveiled that PF has radiosensitizing activity and can suppress the NHEJ activity/repair of the ionizing radiation (IR)-induced DNA double-strand breaks (DSBs). Radiosensitizers heighten the lethal effect of radiotherapy on tumours by boosting DNA damage and free radical production without affecting normal tissue (85) and PF impaired the IR-induced DNA damage repair, enhanced the formation of DSB markers (γ-H2AX and tumour protein 53 binding protein 1 foci) and inhibited the repair of DSB post-IR in HeLa cells, thus promoting the cytotoxicity of IR in HeLa cells (41). Taken together, it can be suggested that PF is likely an excellent chemo-radiotherapeutic agent.

Overall, PF exhibits several anticancer effects in various cancer cell lines. PF affects the proliferation of cancer cells, provides radiosensitizing activity, induces apoptosis and autophagy, inhibits the invasion, metastasis and angiogenesis of cancer, and reduces the immunosuppressive effect of cancer cells.

PF treatment induced apoptosis via the mitochondrial apoptotic pathway in lung cancer and via the suppression of Akt in glioblastoma. In addition, PF instigated apoptosis through nuclear fragmentation, PARP cleavage and caspase 3 activation in leukaemia and through the activation of the BIM, BAX and PUMA proapoptotic proteins in pancreatic cancer. PF inhibited cell proliferation in lung cancer via mitochondrial ATP energy loss and the inhibition of glycolysis, which consequently led to autophagy and cell death. Similarly, in gallbladder, bladder and oesophageal cancer, PF promoted apoptosis via inhibition of glycolysis. However, in pancreatic cancer, PF induced autophagy via induction of ER stress but suppressed the mTOR pathway in numerous cancer cell lines. Upregulation of ROS by PF has been associated with the ability of PF to enhance autophagy in renal cancer and leukaemia. However, ROS led to the downregulation of cMyc and Sp resulting in apoptosis in breast cancer. PF inhibited cell migration via integrin and the VEGF pathway in breast cancer. PF induced cell death via the activation of PP2A and PRLR signalling in pancreatic cancer but PP2A in leukaemia cells. PF modulated the immune system in glioblastoma cells by reducing the number of Tregs and increasing M1 macrophages. In HeLa cells, PF was able to reduce DNA repair by inhibiting the activity of NHEJ.

Limitation of PF and future direction

The dosage of PF as an anticancer drug exceeds the clinical therapeutic range of PF as an antipsychotic. Indeed, the 10 mg/kg dose treatment of PF regularly used in in vivo studies, including in chronic treatment, has successfully reduced the tumour size and weight without triggering any major side effects, as no significant changes in the body weight of the mice and clinical chemical blood analysis parameters were observed (29), and this 10 mg/kg dose of PF is equivalent to the dose of 0.83 mg/kg in humans. Thus, for a person weighing 60 kg, the human equivalent dose of PF would be ~50 mg (21). However, most of the available studies indicate that mice are given PF treatment daily, which exceeds the maximum weekly dose of PF for chronic schizophrenia patients, which varies between 60 and 140 mg (46,47). Of note, this high dose may cause various adverse effects (49). In fact, since PF interacts extensively with most G-protein coupled receptors at levels consistent with the recommended anticancer dosage, which is 50 mg per day for humans, the off-target central nervous system (CNS) activity of PF as an anticancer drug will exacerbate the drug's neurological side effects (86). Therefore, studying other mechanisms of PF delivery to targeted sites, including the use of nanotechnology, could potentially reduce the CNS side effects of the drug.

Nonetheless, a study by Kim et al (33) on an orthotopic mouse glioblastoma model revealed that a lower dose of 0.8 mg/week of PF, which is equivalent to 4 mg/week in an adult patient with a body weight of 60 kg, showed effective antitumor activity. In this study, PF significantly reduced the proliferation, invasion, migration and stemness of glioblastoma cells, as well as the tumour size and invasion in vivo, indicating that a lower dose of PF has a potent anticancer effect (33). Similarly, 0.12 mg/week of PF resulted in significant inhibition of the tumour and prolonged the survival time in an LL/2 lung cancer in vivo model (40). Furthermore, Kim et al (33) demonstrated that a combination of 0.8 mg/kg of PF with TMZ provides better anticancer properties and prolonged survival in the mouse model. Hence, a lower dose of PF alone or in combination with other chemotherapeutic drugs may be considered for future clinical and in vivo studies.

In addition, Ashraf-Uz-Zaman et al (86,87) suggested the usage of PF derivatives with enhanced anticancer activity and a weakened antipsychotic effect. Their study identified 2 PF analogue compounds with less affinity for the CNS and greater anticancer properties than PF (87) (Fig. 3). These compounds, which harbour modifications of PF in the spacer linker by the addition of one carbon in the chain and the introduction of a functional methoxy group (8c), displayed no toxicity in mice and successfully inhibited the cell proliferation of the metastatic triple-negative breast cancer cell line MDA-MB-231 and the lung carcinoma cell line LLC (87). A further study by Ashraf-Uz-Zaman et al (86) identified other PF derivatives with the ability to induce apoptosis in the MDA-MB-231 cell line without affecting the cell cycle. Nonetheless, these compounds have yet to be tested and compared with PF in various cancers. Therefore, more thorough research needs to be done on the PF and PF derivatives to validate them as a clinical therapeutic option for cancer.

Conclusion

This scoping review provides an overview of the anticancer properties of PF in various cancers. It is crucial to investigate potential cost- and time-effective drugs for managing cancer. Based on the available studies, repurposing PF for cancer treatment has promising effects, particularly in cancer with low survival rates and high resistance to the current treatment. PF inhibited the growth, metastasis and migration of various cancers, including glioblastoma, as well as breast, lung, pancreatic, renal, bladder and oesophageal cancer in vitro and in vivo through distinctive mechanisms. Less than 10 µM of PF can provide anticancer activity in vitro. Nonetheless, the in vivo dose of PF exceeds the dose required for antipsychotic treatment; therefore, the subsequent development of PF derivatives with elevated anticancer and reduced antipsychotic activities was deemed necessary. In addition, despite the approval of PF as a valuable anticancer agent, dose-related side effects in the clinical setting and in other types of cancer have yet to be studied. Henceforth, extensive research on the effect of PF on other cancers and in patients needs to be performed. In addition, this scoping review focused on original articles published and indexed in PubMed, Scopus and WoS before November 2023 in English. Therefore, non-indexed, grey literature, studies in other languages and review papers were overlooked. Furthermore, only articles focusing on PF were considered and no clinical studies were included in this review. Thus, future scoping reviews on the anticancer properties of PF should include PF derivatives and clinical studies when available.

Acknowledgements

Not applicable.

Funding

The present study was supported by a grant from the Ministry of Higher Education Malaysia (grant no. FRGS/1/2023/SKK10/UITM/02/1).

Availability of data and materials

Not applicable.

Authors' contributions

AAIM contributed to the conceptualization, methodology, validation, formal analysis, data collection, visualisation, writing of the original draft and reviewing and editing. AAR contributed to the conceptualization of the study, reviewed and edited the manuscript as well as supervised the preparation of and validated the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Authors' information

Asma Ali Ibrahim Mze (ORCID no. 0009-0002-1848-6160); Amirah Abdul Rahman (ORCID no. 0000-0002-3566-1787).

References

1 

Ferlay J, Colombet M, Soerjomataram I, Parkin DM, Piñeros M, Znaor A and Bray F: Cancer statistics for the year 2020: An overview. Int J Cancer. Apr 5–2021.(Epub ahead of print). View Article : Google Scholar

2 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

3 

World Health Organization (WHO), . Cancer. WHO; Geneva: 2022

4 

Chen S, Cao Z, Prettner K, Kuhn M, Yang J, Jiao L, Wang Z, Li W, Geldsetzer P, Bärnighausen T, et al: Estimates and projections of the global economic cost of 29 cancers in 204 countries and territories from 2020 to 2050. JAMA Oncol. 9:465–472. 2023. View Article : Google Scholar : PubMed/NCBI

5 

Gordon N, Stemmer SM, Greenberg D and Goldstein DA: Trajectories of injectable cancer drug costs after launch in the United States. J Clin Oncol. 36:319–325. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Hendouei N, Saghafi F, Shadfar F and Hosseinimehr SJ: Molecular mechanisms of anti-psychotic drugs for improvement of cancer treatment. Eur J Pharmacol. 856:1724022019. View Article : Google Scholar : PubMed/NCBI

7 

Qu LG, Brand NR, Chao A and Ilbawi AM: Interventions addressing barriers to delayed cancer diagnosis in low- and middle-income countries: A systematic review. Oncologist. 25:e1382–e1395. 2020. View Article : Google Scholar : PubMed/NCBI

8 

Masuda T, Tsuruda Y, Matsumoto Y, Uchida H, Nakayama KI and Mimori K: Drug repositioning in cancer: The current situation in Japan. Cancer Sci. 111:1039–1046. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Wouters OJ, McKee M and Luyten J: Estimated Research and Development Investment Needed to Bring a New Medicine to Market, 2009–2018. JAMA. 323:844–853. 2020. View Article : Google Scholar : PubMed/NCBI

10 

Low ZY, Farouk IA and Lal SK: Drug Repositioning: New approaches and future prospects for life-debilitating diseases and the COVID-19 pandemic outbreak. Viruses. 12:10582020. View Article : Google Scholar : PubMed/NCBI

11 

Brown JS: Treatment of cancer with antipsychotic medications: Pushing the boundaries of schizophrenia and cancer. Neurosci Biobehav Rev. 141:1048092022. View Article : Google Scholar : PubMed/NCBI

12 

Vlachos N, Lampros M, Voulgaris S and Alexiou GA: Repurposing antipsychotics for cancer treatment. Biomedicines. 9:17852021. View Article : Google Scholar : PubMed/NCBI

13 

Shaw V, Srivastava S and Srivastava SK: Repurposing antipsychotics of the diphenylbutylpiperidine class for cancer therapy. Semin Cancer Biol. 68:75–83. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Varalda M, Antona A, Bettio V, Vachamaram A, Yellenki V, Massarotti A, Baldanzi G and Capello D: Psychotropic drugs show anticancer activity by disrupting mitochondrial and lysosomal function. Front Oncol. 10:5621962020. View Article : Google Scholar : PubMed/NCBI

15 

Soares BG and Lima MS: Penfluridol for schizophrenia. Cochrane Database Syst Rev. 2006:CD0029232006.PubMed/NCBI

16 

Chokhawala K and Lee S: Antipsychotic medications. StatPearls [Internet]. StatPearls Publishing; Treasure Island, FL: 2023

17 

Tricco AC, Lillie E, Zarin W, O'Brien KK, Colquhoun H, Levac D, Moher D, Peters MDJ, Horsley T, Weeks L, et al: PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. Ann Intern Med. 169:467–473. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Mak S and Thomas A: Steps for conducting a scoping review. J Grad Med Educ. 14:565–567. 2022. View Article : Google Scholar : PubMed/NCBI

19 

Arksey H and O'Malley L: Scoping studies: Towards a methodological framework. Int J Soc Res Methodol. 8:19–32. 2005. View Article : Google Scholar

20 

Hedrick E, Li XX and Safe S: Penfluridol represses integrin expression in breast cancer through induction of reactive oxygen species and downregulation of Sp transcription factors. Mol Cancer Ther. 16:205–216. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Gupta N, Gupta P and Srivastava S: Penfluridol overcomes paclitaxel resistance in metastatic breast cancer. Sci Rep. 9:50662019. View Article : Google Scholar : PubMed/NCBI

22 

Ranjan A, Gupta P and Srivastava SK: Penfluridol: An antipsychotic agent suppresses metastatic tumor growth in triple-negative breast cancer by inhibiting integrin signaling axis. Cancer Res. 76:877–890. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Srivastava S, Zahra FT, Gupta N, Tullar PE, Srivastava SK and Mikelis CM: Low Dose of Penfluridol Inhibits VEGF-Induced Angiogenesis. Int J Mol Sci. 21:7552020. View Article : Google Scholar : PubMed/NCBI

24 

Lai TC, Lee YL, Lee WJ, Hung WY, Cheng GZ, Chen JQ, Hsiao M, Chien MH and Chang JH: Synergistic tumor inhibition via energy elimination by repurposing penfluridol and 2-Deoxy-D-Glucose in lung cancer. Cancers (Basel). 14:27502022. View Article : Google Scholar : PubMed/NCBI

25 

Hung WY, Chang JH, Cheng Y, Cheng GZ, Huang HC, Hsiao M, Chung CL, Lee WJ and Chien MH: Autophagosome accumulation-mediated ATP energy deprivation induced by penfluridol triggers nonapoptotic cell death of lung cancer via activating unfolded protein response. Cell Death Dis. 10:5382019. View Article : Google Scholar : PubMed/NCBI

26 

Xue Q, Liu Z, Feng Z, Xu Y, Zuo W, Wang Q, Gao T, Zeng J, Hu X, Jia F, et al: Penfluridol: An antipsychotic agent suppresses lung cancer cell growth and metastasis by inducing G0/G1 arrest and apoptosis. Biomed Pharmacother. 121:1095982020. View Article : Google Scholar : PubMed/NCBI

27 

Hung WY, Lee WJ, Cheng GZ, Tsai CH, Yang YC, Lai TC, Chen JQ, Chung CL, Chang JH and Chien MH: Blocking MMP-12-modulated epithelial-mesenchymal transition by repurposing penfluridol restrains lung adenocarcinoma metastasis via uPA/uPAR/TGF-β/Akt pathway. Cell Oncol (Dordr). 44:1087–1103. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Ranjan A, German N, Mikelis C, Srivenugopal K and Srivastava SK: Penfluridol induces endoplasmic reticulum stress leading to autophagy in pancreatic cancer. Tumour Biol. 39:10104283177055172017. View Article : Google Scholar : PubMed/NCBI

29 

Ranjan A and Srivastava SK: Penfluridol suppresses pancreatic tumor growth by autophagy-mediated apoptosis. Sci Rep. 6:261652016. View Article : Google Scholar : PubMed/NCBI

30 

Dandawate P, Kaushik G, Ghosh C, Standing D, Ali Sayed AA, Choudhury S, Subramaniam D, Manzardo A, Banerjee T, Santra S, et al: Diphenylbutylpiperidine Antipsychotic Drugs Inhibit Prolactin Receptor Signaling to Reduce Growth of Pancreatic Ductal Adenocarcinoma in Mice. Gastroenterology. 158:1433–1449.e27. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Chien W, Sun QY, Lee KL, Ding LW, Wuensche P, Torres-Fernandez LA, Tan SZ, Tokatly I, Zaiden N, Poellinger L, et al: Activation of protein phosphatase 2A tumor suppressor as potential treatment of pancreatic cancer. Mol Oncol. 9:889–905. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Ranjan A and Srivastava SK: Penfluridol suppresses glioblastoma tumor growth by Akt-mediated inhibition of GLI1. Oncotarget. 8:32960–32976. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Kim H, Chong K, Ryu BK, Park KJ, Yu MO, Lee J, Chung S, Choi S, Park MJ, Chung YG and Kang SH: Repurposing penfluridol in combination with temozolomide for the treatment of glioblastoma. Cancers (Basel). 11:13102019. View Article : Google Scholar : PubMed/NCBI

34 

Ranjan A, Wright S and Srivastava SK: Immune consequences of penfluridol treatment associated with inhibition of glioblastoma tumor growth. Oncotarget. 8:47632–47641. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Hu J, Cao J, Jin R, Zhang B, Topatana W, Juengpanich S, Li S, Chen T, Lu Z, Cai X and Chen M: Inhibition of AMPK/PFKFB3 mediated glycolysis synergizes with penfluridol to suppress gallbladder cancer growth. Cell Commun Signal. 20:1052022. View Article : Google Scholar : PubMed/NCBI

36 

van der Horst G, van de Merbel AF, Ruigrok E, van der Mark MH, Ploeg E, Appelman L, Tvingsholm S, Jäätelä M, van Uhm J, Kruithof-de Julio M, et al: Cationic amphiphilic drugs as potential anticancer therapy for bladder cancer. Mol Oncol. 14:3121–3134. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Zheng C, Yu X, Liang Y, Zhu Y, He Y, Liao L, Wang D, Yang Y, Yin X, Li A, et al: Targeting PFKL with penfluridol inhibits glycolysis and suppresses esophageal cancer tumorigenesis in an AMPK/FOXO3a/BIM-dependent manner. Acta Pharm Sin B. 12:1271–1287. 2022. View Article : Google Scholar : PubMed/NCBI

38 

Wu SY, Wen YC, Ku CC, Yang YC, Chow JM, Yang SF, Lee WJ and Chien MH: Penfluridol triggers cytoprotective autophagy and cellular apoptosis through ROS induction and activation of the PP2A-modulated MAPK pathway in acute myeloid leukemia with different FLT3 statuses. J Biomed Sci. 26:632019. View Article : Google Scholar : PubMed/NCBI

39 

Tung MC, Lin YW, Lee WJ, Wen YC, Liu YC, Chen JQ, Hsiao M, Yang YC and Chien MH: Targeting DRD2 by the antipsychotic drug, penfluridol, retards growth of renal cell carcinoma via inducing stemness inhibition and autophagy-mediated apoptosis. Cell Death Dis. 13:4002022. View Article : Google Scholar : PubMed/NCBI

40 

Wu LL, Liu YY, Li ZX, Zhao Q, Wang X, Yu Y, Wang YY, Wang YQ and Luo F: Anti-tumor effects of penfluridol through dysregulation of cholesterol homeostasis. Asian Pac J Cancer Prev. 15:489–494. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Du J, Shang J, Chen F, Zhang Y, Yin N, Xie T, Zhang H, Yu J and Liu F: A CRISPR/Cas9-Based screening for non-homologous end joining inhibitors reveals ouabain and penfluridol as radiosensitizers. Mol Cancer Ther. 17:419–431. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Janssen PA, Niemegeers CJ, Schellekens KH, Lenaerts FM, Verbruggen FJ, Van Nueten JM and Schaper WK: The pharmacology of penfluridol (R 16341) a new potent and orally long-acting neuroleptic drug. Eur J Pharmacol. 11:139–154. 1970. View Article : Google Scholar : PubMed/NCBI

43 

Airoldi L, Marcucci F, Mussini E and Garattini S: Distribution of penfluridol in rats and mice. Eur J Pharmacol. 25:291–295. 1974. View Article : Google Scholar : PubMed/NCBI

44 

Andrade C: Psychotropic drugs with long half-lives: Implications for drug discontinuation, occasional missed doses, dosing interval, and pregnancy planning. J Clin Psychiatry. 83:22f145932022. View Article : Google Scholar : PubMed/NCBI

45 

Bhattacharyya R, Bhadra R, Roy U, Bhattacharyya S, Pal J and Saha SS: Resurgence of penfluridol: Merits and demerits. East J Psychiatry. 18:23–29. 2015. View Article : Google Scholar

46 

Nikvarz N, Vahedian M and Khalili N: Chlorpromazine versus penfluridol for schizophrenia. Cochrane database Syst Rev. 9:CD0118312017.PubMed/NCBI

47 

Wang RI, Larson C and Treul SJ: Study of penfluridol and chlorpromazine in the treatment of chronic schizophrenia. J Clin Pharmacol. 22:236–242. 1982. View Article : Google Scholar : PubMed/NCBI

48 

Andrade C: The practical importance of half-life in psychopharmacology. J Clin Psychiatry. 83:22f145842022. View Article : Google Scholar : PubMed/NCBI

49 

Clarke Z: Penfluridol. Elsevier; New York, NY: pp. 1–4. 2007

50 

Enyeart JJ, Biagi BA, Day RN, Sheu SS and Maurer RA: Blockade of low and high threshold Ca2+ channels by diphenylbutylpiperidine antipsychotics linked to inhibition of prolactin gene expression. J Biol Chem. 265:16373–16379. 1990. View Article : Google Scholar : PubMed/NCBI

51 

Cabrera M, Gomez N, Remes Lenicov F, Echeverría E, Shayo C, Moglioni A, Fernández N and Davio C: G2/M cell cycle arrest and tumor selective apoptosis of acute leukemia cells by a promising benzophenone thiosemicarbazone compound. PLoS One. 10:e01368782015. View Article : Google Scholar : PubMed/NCBI

52 

Abbas T and Dutta A: p21 in cancer: Intricate networks and multiple activities. Nat Rev Cancer. 9:400–414. 2009. View Article : Google Scholar : PubMed/NCBI

53 

Boudreau RT, Conrad DM and Hoskin DW: Apoptosis induced by protein phosphatase 2A (PP2A) inhibition in T leukemia cells is negatively regulated by PP2A-associated p38 mitogen-activated protein kinase. Cell Signal. 19:139–151. 2007. View Article : Google Scholar : PubMed/NCBI

54 

Nakamura H and Takada K: Reactive oxygen species in cancer: Current findings and future directions. Cancer Sci. 112:3945–3952. 2021. View Article : Google Scholar : PubMed/NCBI

55 

Yang H, Villani RM, Wang H, Simpson MJ, Roberts MS, Tang M and Liang X: The role of cellular reactive oxygen species in cancer chemotherapy. J Exp Clin Cancer Res. 37:2662018. View Article : Google Scholar : PubMed/NCBI

56 

Shah MA and Rogoff HA: Implications of reactive oxygen species on cancer formation and its treatment. Semin Oncol. 48:238–245. 2021. View Article : Google Scholar : PubMed/NCBI

57 

Singh R and Manna PP: Reactive oxygen species in cancer progression and its role in therapeutics. Explor Med. 3:43–57. 2022. View Article : Google Scholar

58 

Perillo B, Di Donato M, Pezone A, Di Zazzo E, Giovannelli P, Galasso G, Castoria G and Migliaccio A: ROS in cancer therapy: The bright side of the moon. Exp Mol Med. 52:192–203. 2020. View Article : Google Scholar : PubMed/NCBI

59 

Kim SJ, Kim HS and Seo YR: Understanding of ROS-Inducing strategy in anticancer therapy. Oxid Med Cell Longev. 2019:53816922019. View Article : Google Scholar : PubMed/NCBI

60 

Miller DM, Thomas SD, Islam A, Muench D and Sedoris K: c-Myc and cancer metabolism. Clin Cancer Res. 18:5546–5553. 2012. View Article : Google Scholar : PubMed/NCBI

61 

Gao FY, Li XT, Xu K, Wang RT and Guan XX: c-MYC mediates the crosstalk between breast cancer cells and tumor microenvironment. Cell Commun Signal. 21:282023. View Article : Google Scholar : PubMed/NCBI

62 

Safe S: Specificity Proteins (Sp) and Cancer. Int J Mol Sci. 24:51642023. View Article : Google Scholar : PubMed/NCBI

63 

Vellingiri B, Iyer M, Devi Subramaniam M, Jayaramayya K, Siama Z, Giridharan B, Narayanasamy A, Abdal Dayem A and Cho SG: Understanding the role of the transcription factor sp1 in ovarian cancer: From theory to practice. Int J Mol Sci. 21:11532020. View Article : Google Scholar : PubMed/NCBI

64 

Dufour S, Broders-Bondon F and Bondurand N: Chapter 13 - β1-Integrin Function and Interplay during Enteric Nervous System Development. Academic Press; Boston, MA: pp. 153–166. 2015

65 

Bergonzini C, Kroese K, Zweemer AJM and Danen EHJ: Targeting integrins for cancer therapy-disappointments and opportunities. Front cell Dev Biol. 10:8638502022. View Article : Google Scholar : PubMed/NCBI

66 

Valdembri D and Serini G: The roles of integrins in cancer. Fac Rev. 10:452021. View Article : Google Scholar : PubMed/NCBI

67 

Yousefi H, Vatanmakanian M, Mahdiannasser M, Mashouri L, Alahari NV, Monjezi MR, Ilbeigi S and Alahari SK: Understanding the role of integrins in breast cancer invasion, metastasis, angiogenesis, and drug resistance. Oncogene. 40:1043–1063. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Desgrosellier JS and Cheresh DA: Integrins in cancer: Biological implications and therapeutic opportunities. Nat Rev Cancer. 10:9–22. 2010. View Article : Google Scholar : PubMed/NCBI

69 

Kim SY: Cancer energy metabolism: Shutting power off cancer factory. Biomol Ther (Seoul). 26:39–44. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Chelakkot C, Chelakkot VS, Shin Y and Song K: Modulating glycolysis to improve cancer therapy. Int J Mol Sci. 24:26062023. View Article : Google Scholar : PubMed/NCBI

71 

Fadaka A, Ajiboye B, Ojo O, Adewale O, Olayide I and Emuowhochere R: Biology of glucose metabolization in cancer cells. J Oncol Sci. 3:45–51. 2017. View Article : Google Scholar

72 

Lu J, Chen S, Bai X, Liao M, Qiu Y, Zheng LL and Yu H: Targeting cholesterol metabolism in Cancer: From molecular mechanisms to therapeutic implications. Biochem Pharmacol. 218:1159072023. View Article : Google Scholar : PubMed/NCBI

73 

Fan YJ and Zong WX: The cellular decision between apoptosis and autophagy. Chin J Cancer. 32:121–129. 2013.PubMed/NCBI

74 

Das S, Shukla N, Singh SS, Kushwaha S and Shrivastava R: Mechanism of interaction between autophagy and apoptosis in cancer. Apoptosis. 26:512–533. 2021. View Article : Google Scholar : PubMed/NCBI

75 

Mulcahy Levy JM and Thorburn A: Autophagy in cancer: Moving from understanding mechanism to improving therapy responses in patients. Cell Death Differ. 27:843–857. 2020. View Article : Google Scholar : PubMed/NCBI

76 

Koukourakis MI, Kalamida D, Giatromanolaki A, Zois CE, Sivridis E, Pouliliou S, Mitrakas A, Gatter KC and Harris AL: Autophagosome Proteins LC3A, LC3B and LC3C have distinct subcellular distribution kinetics and expression in cancer cell lines. PLoS One. 10:e01376752015. View Article : Google Scholar : PubMed/NCBI

77 

Fares J, Fares MY, Khachfe HH, Salhab HA and Fares Y: Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct Target Ther. 5:282020. View Article : Google Scholar : PubMed/NCBI

78 

Ribatti D, Tamma R and Annese T: Epithelial-Mesenchymal transition in cancer: A historical overview. Transl Oncol. 13:1007732020. View Article : Google Scholar : PubMed/NCBI

79 

Huang Y, Hong W and Wei X: The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J Hematol Oncol. 15:1292022. View Article : Google Scholar : PubMed/NCBI

80 

Liu ZL, Chen HH, Zheng LL, Sun LP and Shi L: Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther. 8:1982023. View Article : Google Scholar : PubMed/NCBI

81 

Yang Y and Cao Y: The impact of VEGF on cancer metastasis and systemic disease. Semin Cancer Biol. 86((Pt 3)): 251–261. 2022. View Article : Google Scholar : PubMed/NCBI

82 

Zhang L, Zhang M, Wang L, Li J, Yang T, Shao Q, Liang X, Ma M, Zhang N, Jing M, et al: Identification of CCL4 as an immune-related prognostic biomarker associated with tumor proliferation and the tumor microenvironment in clear cell renal cell carcinoma. Front Oncol. 11:6946642021. View Article : Google Scholar : PubMed/NCBI

83 

Rezayatmand H, Razmkhah M and Razeghian-Jahromi I: Drug resistance in cancer therapy: The Pandora's Box of cancer stem cells. Stem Cell Res Ther. 13:1812022. View Article : Google Scholar : PubMed/NCBI

84 

Zheng HC: The molecular mechanisms of chemoresistance in cancers. Oncotarget. 8:59950–59964. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Gong L, Zhang Y, Liu C, Zhang M and Han S: Application of radiosensitizers in cancer radiotherapy. Int J Nanomedicine. 16:1083–1102. 2021. View Article : Google Scholar : PubMed/NCBI

86 

Ashraf-Uz-Zaman M, Shahi S, Akwii R, Sajib MS, Farshbaf MJ, Kallem RR, Putnam W, Wang W, Zhang R, Alvina K, et al: Design, synthesis and structure-activity relationship study of novel urea compounds as FGFR1 inhibitors to treat metastatic triple-negative breast cancer. Eur J Med Chem. 209:1128662021. View Article : Google Scholar : PubMed/NCBI

87 

Ashraf-Uz-Zaman M, Sajib MS, Cucullo L, Mikelis CM and German NA: Analogs of penfluridol as chemotherapeutic agents with reduced central nervous system activity. Bioorg Med Chem Lett. 28:3652–3657. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 52 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ali Ibrahim Mze A and Abdul Rahman A: Repurposing the antipsychotic drug penfluridol for cancer treatment (Review). Oncol Rep 52: 174, 2024.
APA
Ali Ibrahim Mze, A., & Abdul Rahman, A. (2024). Repurposing the antipsychotic drug penfluridol for cancer treatment (Review). Oncology Reports, 52, 174. https://doi.org/10.3892/or.2024.8833
MLA
Ali Ibrahim Mze, A., Abdul Rahman, A."Repurposing the antipsychotic drug penfluridol for cancer treatment (Review)". Oncology Reports 52.6 (2024): 174.
Chicago
Ali Ibrahim Mze, A., Abdul Rahman, A."Repurposing the antipsychotic drug penfluridol for cancer treatment (Review)". Oncology Reports 52, no. 6 (2024): 174. https://doi.org/10.3892/or.2024.8833