Repurposing Drugs against Cancer
- Introduction
Cancer is a group of
disorders characterized by aberrant cell proliferation and resistance to
growth-suppressing signals. It is a multi-tutorial genetic disease that may be
detected early if diagnostic parameters are promising, and it can be
successfully treated by focusing on potential therapeutic targets. While the emphasis on lifestyle may rule out an epigenetic role in cancer progressions,
such as reducing bodily exposure to carcinogens in both diet and usable
materials for example avoiding excessive sun exposure and tanning materials can
lower the risk of skin cancer (Cancer Statistics, 2018). Today, cancer is one
of the most common causes of death around the world. For the year 2018, there
were an anticipated 1.8 million new cancer diagnoses and 600,000 deaths in the
United States alone (Cancer Statistics, 2018).
Cancer growth is
triggered by a failure in the genetic processes that regulate cell growth and
proliferation. In most situations, oncogenesis is caused by accumulated damage
to numerous genes (the "multi-hit" model) caused by physical and
chemical agents, replication errors, and other factors. However, a person's
inherited genetic background may also play a significant role. In cancer, a
single altered cell develops to create a primary tumor, then metastasizes to
another tissue and becomes a secondary tumor after accumulating more mutations
and becoming more aggressive. Metastasis is often nonrandom, with tissues that
generate growth factors and angiogenic factors being the most susceptible to invasion.
The potential of a malignant tumor to penetrate and metastasize to other
tissues is what distinguishes it from a benign tumor. Tumors are categorized
based on the embryonic origin of the tissue from which they develop. Cancers of
endodermal (e.g., gut epithelial cancers) or ectodermal (e.g., skin, neural
epithelia) origin are referred to as carcinomas. Sarcomas are the cancer of
mesodermal origin (e.g., muscle, blood cells). Carcinomas account for more than
90% of all malignant tumors.
Mutations that impact
growth factor receptors and signal transduction genes, cell cycle regulatory
genes, DNA repair genes, or apoptosis genes induce alterations in cellular
growth. The mutant gene is classified as an oncogene or a tumor-suppressor gene
based on whether it typically stimulates or inhibits proliferation
respectively.
Very often
(particularly in early stages) the diagnosis of cancer involves a histological
examination of the affected tissue. Microscopically, cancer cells show abnormal
location and morphology, loss of structural features characteristic of a
differentiated cell, and an abnormally large nucleus and prominent nucleoli.
The cytoskeleton typically also is abnormal. Karyotyping may show gross
chromosomal abnormalities in advanced tumors. Such large tumors, in fact tumors
larger than 2 mm in diameter, develop their own blood supply. To do this,
cancerous cells secrete growth and angiogenic factors such as basic fibroblast
growth factor (b-FGF), transforming growth factor a (TGFa), and vascular endothelial
growth factor (VEGF).
Most malignant tumor cells eventually acquire the ability to metastasize. To do so they must degrade the basement membranes of connective tissue underlying epithelial cells and surrounding the endothelial cells of blood vessels. This can be accomplished by secretion of plasminogen activator, which activates the blood protease, plasmin. Malignant cells form structures called invadopodia which contain protein components needed for crossing basement membranes.
Table. - Classes of genes implicated in the onset of cancer
Drug repurposing against cancer includes the screening of drugs against cancer already present in the market. It could be human/veterinary and their pharmacokinetics and toxicology studies are done. Moreover, pre-analysis of unexpected side effects and in-vivo efficacy has already been done. There are several classes of drugs based on their type of disease against which it is used.
Sedatives: Bortezomib, antipsychotic drugs, Pomalidomide, lenalidomide, thalidomide, and dexamethasone. Thalidomide - inhibits DNA replication, angiogenesis, chondrogenesis, and cell death, inhibits mRNAprocession of TNFa and VEGF. Their anti-cancer properties have been seen against multiple myeloma. They upregulate p21 expression and induce apoptosis. Lenalidomide - The combination of lenalidomide and dexamethasone along with proteasome inhibitor bortezomib extended the survival rate. Dexamethasone - Along with thalidomide yield a 60% response rate against multiple myeloma.
NSAID: Acetylsalicylic acid (Aspirin) reduces metastasis in mice. Celecoxib against colon, lung, breast cancers and it is COX-2 inhibitor. Risk of breast cancer reduced to 71% in female patients using celecoxib. Taxanes, Letrozoles, Tamoxifen, Anastrozole in combination with COX-2 inhibitor but still further evaluation is needed.
Anticonvulsant//Antiepileptic: these include lamotrigine, Valproic Acid (HDAC inhibitor) interacts with oxidative metabolism, γ−Amino butyric acid, 1-methyl-1-cyclohexanecarboxylic acid (MCCA), GSK-3b, Akt signalling, TCA, Erk pathway etc. Phenytoin is voltage gated sodium channel blocker. It inhibits migration and invasion of MDA-MB-231 cells.
Antihyperlipidemic: Statins include simvastatin, atorvastatin, fluvastatin, lovastatin, pravastatin, rosuvastatin and pitavastatin, are agents that inhibit 3-hydroxy-3-methylglutaryl-coenzyme A (HMGCoA) reductase, that catalyzes the important step in cholesterol biosynthesis. They inhibit proliferation, promote cell cycle arrest, apoptosis and differentiation in tumor cells. Statins potentiate TNF induced apoptosis in CML and reduced incidence of colorectal cancer.
Antidiabetic: Metformin inhibits cancer cells growth and proliferation and different cell lines demonstrated sensitivity to the inhibition of oxygen consumption by metformin, with relatively small variation [1]. The anticancer effects of metformin by both direct (insulin-independent) and indirect (insulin-dependent) mechanisms are discussed in terms of metformin-targeted processes and the ontogenesis of cancer stem cells [2]. Combination of disulfiram and metformin inhibited fibrosarcoma growth in hamsters [3]. Metformin reduces intracellular reactive oxygen species levels via the AMPK-FOXO3 pathway [4]. Metformin has rare but potentially serious side effects, such as idiosyncratic hepatotoxicity but usually Metformin is not considered intrinsically hepatotoxic [5]. Metformin may prevent or ameliorate gentamicin-induced acute renal failure, and therefore it might be beneficial in patients under treatment with this medicine [6]. It can cross the blood-brain barrier. Metformin up-regulated the rat brain endothelial barrier function by increasing the phosphorylation levels of AMPK in brain endothelial cells [7]. Metformin prevents ischemia-induced brain injury by alleviating neutrophil infiltration by down-regulating ICAM-1 in an AMPK-dependent manner [8].
Antimalarial: ART caused dose-dependent decreases in cell number, which were associated with either increased cytotoxicity or cytostasis in MCF and A549 cells [9]. It induces oxidative stress in the mechanism of ART action in tumor cells and suggests that antioxidant defenses act in combination to affect the cellular response to ART [10]. It induces mild liver toxicity but enhances bone marrow toxicity. Administration of artesunate for 14 days caused a significant increase in the levels of ALT, AST, ALP and GGT in serum, inducing hepatic injury [11, 12]. Generation of mitochondrial ROS prompted by artesunate was found to be the principal mechanism of cell death in renal carcinoma [13]. It caused irreversible vascular irritation, reversible nephrotoxicity and no neurotoxicity at high doses. Artesunate could preserve blood–brain barrier integrity and improve neurological outcome after subarachnoid hemorrhage, by activating S1P1 [14]. It has high permeability of artemisinin across biological membranes and can cross the blood-brain barrier [15].
Antiviral: Acyclovir monophosphate exhibited concentration-dependent cytotoxicity against H460 cells and increased S-phase arrest [16]. Ethyl phosphoramidate derivatives show promising antioxidant activity [17]. Acyclovir group showed significant decrease of serum albumin, elevation of alkaline phosphatase activity associated with a severe apoptotic changes as evidenced by Bc12 overexpression in liver cells [18]. Acute renal insufficiency associated with high-dose of acyclovir administered intravenously. Nephrotoxicity developed despite precautions to avoid volume contraction [19, 20]. Zidovudine is a dideoxynucleoside used in the treatment of HIV infection [21]. Zidovudine induced liver toxicity and oxidative stress [22]. hepatotoxicity, oxidative stress and hyperlipidaemia induced by AZT in rat liver [23]. It induces liver toxicity and oxidative stress. It is responsible for mitochondrial myopathy, mitochondrial DNA depletion, lactic acidosis alone or associated with hepatic abnormalities [24]. AZT at a dose of 400 mg/kg/day for 17 days caused clear hematotoxicity and nephrotoxicity[25]. It can cross the blood brain barrier and Zidovudine passes the monolayer mainly by passive diffusion [26] [27]. DDI is transported from the brain into circulating blood across the blood-brain barrier via a probenecid-sensitive carrier-mediated efflux transport system [28]. Didanosine is a hypoxanthine attached to the sugar ring, unlike other nucleoside analogues. Dideoxyinosine derivatives show anti-cancer activity [29]. DDI may or may not be the only potential cause of hepato-toxicity [30] and passage across the BBB was demonstrated to be via non-saturable and saturable processes [31, 32].
Antifungal: Triazole induces anti-angiogenic activity and inhibits hedgehog signalling pathway, induces autophagic growth arrest. Against leukemia, ovarian, breast and pancreatic cancers. Thiabendazole TBZ induces anti-angiogenic activity and decreases vascular density. Rapamycin induces its analogues against breast cancer. Inhibits mTOR signalling pathway.
Anthelmintic: Cyclodextrin complexes of niclosamide show enhanced anticancer activity both in in-vitro and in-vivo mice model [33, 34]. Antioxidant - it increases ROS production in AML, lung cancer cells and enhances ROS production mediated cell death via c-jun activation [35, 36]. It targets Wnt/b-catenin signalling and attenuates cancer stemness [37]. No known nephrotoxicity of niclosamide has been reported even improvement of renal ischemia/reperfusion injury has been validated [38]. Mebendazole inhibited the growth of breast, ovary, colon carcinomas and osteosarcoma with IC50 values in the range of 0.1 to 0.8 μM. Mebendazole is a benzimidazole anthelmintic used to treat helminth infections. Inhibitory effects of MBZ against breast, ovary, colon carcinomas, and osteosarcoma, producing IC50s that varied from 0.1 to 0.8 μM [39]. Mebendazole blocked anticancer activity of disulfiram and metformin combination [3]. The comparative analysis of results between intact group and control pathology showed a significant mebendazole-induced neurotoxicity [40]. Mebendazole has not shown any major liver toxicity and renal toxicity [41, 42]. Mebendazole crosses the blood-brain barrier and among patients with cystic echinococcosis, there is significant variab ility in absorption [43]. Its different polymorphs can cross BBB and reach upto brain tumor as seen in mice [44]. It crosses BBB and reaches well into brain tumor tissue [45].
Drugs for osteoporosis: Zoledronic acid induces caspase-dependent apoptosis in renal cancer cell lines [46]. ZA induces rabbit liver oxidative stress and decreases antioxidant levels in liver tissue [47]. ZA induces apoptosis through promoting ROS production and subsequently activating chloride channels in nasopharyngeal carcinoma [48]. Zoledronic acid may cause hepato-toxicity [49, 50] but helps in preventing bone loss after liver transplantation [51]. Patients with multiple myeloma or different types of metastatic cancer, who had experienced renal impairment during zoledronic acid therapy [52]. It causes renal toxicity in patients with multiple myeloma and bone metastasis [53, 54]. Blood-brain barrier - Information not found but Transferrin-targeted self-assembled nanoparticles (NPs) incorporating zoledronic acid increases the antitumor efficacy of this drug in glioblastoma through the acquisition of ability to cross the BBB [55].
- Potential Anticancer Drugs
Celecoxib
4-[5-(4-methylphenyl)–3-(trifluoromethyl)-1H-pyrazol-1-yl]
benzenesulfonamide, a COX-2-selective NSAID, is known as celecoxib. The US Food
and Drug Administration (FDA) authorised the oral capsule version in 1999, and
Pfizer, Inc. (New York, NY, USA) began marketing it in 2000. Celecoxib is an
analgesic, anti-inflammatory, and antipyretic medication and known for its
decreased risk of causing gastrointestinal. Celecoxib appears to have a
substantial chemopreventive effect on breast cancer, according to preclinical
studies [56,
57]. In various animal models of
breast cancer, celecoxib therapy (500–1,500 mg/kg diet) reduces tumour
incidence, multiplicity, and volume [58, 59]. Furthermore, metastasis to
the lungs and brain might be avoided and clinical trials, in addition to
preclinical investigations, yielded good findings. A standard dose of celecoxib
(200 mg/day) for more than 12 months was related to a significantly decreased
risk of breast cancer in two cases–control trials [60, 61]. It reduces precancerous
polyps in the colon and NSAIDS are known to exhibit anti-colon cancer effects
for 2 decades [62]. Cyclooxygenase 2 converts
arachidonic acid into prostaglandins E2 (PGE2) and its overexpression helps in
breast cancer progression [63]. In turn, PGE2 governs
anti-tumor immunity suppression, increased metastasis, lymphangiogenesis and
stem cell like properties [64-67].
Cyclooxygenase 2 is
associated with breast cancer cell motility, metastasis and cancer progression [68]. Cyclooxygenase is highly
expressed in breast cancer and ductal carcinoma in-situ as compared to normal
tissue while it is also expressed in different types of tumor [69-71]. Celecoxib exerts anticancer effects by binding to the
cadherin-11 (CDH11) protein, which is thought to be involved in the progression
of tumors. Celecoxib inhibits promotion of mammary tumorigenesis in rats. It
might contain antioxidant activity, not validated [72] and may also have a
pro-oxidative anti-cancer effect [73]. The cholestatic hepatitis
is temporally related to celecoxib use [74] while Celecoxib induced
renal Injury in rats has also been reported [75]. The use of celecoxib is
associated with renal effects similar to that of conventional nonselective
NSAIDs [76].
Celecoxib was shown to reduce
glioma cell viability by inducing DNA damage, leading to p53-dependent G1
cell-cycle arrest and autophagy. However, the use of celecoxib in patients with
glioblastoma has seen mixed results, due in part to the low potential of the
drug to cross the blood–brain barrier (BBB) [77]. Celecoxib showed a
significant protective effect on HBMECs against irradiation, which involves
inhibited apoptosis; celecoxib could alleviate radiation-induced brain injury
in rats [78]. Celecoxib inhibits the
expression of Snail, Slug, ZEB1, vimentin, focal adhesion kinase etc, thus
blocking tumor cell mobility in in-vitro studies.
In-vivo administration of celecoxib
in tumor xenograft mice model exhibit 65% inhibition of oral squamous cell
carcinoma. celecoxib administered tumor xenografts were well differentiated as
compared to control [68].
Celecoxib inhibits the growth
of breast cancer cells in vitro and protects rats from developing BC caused by
7,12-dimethylbenzanthracene (DMBA). As a result, celecoxib has anticancer
properties and appears to be useful in cancer treatment. Bocca et al
investigated celecoxib's antiproliferative efficacy in human breast cancer
cells with varying levels of COX-2 expression. Celecoxib administration causes
a significant reduction in cell growth in oestrogen receptor (ER) (+) MCF-7
cells, as well as a decrease in aromatase and ER expression and activation.
Inhibition of ERK and Akt, as well as activation of PP2A and PTEN, may be
involved in the linked pathway [79, 80]. Short-term COX-2 inhibition
by celecoxib promotes transcriptional programmes that enable antitumor activity
in primary breast cancer tissue, according to a research finding
transcriptional alterations in breast cancer tissues of patients treated with
the medication. Basu et al investigated the mechanisms by which celecoxib
impacts tumour development in MDAMB-231 (very invasive) and MDA-MB-468
(moderately invasive) human breast cancer cell lines. In diverse human breast
cancer cell lines, they established that the varied molecular pathways of
celecoxib-induced growth suppression are dependent on COX-2 expression levels
and invasiveness [81]. Celecoxib's tumour growth
inhibition effect was found to be mostly due to promoting apoptosis rather than
affecting cell proliferation [82].
COX-2 overexpression in
celecoxib-resistant cell lines results in lower levels of Bax, a pro-apoptosis
protein, and higher levels of anti-apoptosis proteins such as Bcl-2 or Bcl-xL [83]. COX-2 knockdown using a
particular siRNA reduces clonogenicity and Bcl-xL and Bcl-2 levels in these
cells considerably. Celecoxib causes apoptosis in breast cancer cells by
lowering Akt phosphorylation, raising Bax expression, and activating caspase 3
and caspase 7. Celecoxib also inhibits the NF-B pathway, which causes apoptosis
in the breast cancer cells. Another route of celecoxib-induced apoptosis is the
p53-independent mitochondrial apoptosis pathway, which is independent of COX-2
and cannot be blocked by Bcl-2 overexpression [84, 85].
Dietary administration of celecoxib also produces anti-tumor effects in mice, decreasing tumor volume and cancer progression [86]. Despite the promising effectiveness described above, the mechanisms of celecoxib's anticancer activity remain unknown. In carcinogenesis and development, COX-2 plays a critical function. As a selective COX-2 inhibitor, celecoxib is thought to have multiple anticancer mechanisms, including proliferation suppression, apoptosis induction, immunoregulation, tumour microenvironment regulation, antiangiogenic action, and resensitization of other antitumor medicines. Meanwhile, celecoxib's anticancer action is aided by COX-2-independent mechanisms.
Lamotrigine
Lamotrigine is an
antiepileptic drug belonging to the phenyltriazine class. It is used in the
treatment of both epilepsy and as a mood stabilizer in bipolar disorder. It
inhibits the proliferation, the anchorage-dependent, and independent cell
growth in breast cancer cells [87]. Lamotrigine has the most
protective effects on oxidative stress [88]. Acute hepatitis is also
associated with Lamotrigine [89], chronic exposure of rats to
high doses of LTG reflects hepatocellular damage that may lead to hepatitis [90]. Lamotrigine induced
neuroleptic malignant syndrome in a patient with renal failure caused by
lithium [91]. Lamotrigine can also cross
the blood-brain barrier [92]. Lamotrigine dramatically
improved the oxidant/antioxidant milieu while decreasing acetylcholine esterase
and glutamate levels in brain tissue [93]. Lamotrigine is a
second-generation anti-epileptic drug with a broad activity range, a manageable
adverse effect profile, easier dosage than previous medications, and improved
effectiveness in a variety of epileptic syndromes. Unfortunately, lamotrigine
is the most interacting medication among the new anti-epileptic drugs, and its
metabolism is vulnerable to both enzyme inhibition and induction [93].
Lamotrigine is a
voltage-gated sodium channel blocking medication that has anti-cancer efficacy
based mostly on other mechanisms and is thought to work against cancer by
inhibiting histone deacetylase [94]. Because there is very
little and insufficient information in the current literature, we sought to
investigate the potential antitumoral activity of lamotrigine, a synthetic
phenyltriazine with anticonvulsant properties that has been commercially
available since the mid-1990s for the treatment of epilepsy and bipolar
disorder [95]. The capacity of lamotrigine
to inhibit voltage-activated Na and Ca channels is thought to be the mechanism
by which it exerts its anti seizure activity [96]. Lamotrigine inhibits breast
cancer cell proliferation, anchorage-dependent and independent cell growth.
These effects were linked to cell-cycle arrest and changes in related proteins
and target genes of FoxO3a, a ubiquitous transcription factor that is adversely
regulated by AKT. Lamotrigine also enhances the expression of another FoxO3a
target, PTEN, which downregulates the PI3K/Akt signalling pathway, resulting in
FoxO3a dephosphorylation and activation. Furthermore, in an autoregulatory
mechanism, lamotrigine stimulates FoxO3a expression by boosting transcription [87].
Lamotrigine's site of action
has already been explored using binding and neurochemical techniques.
Lamotrigine does not bind to dopamine, adenosine, muscarinic, or sigma neuronal
receptors, and it does not appear to be an N-methyl-D-aspartate receptor
antagonist since it does not reduce the production of cyclic GMP in response to
N-methyl-D-aspartate. Lamotrigine reduces veratrine-induced neurotransmitter
release in rat cortical brain slices, according to neurochemical research [97]. It is used to treat
seizures, other types of convulsions, and bipolar disorders. LTG's antiepileptic
action is caused by its binding to voltage-gated sodium channels, which
inhibits the release of endogenous amino acids and acetylcholine. LTG can also
be used to treat colon cancer and chronic myelogenous leukaemia. Furthermore,
both in vitro and in vivo investigations have shown that LTG has potent
antiproliferative effects in breast cancer cells. LTG, at modest dosages, can
boost felbamate's protective index against seizures by strengthening the
anticonvulsant action without significantly increasing neurotoxicity [98].
However, because the effect
of lamotrigine on signal transduction has not been widely studied, and because
the most common therapeutic targets for antitumor drugs are receptors, protein
tyrosine kinases, and the phosphatidylinositol 3-kinase (PI3K)/Akt cell
survival pathway, we will examine the potential interference that lamotrigine
could exert on intracellular signalling molecules in different cancer cells,
either directly or indirectly. Lamotrigine's biological effects and molecular
mechanism in many types of human malignancies have yet to be discovered, as has
its impact on tumour growth. However, the positive benefits of the current
generation of anti-epileptic drugs such as lamotrigine outweigh the harmful
effects of mitochondria [93, 99].
References
1. Chowdhury, S., et al., MATE2 expression is associated with cancer
cell response to metformin. PLoS One, 2016. 11(12): p. e0165214.
2. Del
Barco, S., et al., Metformin:
multi-faceted protection against cancer. Oncotarget, 2011. 2(12): p. 896.
3. Popović,
K.J., et al., Disulfiram and metformin
combination anticancer effect reversible partly by antioxidant nitroglycerin
and completely by NF-κB activator mebendazole in hamster fibrosarcoma.
Biomedicine & Pharmacotherapy, 2021. 143:
p. 112168.
4. Hou,
X., et al., Metformin reduces
intracellular reactive oxygen species levels by upregulating expression of the
antioxidant thioredoxin via the AMPK-FOXO3 pathway. Biochemical and
biophysical research communications, 2010. 396(2):
p. 199-205.
5. Miralles-Linares,
F., et al., Metformin-induced
hepatotoxicity. Diabetes Care, 2012. 35(3):
p. e21-e21.
6. Amini,
F.G., et al., Ameliorative effects of
metformin on renal histologic and biochemical alterations of gentamicin-induced
renal toxicity in Wistar rats. Journal of research in medical sciences: the
official journal of Isfahan University of Medical Sciences, 2012. 17(7): p. 621.
7. Takata,
F., et al., Metformin induces
up-regulation of blood–brain barrier functions by activating AMP-activated
protein kinase in rat brain microvascular endothelial cells. Biochemical
and biophysical research communications, 2013. 433(4): p. 586-590.
8. Liu,
Y., et al., Metformin attenuates
blood-brain barrier disruption in mice following middle cerebral artery
occlusion. Journal of neuroinflammation, 2014. 11(1): p. 1-12.
9. Liu,
W.M., A.M. Gravett, and A.G. Dalgleish, The
antimalarial agent artesunate possesses anticancer properties that can be
enhanced by combination strategies. International Journal of Cancer, 2011. 128(6): p. 1471-1480.
10. Efferth,
T., M.M. Briehl, and M.E. Tome, Role of
antioxidant genes for the activity of artesunate against tumor cells.
International journal of oncology, 2003. 23(4):
p. 1231-1235.
11. Uhl,
M., S. Schwab, and T. Efferth, Fatal
liver and bone marrow toxicity by combination treatment of dichloroacetate and
artesunate in a glioblastoma multiforme patient: case report and review of the
literature. Frontiers in oncology, 2016. 6: p. 204.
12. Jahas,
J., et al., Protective Role of Tylophora
Indica Ethanolic Extract on Artesunate Induced Liver Toxicity. 2014.
13. Chauhan,
A.K., K.-j. Min, and T.K. Kwon, RIP1-dependent
reactive oxygen species production executes artesunate-induced cell death in
renal carcinoma Caki cells. Molecular and cellular biochemistry, 2017. 435(1): p. 15-24.
14. Zuo,
S., et al., Artesunate protected blood–brain
barrier via sphingosine 1 phosphate receptor 1/phosphatidylinositol 3 kinase
pathway after subarachnoid hemorrhage in rats. Molecular neurobiology,
2017. 54(2): p. 1213-1228.
15. Augustijns,
P., et al., Transport of artemisinin and
sodium artesunate in Caco‐2 intestinal
epithelial cells. Journal of pharmaceutical
sciences, 1996. 85(6): p. 577-579.
16. Yao,
J., et al., Turning an antiviral into an
anticancer drug: nanoparticle delivery of acyclovir monophosphate. Journal
of Controlled Release, 2013. 170(3):
p. 414-420.
17. Devineni,
S.R., et al., Ethyl phosphoramidates of
acyclovir: design, synthesis, molecular docking (HN Protein), and evaluation of
antiviral and antioxidant activities. Medicinal Chemistry Research, 2017. 26(5): p. 999-1009.
18. Hanan
MA, H. and R. Mona, Comparative
hepatotoxic study between acyclovir and famciclovir in adult male albino rats.
2007.
19. Sawyer,
M.H., et al., Acyclovir-induced renal
failure: clinical course and histology. The American journal of medicine,
1988. 84(6): p. 1067-1071.
20. Pouplin,
T., et al., Valacyclovir for herpes
simplex encephalitis. Antimicrobial agents and chemotherapy, 2011. 55(7): p. 3624-3626.
21. Heidari,
A. and R. Gobato, A Novel Approach to
Reduce Toxicities and to Improve Bioavailabilities of DNA/RNA of Human Cancer
Cells–Containing Cocaine (Coke), Lysergide (Lysergic Acid Diethyl Amide or
LSD), Δ⁹–Tetrahydrocannabinol
(THC)[(–)–trans–Δ⁹–Tetrahydrocannabinol],
Theobromine (Xantheose), Caffeine, Aspartame (APM)(NutraSweet) and Zidovudine
(ZDV)[Azidothymidine (AZT)] as Anti–Cancer Nano Drugs by Coassembly of Dual
Anti–Cancer Nano Drugs to Inhibit DNA/RNA of Human Cancer Cells Drug
Resistance. Parana Journal of Science and
Education, 2018. 4(6): p. 1-17.
22. Raghu,
R. and S. Karthikeyan, Zidovudine and
isoniazid induced liver toxicity and oxidative stress: Evaluation of mitigating
properties of silibinin. Environmental toxicology and pharmacology, 2016. 46: p. 217-226.
23. Raghu,
R., et al., Silibinin mitigates
zidovudine-induced hepatocellular degenerative changes, oxidative stress and
hyperlipidaemia in rats. Human & experimental toxicology, 2015. 34(11): p. 1031-1042.
24. Chariot,
P., et al., Zidovudine-induced
mitochondrial disorder with massive liver steatosis, myopathy, lactic acidosis,
and mitochondrial DNA depletion. Journal of hepatology, 1999. 30(1): p. 156-160.
25. Omar,
R.F., et al., In VivoToxicity of
Foscarnet and Zidovudine Given Alone or in Combination. Toxicology and
applied pharmacology, 1996. 139(2):
p. 324-332.
26. Glynn,
S.L. and M. Yazdanian, In vitro
blood–brain barrier permeability of nevirapine compared to other HIV
antiretroviral agents. Journal of pharmaceutical sciences, 1998. 87(3): p. 306-310.
27. Masereeuw,
R., et al., In vitro and in vivo
transport of zidovudine (AZT) across the blood–brain barrier and the effect of
transport inhibitors. Pharmaceutical research, 1994. 11(2): p. 324-330.
28. Gibbs,
J., P. Jayabalan, and S. Thomas, Mechanisms
by which 2′, 3′‐dideoxyinosine
(ddI) crosses the guinea‐pig CNS barriers;
relevance to HIV therapy. Journal of
neurochemistry, 2003. 84(4): p.
725-734.
29. Borek,
W., et al., Synthesis of Novel 2′,
3′-Didehydro-2′, 3′-dideoxyinosine Phosphoramidate Prodrugs and Evaluation of
their Anticancer Activity. Nucleosides, Nucleotides and Nucleic Acids,
2014. 33(8): p. 507-518.
30. Lacaille,
F., et al., Hepatic toxicity associated
with 2'-3'dideoxyinosine in children with AIDS. Journal of pediatric
gastroenterology and nutrition, 1995. 20(3):
p. 287-290.
31. Takasawa,
K., et al., In Vivo Evidence for
Carrier-Mediated Efflux Transport of 3′-Azido-3′-Deoxythymidine and 2′,
3′-Dideoxyinosine Across the Blood-Brain Barriervia a Probenecid-Sensitive
Transport System. Journal of Pharmacology and Experimental Therapeutics,
1997. 281(1): p. 369-375.
32. Johnson,
M.D., J. Chen, and B.D. Anderson, Investigation
of the Mechanism of Enhancement of Central Nervous System Delivery of
2′-β-Fluoro-2′, 3′-Dideoxyinosine Via a Blood-Brain Barrier Adenosine
Deaminase-Activated Prodrug. Drug metabolism and disposition, 2002. 30(2): p. 191-198.
33. Lodagekar,
A., et al., Formulation and evaluation of
cyclodextrin complexes for improved anticancer activity of repurposed drug:
Niclosamide. Carbohydrate polymers, 2019. 212: p. 252-259.
34. Yu,
X., et al., Niclosamide exhibits potent
anticancer activity and synergizes with sorafenib in human renal cell cancer
cells. Cellular Physiology and Biochemistry, 2018. 47(3): p. 957-971.
35. Hamdoun,
S., P. Jung, and T. Efferth, Drug
repurposing of the anthelmintic niclosamide to treat multidrug-resistant
leukemia. Frontiers in pharmacology, 2017. 8: p. 110.
36. Son,
A.-R., J. Ahn, and J.-Y. Song, Niclosamide
enhances ROS-mediated cell death through c-Jun activation. Biomedicine
& Pharmacotherapy, 2014. 68(5):
p. 619-624.
37. Cerles,
O., et al., Niclosamide inhibits
oxaliplatin neurotoxicity while improving colorectal cancer therapeutic
response. Molecular cancer therapeutics, 2017. 16(2): p. 300-311.
38. Zhang,
L.X., et al., Niclosamide attenuates inflammatory
cytokines via the autophagy pathway leading to improved outcomes in renal
ischemia/reperfusion injury. Molecular medicine reports, 2017. 16(2): p. 1810-1816.
39. Pantziarka,
P., et al., Repurposing Drugs in Oncology
(ReDO)—mebendazole as an anti-cancer agent. ecancermedicalscience, 2014. 8.
40. Yemets,
M. and D. Lytkin, The Comparative Study
of AntioxidantsNeuroprotective Properties Under the Condition of
Mebendazole-induced Neurotoxicity. 2020.
41. Liu,
C.-S., et al., An alternative mebendazole
formulation for cystic echinococcosis: the treatment efficacy, pharmacokinetics
and safety in mice. Parasites & vectors, 2014. 7(1): p. 1-10.
42. Al-Waili,
N.S. and S. Al-Waili, Mebendazole in
cutaneous leishmaniasis. J Pak Med Assoc, 1989. 39: p. 20-21.
43. McCarthy,
J.S. and T.A. Moore, Drugs for helminths.
Mandel, Douglas and Bennett’s Principles and Practice of Infectious Diseases.
8th ed. Philadelphia: Saunders, 2015.
44. Bai,
R.-Y., et al., Brain penetration and
efficacy of different mebendazole polymorphs in a mouse brain tumor model.
Clinical Cancer Research, 2015. 21(15):
p. 3462-3470.
45. De
Witt, M., et al., Repurposing mebendazole
as a replacement for vincristine for the treatment of brain tumors.
Molecular Medicine, 2017. 23(1): p.
50-56.
46. Ullén,
A., et al., Zoledronic acid induces
caspase-dependent apoptosis in renal cancer cell lines. Scandinavian
journal of urology and nephrology, 2009. 43(2):
p. 98-103.
47. Karabulut,
A., et al. Oxidant and antioxidant
activity in rabbit livers treated with zoledronic acid. in Transplantation proceedings. 2010.
Elsevier.
48. Wang,
L., et al., The apoptotic effect of
zoledronic acid on the nasopharyngeal carcinoma cells via ROS mediated chloride
channel activation. Clinical and Experimental Pharmacology and Physiology,
2018. 45(10): p. 1019-1027.
49. Polyzos,
S., et al., Zoledronic acid-induced
transient hepatotoxicity in a patient effectively treated for Paget’s disease
of bone. Osteoporosis international, 2011. 22(1): p. 363-367.
50. Lu,
Y., et al., Hepatotoxicity induced by
zoledronic acid in an aged woman with primary osteoporosis. EXCLI journal,
2013. 12: p. 115.
51. Crawford,
B.A., et al., Zoledronic acid prevents
bone loss after liver transplantation: a randomized, double-blind,
placebo-controlled trial. Annals of internal medicine, 2006. 144(4): p. 239-248.
52. Munier,
A., et al., Zoledronic acid and renal
toxicity: data from French adverse effect reporting database. Annals of
Pharmacotherapy, 2005. 39(7-8): p.
1194-1197.
53. Bujanda,
D.A., et al., Assessment of renal
toxicity and osteonecrosis of the jaws in patients receiving zoledronic acid
for bone metastasis. Annals of oncology, 2007. 18(3): p. 556-560.
54. Weide,
R., et al., Renal toxicity in patients
with multiple myeloma receiving zoledronic acid vs. ibandronate: a
retrospective medical records review. Journal of cancer research and
therapeutics, 2010. 6(1): p. 31.
55. Porru,
M., et al., Medical treatment of
orthotopic glioblastoma with transferrin-conjugated nanoparticles encapsulating
zoledronic acid. Oncotarget, 2014. 5(21):
p. 10446.
56. Lanza-Jacoby,
S., et al., The cyclooxygenase-2
inhibitor, celecoxib, prevents the development of mammary tumors in Her-2/neu
mice. Cancer Epidemiology and Prevention Biomarkers, 2003. 12(12): p. 1486-1491.
57. Liu,
Y., et al., Premetastatic soil and
prevention of breast cancer brain metastasis. Neuro-oncology, 2013. 15(7): p. 891-903.
58. Woditschka,
S., et al., Chemopreventive effects of
celecoxib are limited to hormonally responsive mammary carcinomas in the
neu-induced retroviral rat model. Breast Cancer Research, 2008. 10(1): p. 1-9.
59. Kang,
H., et al., Chemopreventive effect of
celecoxib against DMBA-induced breast cancer and its mechanism. Journal of
Southern Medical University, 2006. 26(11):
p. 1599-1602.
60. Harris,
R.E., J. Beebe-Donk, and G.A. Alshafie, Reduction
in the risk of human breast cancer by selective cyclooxygenase-2 (COX-2)
inhibitors. BMC cancer, 2006. 6(1):
p. 1-5.
61. Ashok,
V., et al., Selective cyclooxygenase-2
(COX-2) inhibitors and breast cancer risk. The Breast, 2011. 20(1): p. 66-70.
62. Kune,
G.A., S. Kune, and L.F. Watson, Colorectal
cancer risk, chronic illnesses, operations, and medications: case control
results from the Melbourne Colorectal Cancer Study. Cancer research, 1988. 48(15): p. 4399-4404.
63. Bennett,
A., et al., Breast cancer, its
recurrence, and patient survival in relation to tumor prostaglandins.
Advances in prostaglandin, thromboxane, and leukotriene research, 1983. 12: p. 299-302.
64. Lala,
P., N. Al‐Mutter, and A. Orucevic, Effects
of chronic indomethacin therapy on the development and progression of
spontaneous mammary tumors in C3H/HEJ mice. International journal of
cancer, 1997. 73(3): p. 371-380.
65. Timoshenko,
A.V., et al., Role of prostaglandin E2
receptors in migration of murine and human breast cancer cells.
Experimental cell research, 2003. 289(2):
p. 265-274.
66. Majumder,
M., et al., Prostaglandin E2 receptor EP
4 as the common target on cancer cells and macrophages to abolish angiogenesis,
lymphangiogenesis, metastasis, and stem‐like cell functions. Cancer science,
2014. 105(9): p. 1142-1151.
67. Chang,
S.-H., et al., Role of prostaglandin
E2-dependent angiogenic switch in cyclooxygenase 2-induced breast cancer
progression. Proceedings of the National Academy of Sciences, 2004. 101(2): p. 591-596.
68. Chiang,
S.-L., et al., Preventive effect of
celecoxib use against cancer progression and occurrence of oral squamous cell
carcinoma. Scientific reports, 2017. 7(1):
p. 1-11.
69. Bosco,
J.L., et al., Regular aspirin use and
breast cancer risk in US Black women. Cancer Causes & Control, 2011. 22(11): p. 1553-1561.
70. Díaz-Cruz,
E.S., C.L. Shapiro, and R.W. Brueggemeier, Cyclooxygenase
inhibitors suppress aromatase expression and activity in breast cancer cells.
The Journal of Clinical Endocrinology & Metabolism, 2005. 90(5): p. 2563-2570.
71. Masferrer,
J.L., A. Koki, and K. Seibert, COX‐2 inhibitors: a new class of antiangiogenic agents. Annals of the New York Academy of Sciences, 1999. 889(1): p. 84-86.
72. Kirkova,
M., et al., Potential antioxidant
activity of celecoxib and amtolmetin guacyl: in vitro studies. Autonomic
and autacoid pharmacology, 2007. 27(1):
p. 13-18.
73. Ralph,
S.J., et al., NSAID celecoxib: a potent
mitochondrial pro-oxidant cytotoxic agent sensitizing metastatic cancers and
cancer stem cells to chemotherapy. Journal of Cancer Metastasis and
Treatment, 2018. 4.
74. Grieco,
A., et al., Acute cholestatic hepatitis
associated with celecoxib. Annals of Pharmacotherapy, 2002. 36(12): p. 1887-1889.
75. Cooper,
D.L., et al., Exacerbation of
celecoxib-induced renal injury by concomitant administration of misoprostol in
rats. PloS one, 2014. 9(2): p.
e89087.
76. Ahmad,
S.R., et al., Renal failure associated
with the use of celecoxib and rofecoxib. Drug Safety, 2002. 25(7): p. 537-544.
77. Yerokun,
T. and L.L. Winfield, Celecoxib and
LLW-3-6 reduce survival of human glioma cells independently and synergistically
with sulfasalazine. Anticancer research, 2015. 35(12): p. 6419-6424.
78. Xu,
X., et al., Celecoxib Alleviates
Radiation-Induced Brain Injury in Rats by Maintaining the Integrity of
Blood-Brain Barrier. Dose-Response, 2021. 19(2): p. 15593258211024393.
79. Bocca,
C., et al., Antiproliferative effects of
COX-2 inhibitor celecoxib on human breast cancer cell lines. Molecular and
cellular biochemistry, 2011. 350(1):
p. 59-70.
80. Dai,
Z.-J., et al., Antitumor activity of the
selective cyclooxygenase-2 inhibitor, celecoxib, on breast cancer in vitro and
in vivo. Cancer cell international, 2012. 12(1): p. 1-8.
81. Basu,
G.D., et al., Mechanisms underlying the
growth inhibitory effects of the cyclo-oxygenase-2 inhibitor celecoxib in human
breast cancer cells. Breast Cancer Research, 2005. 7(4): p. 1-14.
82. Basu,
G.D., et al., Cyclooxygenase-2 Inhibitor
Induces Apoptosis in Breast Cancer Cells in an In vivo Model of Spontaneous
Metastatic Breast Cancer11Susan G. Komen Breast Cancer Foundation. Note: GD
Basu and LB Pathangey contributed equally to this work. Molecular Cancer
Research, 2004. 2(11): p. 632-642.
83. Barnes,
N., et al., Cyclooxygenase-2 inhibition:
effects on tumour growth, cell cycling and lymphangiogenesis in a xenograft
model of breast cancer. British journal of cancer, 2007. 96(4): p. 575-582.
84. Singh,
B., et al., Overexpression of COX-2 in
celecoxib-resistant breast cancer cell lines. Journal of Surgical Research,
2010. 163(2): p. 235-243.
85. Baumann,
K.H., et al., Effects of celecoxib and
ly117018 combination on human breast cancer cells in vitro. Breast cancer:
basic and clinical research, 2009. 3:
p. BCBCR. S2291.
86. Harris,
R.E., et al., Chemoprevention of breast
cancer in rats by celecoxib, a cyclooxygenase 2 inhibitor. Cancer research,
2000. 60(8): p. 2101-2103.
87. Pellegrino,
M., et al., FoxO3a mediates the
inhibitory effects of the antiepileptic drug Lamotrigine on breast cancer
growth. Molecular Cancer Research, 2018. 16(6): p. 923-934.
88. Eren,
İ., M. Nazıroğlu, and A. Demirdaş, Protective
effects of lamotrigine, aripiprazole and escitalopram on depression-induced
oxidative stress in rat brain. Neurochemical research, 2007. 32(7): p. 1188-1195.
89. Mecarelli,
O., et al., Acute hepatitis associated
with lamotrigine and managed with the molecular adsorbents recirculating system
(MARS). Epilepsia, 2005. 46(10):
p. 1687-1689.
90. Meshkibaf,
M., et al., Choronic effects of
lamotrigine on liver function in adult male rats. Indian Journal of
Clinical Biochemistry, 2006. 21(1):
p. 161-164.
91. Szota,
A.M., et al., Lamotrigine, quetiapine and
aripiprazole-induced neuroleptic malignant syndrome in a patient with renal
failure caused by lithium: a case report. BMC psychiatry, 2020. 20(1): p. 1-5.
92. Potschka,
H., M. Fedrowitz, and W. Löscher, P-Glycoprotein-mediated
efflux of phenobarbital, lamotrigine, and felbamate at the blood–brain barrier:
evidence from microdialysis experiments in rats. Neuroscience letters,
2002. 327(3): p. 173-176.
93. Chogtu,
B., et al., Comparison of the efficacy of
carbamazepine, gabapentin and lamotrigine for neuropathic pain in rats.
Indian journal of pharmacology, 2011. 43(5):
p. 596.
94. Koltai,
T., Voltage-gated sodium channel as a
target for metastatic risk reduction with re-purposed drugs. F1000Research,
2015. 4.
95. Brodie,
M.J., Lamotrigine. The lancet, 1992. 339(8806): p. 1397-1400.
96. Xie,
X. and R. Hagan, Cellular and molecular
actions of lamotrigine: possible mechanisms of efficacy in bipolar disorder.
Neuropsychobiology, 1998. 38(3): p.
119-130.
97. Lang,
D.G., C.M. Wang, and B. Cooper, Lamotrigine,
phenytoin and carbamazepine interactions on the sodium current present in N4TG1
mouse neuroblastoma cells. Journal of Pharmacology and Experimental
Therapeutics, 1993. 266(2): p.
829-835.
98. Najm,
S., et al., Schiff-Based Metal Complexes
of Lamotrigine: Design, Synthesis, Characterization, and Biological Evaluation.
ACS omega, 2021. 6(11): p.
7719-7730.
99. Yasam, V.R., et al., A pharmacological overview of lamotrigine for the treatment of epilepsy. Expert review of clinical pharmacology, 2016. 9(12): p. 1533-1546.
Comments
Post a Comment