Skip to main content
Erschienen in: European Journal of Medical Research 1/2023

Open Access 01.12.2023 | Review

Repurposing approved non-oncology drugs for cancer therapy: a comprehensive review of mechanisms, efficacy, and clinical prospects

verfasst von: Roohi Mohi-ud-din, Apporva Chawla, Pooja Sharma, Prince Ahad Mir, Faheem Hyder Potoo, Željko Reiner, Ivan Reiner, Dilek Arslan Ateşşahin, Javad Sharifi-Rad, Reyaz Hassan Mir, Daniela Calina

Erschienen in: European Journal of Medical Research | Ausgabe 1/2023

Abstract

Cancer poses a significant global health challenge, with predictions of increasing prevalence in the coming years due to limited prevention, late diagnosis, and inadequate success with current therapies. In addition, the high cost of new anti-cancer drugs creates barriers in meeting the medical needs of cancer patients, especially in developing countries. The lengthy and costly process of developing novel drugs further hinders drug discovery and clinical implementation. Therefore, there has been a growing interest in repurposing approved drugs for other diseases to address the urgent need for effective cancer treatments. The aim of this comprehensive review is to provide an overview of the potential of approved non-oncology drugs as therapeutic options for cancer treatment. These drugs come from various chemotherapeutic classes, including antimalarials, antibiotics, antivirals, anti-inflammatory drugs, and antifungals, and have demonstrated significant antiproliferative, pro-apoptotic, immunomodulatory, and antimetastatic properties. A systematic review of the literature was conducted to identify relevant studies on the repurposing of approved non-oncology drugs for cancer therapy. Various electronic databases, such as PubMed, Scopus, and Google Scholar, were searched using appropriate keywords. Studies focusing on the therapeutic potential, mechanisms of action, efficacy, and clinical prospects of repurposed drugs in cancer treatment were included in the analysis. The review highlights the promising outcomes of repurposing approved non-oncology drugs for cancer therapy. Drugs belonging to different therapeutic classes have demonstrated notable antitumor effects, including inhibiting cell proliferation, promoting apoptosis, modulating the immune response, and suppressing metastasis. These findings suggest the potential of these repurposed drugs as effective therapeutic approaches in cancer treatment. Repurposing approved non-oncology drugs provides a promising strategy for addressing the urgent need for effective and accessible cancer treatments. The diverse classes of repurposed drugs, with their demonstrated antiproliferative, pro-apoptotic, immunomodulatory, and antimetastatic properties, offer new avenues for cancer therapy. Further research and clinical trials are warranted to explore the full potential of these repurposed drugs and optimize their use in treating various cancer types. Repurposing approved drugs can significantly expedite the process of identifying effective treatments and improve patient outcomes in a cost-effective manner.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
Akt
Protein kinase B
Bax
Bcl-2-associated X protein
Bcl2
B-cell lymphoma 2
BSE
Bovine spongiform encephalopathy
c-myc
Cellular myelocytomatosis
COX-2
Cyclooxygenase-2
CSCs
Cancer stem cells
CSCs
Cancer stem cells
CTGF
Connective tissue growth factor
DAPK
Death-associated protein kinase
DHFR
Dihydrofolate reductase
EGFR
Epidermal growth factor receptor
eIF4E
Eukaryotic translation initiation factor 4E
ER
Endoplasmic reticulum
FACT
Facilitates chromatin transcription
FDA
Food and drug administration
HDACs
Histone deacetylase
HER2
Human epidermal growth factor receptor-2
HIV
Human immunodeficiency virus
HUVEC
Human vascular endothelial cells
MCF-7
Michigan Cancer Foundation-7 cell line
MDA-MB-231
MD Anderson-Metastatic Brest-231 cell line
MMPs
Matrix metalloproteinases
mTOR
Mammalian target of rapamycin
MZ
Mebendazole
NF-Kb
Nuclear factor-kB
NIH
National Institute of Health
NO
Nitric oxide
NSAID
Non-steroidal anti-inflammatory drug
P53
Tumor protein TP53
PAK-1
Protein-activated kinases-1
PARP
Poly (ADP-ribose) polymerase
PCNA
Proliferating cell nuclear antigen
PSADT
PSA doubling time
PTB
Phosphotyrosine binding
ROS
Reactive oxygen species
Shc-PTB
Shc-Phosphotyrosine binding
SNEA
Sub-network enrichment analysis
SOD2
Superoxide dismutase 2
STAT3
Signal transducer and activator of transcription-3
TfR1
Transferrin receptor protein-1
TKIs
Tyrosine kinase inhibitors
TRAIL
TNF-related apoptosis-inducing ligand
VEGF
Vascular endothelial growth factor
XIAP
X-linked blocker of apoptosis
YAP1
Yes-associated protein-1

Introduction

Cancer is a serious disease that causes high mortality rates worldwide [13]. The occurrence of different types of cancer and particularly the possibilities of treatment are a big challenge for clinicians [49]. Malignant tumors such as liver cancer, breast, prostate, pancreas and colorectal cancer are generally difficult to cure at advanced phases with existing conventional treatments [1014]. Finding new substances for the treatment of cancers was the result of technological development and innovative approaches. Surprisingly, the research on novel agents for cancer treatment lasts long and is a complicated exercise due to the various steps necessary for the isolation, synthesis, and purification of new anticancer substances [13, 1518]. After drug discovery, synthesis and selection of appropriate formulation and preclinical pharmacology and toxicology, phase I clinical trials have to be made which begin with the administration of an investigational drug into healthy humans This phase involves the estimation of initial safety and tolerability, pharmacokinetics and assessments of pharmacodynamics. If successful, phase II is performed with the primary objective to explore therapeutic efficacy in patients. After that, for many of them, there is only a small probability to finish successfully phase III clinical trials which are designed to confirm the preliminary evidence obtained in phase II to verify that the potential drug is safe and effective for use in the intended indication and recipient patients. The amount of time and money for the production of new anti-cancer drugs is a big obstacle for the pharma industry for new drugs development, which will have proven therapeutic efficacy [1923]. Therefore, a novel concept of the use of old FDA-approved drugs is the recent direction to help clinicians and researchers, and, of course, the patients [24]. In this strategy, the advantage is that the assessment of drug safety, such as pharmacodynamics, pharmacokinetics, toxicity, and safety profiles, has been already previously established in Phase I studies. Moreover, old approved drugs can rapidly proceed for further Phase II clinical trials. Therefore, the interest is more and more focused on this strategy because of the lower cost and less time for developing new use of old drugs [19, 25].

Drug repurposing landscape: a brief synopsis

Drug development is a multistep process that includes identifying the therapeutic drug molecule that is clinically effective in the treatment of a disease [26]. This conventional drug discovery strategy implicates the de novo identification of new molecular entities. It has five stages, such as the discovery of the molecule and preclinical study, safety review, clinical studies, FDA review, and FDA post-market safety monitoring [27]. This process involves the identification of candidate molecules, synthesis, characterization, validation, optimization, screening, and assays for therapeutic efficacy [28]. When a product shows favorable outcomes in these studies, then the molecule has to go through a drug development process and subsequently testing in clinical trials [28]. De novo drug development is a time-consuming process and involves significant investments. It usually takes years of work (10–17 years) and costs millions of dollars. Furthermore, it is associated with high failure rates, with roughly 90% of molecules being rejected due to unexpected characteristics, such as safety and efficacy concerns [29, 30]. Despite massive expenditures in drug discovery and tremendous advancements in biological and informational technology over the past several decades, the number of new drugs brought to the phase of clinical trials has not expanded so much and remained relatively constant. Even though total research and development cost for drug discovery has expanded tenfold from 1975 (US $4 billion) to 2009 ($40 billion), there has been no substantial change in the number of new molecules approved since 1975 (in 2013 there were only 6 new drug moieties approved in comparison with the year 1976, where the number of newly approved pharmaceuticals was 27) [31]. The situation has improved in the last decade but not to a large extent. The increasing cost and time in the drug discovery process have resulted in the possibility that if resistance to the available drugs emerges, people with advanced diseases will end up dying before a substitute treatment option could become accessible [32]. Drug development is undoubtedly one of the most complex tasks in pharmaceutical research. In addition to already intimidating complications in pharmacological drug design, numerous obstacles occur due to regulatory, clinical intellectual property, and economic concerns. As a result of these challenging circumstances, the drug development process has become even more prolonged and uncertain. In pursuit of new treatment alternatives for patients with diseases, such as cancer, researchers have turned to drug repurposing tactics [33]. Drug repurposing also termed as drug repositioning, drug rescuing, drug reprofiling, drug recycling, or therapeutic switching, involves identifying and exploring the new therapeutic use of already FDA-approved and in clinical practice used drugs, but used for the treatment of other indications [24]. It is regarded as the most effective strategy in developing drug candidates using novel pharmacological properties and therapeutic characteristics of well-known drugs. Considering that traditional drug discovery is a time-consuming and costly process, the revolutionary strategy of drug repositioning is used to boost the success rate of medication development. Compared to the traditional drug discovery approach, this strategy is more favorable in terms of minimizing the length of time required for drug development while maintaining low costs, high efficiency, and minimal risk of failure [34]. The drug repurposing also offers a significant advantage not only in terms of the availability of preclinical information about the existing drug (a drug to be repurposed) but also provides additional data about clinical aspects such as pharmacokinetic, pharmacodynamics, and toxicity profile of that particular drug [35]. Because of this, these drugs might quickly be tested in Phase II and Phase III clinical trials, and the accompanying development costs could be significantly lower. The risk of failure is reduced, because in-vitro and in-vivo studies, toxicology profiles, chemical optimization, and formulation development have previously been explored. As a result, pharmaceutical companies have directed more and more of their attention to drug repurposing, since it might give a considerable advantage compared to traditional drug development (Fig. 1) [36]. In this context, it is not unexpected that approximately 30% of newly approved drugs in the United States are repurposed drugs [37].

Experimental drug repurposing approach

Binding assay

Techniques such as affinity chromatography, proteomics, and mass spectroscopy are used to identify novel targets for old drugs [38]. The protein target of gefitinib was investigated using HeLa cell extract. Mass spectroscopy results indicated that gefitinib could potentially interact with 20 different protein kinases that might be a target for gefitinib [27].

Phenotypic approaches

The phenotypic drug discovery approach is an experimental strategy that uses the library of accessible drug collections and focuses on finding their biological activities in cells and living organisms. It does not depend on the direct interaction with the target. Changes in in-vitro, in-vivo models and clinical studies can lead to the discovery of new drugs [39]. This is a screening procedure that does not presume the mechanism of action, and the primary output is a change in phenotype or physiological parameters. Cells, physiological systems, and whole organisms can all be used in this process. Depending upon the purpose or phase of drug development, each of these several systems can be used. Cell-based screening provides an increased insight into in vivo processes. On the other hand, in-vivo animal models help to assess the possible use of the existing medications for new phenotypic characteristics [40]. This method led to the discovery that astemizole and its metabolite desmethyl astemizole as effective inhibitors of Plasmodium falciparum growth and development [41].

Drug-centric approach

Drug-centric repurposing strategy is focused on forecasting new use for already approved drugs. This strategy relies heavily on substances that have the potential to interact with a wide range of targets (polypharmacological agents). Even though polypharmacological substances are responsible for triggering undesirable side effects, their activities can be used, because they offer the possibility of additional indications for a specific drug [42]. Polypharmacology seems to become the next major drug development paradigm. A considerable number of drugs are known for their ability to affect many targets simultaneously. Aspirin is used to relieve mild pain, fever and rheumatoid arthritis. It is also used as an anti-inflammatory agent in the treatment of Kawasaki disease and pericarditis. Transient ischemic attacks, ischemic stroke, myocardial infarction and even some types of cancer have all been successfully treated with this drug. Sildenafil, a phosphodiesterase inhibitor, was initially used for the management of erectile dysfunction. However, today, it is widely used for the management of pulmonary hypertension [43]. The majority of kinase inhibitors can inhibit several targets which makes them attractive options for the treatment of some types of cancer. Different multi-targeted tyrosine kinase inhibitors (TKIs) such as imatinib, nilotinib, and vandetanib were approved for clinical use in 2010 to treat solid cancers [44].

Target-based approach

The target-based drug repurposing approach involves the study of candidate drugs with biological targets such as receptors and proteins to different physiological responses to them. According to this method, new indications were discovered by relating a certain drug to a specific disease depending on the protein which it might target [42]. Proteins have many pathophysiological roles both in diseases and in healthy humans; dysfunctional, mutated, or misfolded proteins may trigger pathological responses that cause the development of a disease. The studies of proteins or biomarkers implicated in pathophysiological processes focus on target-based drug repurposing strategy [45]. The target-based strategy involves in silico or virtual high-throughput screening of drugs from various drug libraries or substances databases such as ligand-based screening or molecular docking proceeded by in vitro and in vivo high throughput or high content screening of drugs against a specific protein or biomarker of interest [27]. For instance, a new pharmaceutical molecule N-myristoyl transferase was discovered by this target-based approach for the treatment of filarial nematodes [46].

Knowledge-based

In this drug repurposing approach, models are developed that incorporate drug-related information, such as drug targets, chemical structures, route information, adverse effects, etc. These models are then used to anticipate unknown targets, biomarkers, or disease mechanisms [35].

Pathway or network-based

Pathway-based drug repurposing approach uses information about metabolic pathways, signaling pathways, and protein interaction networks to anticipate the similarity or relationship between a certain drug and a disease [35]. Network or pathway-based drug repurposing uses omics data to understand how drugs interact and communicate with disease targets. As a result, a specialized network with a few targets can be found in a vast network of pathways. Network-based strategies for drug repurposing are more and more in the focus of interest. Network-based computational biology focused on biomolecular interactions and omics data integration seem to be very promising. New drug repurposing research has discovered previously unknown signaling pathways in breast carcinoma subtypes [47, 48]. Kotelnikova et al. described a novel computational algorithm for the treatment planning of glioblastoma. The study analyzed gene expression data using a proprietary algorithm designed for pathway studio called sub-network enrichment analysis (SNEA). This method led to the discovery and FDA approval of fulvestrant (Faslodex1), a drug used to treat hormone receptor-positive metastatic breast carcinoma [49]. A study by Yu et al. suggested an approach for predicting potential drug–disease interactions that may be used for drugs or diseases that have or do not have associated genes. The adverse effects of drugs and disease symptoms were associated with identify drug–module and disease–module pairs using this strategy [50].

Repurposing of non-oncology drugs to treat cancer

Repurposing non-oncology drugs to use against cancer cells is an alternative approach to provide better mitigation possibilities for people with cancer at a lower cost and more quickly. Many methods were used to explore the probable anticancer function of non-cancerous drugs. For drug repurposing of non-oncology medications many in vivo and in vitro trials on pharmacological models and cancer cell lines were performed. Numerous wide-ranging electronic databases, such as the National Institute of Health (NIH) and Molecular Libraries Initiative [51], in which the chemical substances, biological evaluation assays, and genetic relevance of the active chemical substances were used to analyze them as a tool for utilization of drugs repositioning [52]. Repurposing of non-oncology drugs works through many mechanisms, such as cancer monotherapy, inhibiting proliferative signaling, inducing cell death, regulation of cellular metabolism, activation of antitumor immunity, drug combinatorial therapy, reactivating growth suppressors, interfering with replication, decreasing angiogenesis, suppression of invasion, and metastasis [32].

Anthelmintic drugs

Anthelmintic are a class of drugs that are used to treat unicellular protozoa as well as parasite worms in the intestine [53]. Anti-parasitic drugs such as mebendazole, flubendazole, albendazole, ivermectin, and chloroquine are commonly used antiparasitic drugs. Initially, these drugs were used to treat cattle parasites and then subsequently they were recommended for helminthiasis in humans as well. Many studies showed that some anthelmintic drugs have beneficial effects as anticancer agents on pathways, such as activator transcription proteins, signal transducer, and nuclear factor-kappa B (NF-kB) and Wnt/β-catenin (Table 1 and Fig. 2) [53]. Therefore, these anthelmintic drugs might be potential candidates as anticancer drugs.
Table 1
Drug repurposing for cancer therapy
Class of drug
Name of drug
Chemical structure of drug
Type of cancer cells tested
Mechanism/results
Refs.
Anthelmintic Antiprotozoal
Flubendazole
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Stra_HTML.gif
breast cancer, leukemia,
multiple myeloma, neuroblastoma colorectal cancer melanoma cells
↑Microtubule damage
↑ROS,
↑Cell cycle arrest in G2/M phase
↑Caspases 3, 7
↑Apoptosis
↑Cytotoxicity
↓Cancer cells growth
↓Metastasis
↓Resistance to anticancer drug trastuzumab
[5759]
Mebendazole
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strb_HTML.gif
breast cancer, prostate cancer
colon cancer
ovarian cancer
thyroid cancer
Synergistic effects with docetaxel
↓Polymerization of tubulin
↑ Cell cycle arrest in G2/M phase
↑Caspase-3
↑Apoptosis
↑Cytotoxicity
↓ Cancer cells multiplication
↓ Tumor growth
↓Metastases
[63, 65, 68, 144]
Niclosamide
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strc_HTML.gif
colorectal, breast, prostate and ovarian cancer
↑Cytotoxicity
↓Anaerobic metabolism, ↓glucose uptake in cancer cells
↓Signaling pathways associated with metastasis,
↓NF-κB,
↓Wnt/β-catenin, ↓STAT3
[145, 146]
Praziquantel
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strd_HTML.gif
colorectal
adenocarcinoma,
gastrointestinal cancers
↓XIAP
↓Anti-apoptotic proteins
↓Caspases
↑Apoptosis
Synergic effects with paclitaxel
[53]
Eprinomectin
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Stre_HTML.gif
prostate cancer
↑Apoptosis
↑Caspases 3, 9
↓ROS
↑Mitotic cell arrest in G1 phase
↑Translocation of β-catenin
[87]
Ivermectin
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strf_HTML.gif
colon, prostate, breast and gastric cancer
↓Cancer cells growth
↓AKT–mTOR
↓Wnt/β-catenin
↓PAK1
↓cyclin D
↓β-catenin
↓AKT/ERK//NF-kB
↓YAP1
↓CTGF
↓EGFR
[90, 147]
Nitazoxanide
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strg_HTML.gif
epithelial cancer cells
↑Apoptosis
↓c-MYC
↓mTOR
↑DNA fragmentation and damage
[148]
Clioquinol
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strh_HTML.gif
leukemic and myeloma malignant cells
↑Apoptosis
↓HDACs
↑Cell cycle arrest
↓p53, ↓p21
[95]
Chloroquine
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Stri_HTML.gif
pancreatic, liver cancer, cancer stem cells
breast cancer
↑Autophagy,
↓Janus kinase 2
↓DNA methylase 1
Synergistic effect in the combination with paclitaxel
↓Growth of cancer,
↓Signaling cascade of CXCL12/CXCR4
[149]
Antiviral
Ritonavir
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strj_HTML.gif
breast, pancreatic, ovarian and lymphocytic leukemia
↓Akt phosphorylation
↑Apoptosis
↓Progression of cancer cells
[150]
Nelfinavir
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strk_HTML.gif
ovarian, breast, lung cancer and liposarcoma
↑Apoptosis
↓Phosphorylation of Akt
↓STAT-3
↓Erk 1/2
[151]
Acyclovir
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strl_HTML.gif
breast cancer
↓Cell proliferation
↑Apoptosis
↑Caspase-3
↓ALDH
↑Proteins expression of E-cadherin,
↓Proliferation rate,
↓Tumor growth
[125]
Ribavirin
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strm_HTML.gif
human lymphocytes and human squamous cell carcinoma
↓Cyclin D1
↓Proteins cells,
↓elF4E and competing for guanylyl transferase
↓Translation of VEGF
↓mRNA, inhibit 5'-mRNA
[152]
Cidofovir
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strn_HTML.gif
glioblastoma and epithelial cells cancer
↑Apoptosis
↑PARP
↑Caspases
↑Cell cycle arrest in S-phase ↓DNA synthesis
[153]
Symbols: ↑increase, ↓decrease
Abbreviations: Akt protein kinase B, ERK extracellular signal-regulated kinase, NF-Kb Nuclear factor kappa B, mTor mammalian target of rapamycin, ALDH aldehyde dehydrogenase, MMP-9 MATZRIX metalloproteinase-9, Atg7 Autophagy-related E1 ligase 7, Bax Bcl-2-associated X protein, Bcl2 B-cell leukemia/lymphoma 2 protein, Bcl-XL B-cell lymphoma-extralarge, CAFs cancer-associated fibroblasts, CDC–CDK cyclin-dependent kinase 1, CK2 protein kinase CK2(casein kinase 2), c-MYC cellular myelocytomatosis oncogene, CTGF connective tissue growth factor, CXCL12/CXCR4 stromal cell-derived factor-1 (CXCL12) and chemokine (C–X–C motif) receptor 4 (CXCR4) cyclin D, DAPK death-associated protein kinase, DNA deoxyribonucleic acid, DR4/5 death receptor 4, EGFR epidermal growth factor receptor, elF4E eukaryotic translation initiation factor 4E

Flubendazole

Flubendazole is a well-known benzimidazole which is an antihelmintic drug that has also antineoplastic effects in different types of malignant diseases, including breast cancer, leukemia, multiple myeloma, and neuroblastoma [54, 55]. Flubendazole initiates significant changes in microtubule targeting sites, induction of apoptosis, induction of reactive oxygen species followed by G2/M phase accretion, and caspases 3, and 7 initiations in malignant cells. Flubendazole acts via different mechanisms such as inhibiting tumor growth, angiogenesis, etc. in pulmonary, liver, and breast cancer [56]. It inhibits trastuzumab resistance by targeting cancer cells, induction of apoptosis, and overexpression of human epidermal growth factor receptor 2 (HER2) in breast cancer [57]. This drug has cytotoxic activities in human colorectal cancer by blocking transcription proteins 3, signal transducer, and autophagy pathway [58]. It also blocks human melanoma cells' growth and metastasis and suppresses programmed cell death protein-1 and myeloid-derived suppressor cell accumulation [59].

Mebendazole

Mebendazole (MZ) is a drug that is frequently prescribed to manage gut parasitic infections. It inhibits tubulin polymerization which has an antiparasitic effect [60]. Mebendazole is a synthetic benzimidazole antihelmintic and a repositioned drug that has already proven its pharmacokinetics and toxicity profile [61]. MZ could be used in combination with temozolomide, which is a drug commonly prescribed in the treatment of malignant gliomas. In both xenograft and syngenic forms of glioma, this combination therapy suppressed tumor development more than temozolomide alone [62]. It also has synergistic effects with docetaxel inhibiting the polymerization of tubulin, mitotic arrest in the G2/M phase, augmenting apoptosis, reducing cell multiplication of prostate cancer, and suppressing tumor growth [63]. It stops mitotic growth in the G2/M phase, double-stranded breaks, and apoptosis in breast cancer which was shown using in vivo and in vitro biological assays [64]. Mebendazole activates the caspase-3 pathway and induces apoptosis. It also inhibits tumor development and stops pulmonary metastases in the later stages of thyroid cancer [65]. Mebendazole has also cytotoxic activity specific for colon cancer, ovarian cancer, endocrine malignancy, and brain tumors [62, 66, 67]. In cholangiocarcinoma, mebendazole induces apoptosis by inhibiting cell multiplication via increasing the expression of caspase-3 [68]. Other studies have found that MZ causes growth suppression in cell lines from many other different types of cancer ex-vivo and in vivo, especially pulmonary cancer [69], colorectal cancer [70], melanoma [71, 72], glioblastoma [62], and medulloblastoma [73]. The effects of MZ in melanoma are achieved by inducing apoptotic cell death, especially by activation of caspases, the pro-apoptotic Bcl-2, and suppression of the repressor of the apoptotic pathway, X-linked blocker of apoptosis (XIAP) [71, 72].
MZ has significant binding interaction potential in colon cancer cells, suggesting that it might be an antagonist of different kinases and oncogenes, such as ABL and BRAF [70], as well as a Hedgehog modulator in medulloblastoma [74]. MZ therapy in rodents caused a reduction in the size of tumors and decreased angiogenesis in comparison with normal animals. Furthermore, the incidence of metastasis in the therapy group was lower [67].
According to several case studies, MZ can also have antitumor effects in a clinical environment when given to patients with cancer [75, 76]. Mukhopadhyay et al. published one of the first studies on the antitumor effects of MZ [67]. It slowed down the growth of lung carcinoma cells but did not affect normal endothelium cells or fibroblasts. After the first- and second-line therapy in refractory tumors, MZ was given to patients with metastatic colorectal carcinoma. The patients had no adverse effects other than an increase in hepatic enzymes, and after that, the dose of the drug was reduced, and therapy efficiently eliminated practically all pulmonary and lymphovascular metastases, and partial recovery of hepatic metastases occurred as well [75]. MZ was prescribed to patients with adrenocortical cancer after the failure of different chemotherapeutics. During approximately one and a half years, there were no adverse effects, the size of the metastases decreased, and the illness was stable [76].

Niclosamide

Niclosamide is a drug of choice [77] and got approval from FDA as an anthelminthic drug. It has also been reported in a series of studies to have cytotoxic effects on ovarian cancer, breast cancer and prostate cancer cells [32, 78]. It targets cancer cells by interfering with the anaerobic metabolism and glucose uptake of these cells [79]. It also has an anticancer effect on multiple signaling pathways, such as metastasis, and signal activation, and as a transducer of transcription proteins Wntβ/-catenin and NF-KB [80, 81]. Some studies showed that it significantly suppressed the development of cancer of breast, liver, and colorectal cancer. Its anti-metastasis effect seems to prevent liver metastasis of colorectal cancer cells. It also has a beneficial effect on pulmonary metastases of breast cancer [82, 83]. However, less bioavailability and the poor solubility of the drug is the biggest obstacle to its clinical development [84]. The intravenous route might help to use the development of this drug as a repositioned drug [85, 86].

Praziquantel

Praziquantel is a broad-spectrum antiparasitic drug, and its mechanism of action is still unclear. Nevertheless, it can intensify the concentration of intracellular Ca2+ and it also causes contractions of muscles [53]. It has been shown that this drug also increases the cytotoxic action of paclitaxel at 20–40 µM. The combination of both drugs, i.e., praziquantel and paclitaxel, acts synergistically reducing the expression of anti-apoptotic protein and X-linked inhibitor of apoptosis protein (XIAP) [53].

Eprinomectin

Eprinomectin has a broad spectrum of effects against different parasitic infections. It showed also a cytotoxic potential against prostate cancer cells. It induces apoptosis in PC-3 cells by affecting reactive oxygen species (ROS). Eprinomectin also causes the stop of mitotic cells at the G1 phase. Moreover, this drug stimulates the translocation of β-catenin and has a significant apoptotic effect and activates caspase-3 and caspase-9. The abovementioned findings might explain why eprinomectin could have cytotoxic effects against advanced prostate cancer [87].

Ivermectin

Ivermectin is an FDA-approved macrocyclic lactone. It has antitumor effect in smaller doses against the Wnt-TCF, and β-catenin, and suppresses the expression of cyclin D in cancer of the colon [88]. Another approach documented the therapeutic effect of this drug in cancer xenografts and melanoma [89]. Reports on neurofibromatosis tumor cells and ovarian cancer cells suggested that this drug might suppress the growth of protein-activated kinases (PAK-1) and thus have a beneficial effect on prostate, breast, and gastric cancers [90]. This drug tends to reduce the growth of a tumor enhancing mitochondrial biogenesis compared to normal cells [91]. Similarly, in another research, it has been shown that this drug also inhibits PAK1 via AKT–mTOR in breast cancer. It reduces yes-associated protein (YAP1) expression and targets connective tissue growth factor (CTGF) in gastric cancer. It also suppresses resistance by dropping p-glycoprotein due to inhibition of ERK/AKT/NF-kB and epidermal growth factor receptor (EGFR) [92]. This drug is a self-renewal marker of cancer stem cells (CSCs) in breast cancer. Therefore, this drug could be a future therapeutic molecule for cancer management [93].

Nitazoxanide

Nitazoxanide is a thiazole-based molecule and it has been reported that it might have antitumor effects due to different mechanisms, such as inhibition of cellular myelocytomatosis (c-MYC), induction of apoptosis, and DNA fragmentation. It also causes autophagy against epithelial tumor cells via mechanistic targeting rapamycin (mTOR) inhibition besides its antihelmintic effects. Similarly, this drug can induce apoptosis by DNA fragmentation condensation of the nucleus along with its anti-parasitic effects [53, 94].

Clioquinol

Clioquinol also seems to have anticancer effects besides its antiparasitic effects. It causes downregulation expression in histone deacetylase (HDACs) in leukemic and malignant myeloma cells. This drug causes apoptosis via mitotic arrest and downregulation of HDAC, resulting in the expression of p53 and p21 [95].

Pyrimethamine

Pyrimethamine is an antiparasitic drug which inhibits tumor growth and metastasis in pulmonary carcinoma by attacking the dihydrofolate reductase (DHFR) and thymidine phosphorylase (TP) [96, 97]. Pyrimethamine, which is a STAT3 antagonist, has chemotherapeutic and immune-stimulatory effects in breast cancer models in mice. Pyrimethamine suppresses STAT3 action in metastatic breast cancer cell lines ex-vivo by reducing tumor growth and invasion and by increasing Lamp1 production in tumour-infiltrating CD8+T lymphocytes [98]. Pyrimethamine also inhibits the growth of ovarian cancer cells both ex-vivo and in-vivo [99].
The most representative mechanisms of anthelmintic drug repurposing in cancer therapy are summarized in Fig. 2.

Antiviral drugs

The combinations of antiviral drugs and conventional chemotherapeutic agents are used to treat different types of malignant diseases such as lymphoma, nasopharyngeal carcinoma, hepatocellular carcinoma, and Kaposi sarcoma using protease inhibitors directed towards the human immune-deficiency virus. Studies on the viruses accompanying cancers can be a useful platform for the development of novel therapeutic approaches not only for treating viral infections but also consequently influencing tumorigeneses [100].

Ritonavir

Ritonavir is a thiazole-based anti-viral drug widely used for the treatment of different viral diseases such as HIV (human immunodeficiency virus) and to increase the effectiveness of protease inhibitors. Several studies have shown antitumor effects of ritonavir. It seems that it can cause apoptosis and inhibit the progression of malignant cells in breast, pancreatic and ovarian carcinoma [101103]. The drug also strengthens the effects of several substances such as temozolomide towards glioma cancer cells [104]. It seems that it could also be used in combination with bortezomib for the treatment of renal cancer [105]. Ritonavir also has anticancer effects against breast cancer by inhibiting Akt phosphorylation and seems to be effective also in lymphocytic leukemia [106].

Nelfinavir

Nelfinavir is a protease inhibitor and is widely used in managing HIV-1 and HIV-2. This drug can decrease phosphorylation of Akt, signal transducer and activator of transcription 3 (STAT-3), and xenografts tumors [107] Several studies reported the beneficial effects of this drug against different types of cancers such as ovarian, breast, lungs, and liposarcoma by inhibiting the signals of Erk 1/2, STAT-3, and Akt [108113]. It has been reported that nelfinavir induces endoplasmic reticulum (ER) stress and also increases the effect of other drugs used for the management of prostate and breast cancers [114, 115]. Numerous studies showed that it inhibits autophagy and cyclooxygenase (COX-2) inhibitors in breast cancers [116]. The mixture of COX-2 and nelfinavir inhibit autophagy and could increase cytotoxic effects directed against cancer of the breast [116]. It also induces pro-apoptotic effects by activating caspase-4 [117, 118]. Some studies found that this drug induces apoptosis mediated by ROS [119, 120]. Other studies also reported antiproliferative effects and limited toxicity against liposarcoma and cystic carcinomas [120, 121].

Acyclovir

Acyclovir was discovered about four decades ago and has become a drug of choice against viral infections, such as Herpes Simplex [122]. A plethora of evidence suggests that this drug is effective in the treatment of different types of cancers [123, 124]. This drug suppresses cell proliferation and induces apoptosis in malignant tumors of of the breast. Another study reported antiproliferative effects of acyclovir in Michigan Cancer Foundation (MCF-7) cell lines by increasing the proteins expression of E-cadherin, reducing the proliferation rate, and increasing apoptosis caspase-3 and wound healing in MCF-7 malignant cells. The results suggested that this drug could be used alone or in combination therapy as a potential candidate for breast cancer treatment. The results of the calorimetric assay indicated the downregulation of aldehyde dehydrogenase (ALDH) activity towards breast cancer cell lines [125].

Ribavirin

Ribavirin is a member of the antiviral drugs class and it seems that it has beneficial effects in different viral diseases, such as polio, hepatitis-C and SARS–coronavirus infection [126128]. It is a guanosine ribonucleoside-based drug. Since it is similar to guanosine it tends to compete for guanylyl transferase and inhibit 5'-mRNA. This drug at micromolecular concentrations binds with eukaryotic translation initiation factor (elF4E) on a purposeful site using a 7-methyl guanosine mRNA cap. This drug suppresses elF4E facilitated oncogenes transformation using 7-methyl guanosine. In another approach, it has been found that it can inhibit elF4E competitive binding to cyclin D1 and thus reduce cyclin D1 protein cells [124]. This drug also causes the translation of VEGF mRNA and other genes [129, 130]. Ribavirin at 2 µm concentration suppresses the progression of human lymphocytes [131].

Cidofovir

Cidofovir is a FDA accepted nucleoside broad-spectrum antiviral drug which is used to treat different viral infections by inhibiting the viral DNA polymerase through its metabolite diphosphate [132134]. Because of this mechanism, the drug can decrease the growth of several types of human cancers [135139]. This drug also induces apoptosis mitotic arrest in S-phase and causes activation of Poly (ADP-ribose) polymerase (PARP) and caspase in epithelial cells [140142]. It also inhibits DNA synthesis and blocks the growth of cancer cells. It has also been reported that this drug has cytotoxic effects on human glioblastoma cell lines. This drug shows antiproliferative effects both in xenograft models and in vitro studies. It inhibits the gene expression associated with apoptosis in glioblastoma. This drug has antitumor effects because of its different mechanisms, including effects on mitogenic pathways and activation of proapoptotic pathways in glioblastoma [143].

Antibiotics

Sulfisoxazole

Sulfisoxazole is an antibiotic that may inhibit small extracellular vesicular exudation from cancerous mammary cells by interfering with endothelin receptor A. In animal studies of breast cancer xenografts, sulfisoxazole had antitumor and antimetastatic effects (Table 2) [154].
Table 2
Antibiotics, antifungals, antimalarial, anti-inflammatory as potential drug candidate against cancer
Class of Drug
Name of drug
Structure
Type of cancer to be used
Mechanism
Refs.
Antibiotics
Sulfisoxazole
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Stro_HTML.gif
Breast cancer
Interfering with endothelin receptor A to stop breast cancer cells from exuding tiny extracellular vesicles
[154]
Azithromycin
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strp_HTML.gif
Colon cancer
↑TNF-α-related apoptosis
↑TRAIL
↑DR4/5
↓Autophagy
[155]
Doxycycline
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strq_HTML.gif
Osteosarcoma, prostate carcinoma, myeloid and colon cancer
In myeloid and colon cancer, it prevents permeation by lowering MMP-2 and MMP-9
[166]
Anthracycline
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strr_HTML.gif
Endometrial, breast, bladder, hepatic, thyroid, and pulmonary malignancies
Attaches to DNA, causing it to get alkylated, which stops the cell cycle
[182]
Antifungals
Itraconazole
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strs_HTML.gif
Non-small cell lung cancer
By eliminating lipids from the plasma membrane, it decreases AKT1 activity, which inhibits its downstream target mTOR, resulting in mortality and growth slowering
[188]
Rapamycin
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strt_HTML.gif
Breast cancer
It promotes intracellular autophagy and boosts the function of Atg7 and DAPK via transcriptional activation
[196]
Griseofulvin
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Stru_HTML.gif
Colorectal and cervical cancers
Affects microtubule assembly in MCF-7 cells, causing programmed cell death
↑Cell cycle arrest
[199201]
Clotrimazole
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strv_HTML.gif
Breast, colon and pulmonary cancer
Blockes actin polymerization and activates glycolytic inflow
[203]
Ciclopirox
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strw_HTML.gif
Breast, colorectal cancer rhabdomyosarcoma
↓CDC–CDK,
↓Bcl-XL
↑Caspase-dependent cascade causing apoptotic cell death
[208, 209]
Nannocystin A
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strx_HTML.gif
Colorectal and breast cancer cells
Target eukaryotic elongation factor 1 in proteome investigations
[213, 214]
NSAIDS drugs
Aspirin
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Stry_HTML.gif
Hepatocellular carcinoma
Affects P4HA2 by suppressing NF-κB and LMCD1-AS1/let-7g of Aspirin prevents tumour growth and accumulation of collagen
[313, 314]
Ibuprofen
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strz_HTML.gif
Adenocarcinoma
Modulates the expression levels of cancer-related genes Akt, P53, PCNA, Bax, and Bcl2
[228]
Naproxen
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Straa_HTML.gif
bladder carcinoma
↑Cell arrest
↑Cancer cells death
↑PI3K
[233]
Diclofenac
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strab_HTML.gif
Ovarian cancer
↑Apoptotic cell death
↓SOD2
↓Proportion of free radicals
[243, 244]
Celecoxib
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strac_HTML.gif
Bladder cancer
Blocks epithelial-to-mesenchymal transformation
↓miRNA-145/TGFBR2/Smad3 axis
[250]
Indomethacin
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strad_HTML.gif
Colon cancer
↓Cancer cell proliferation
↓PKC-p38-DRP1
↓Wnt/-βcatenin signalling, to effectively target MAPK mechanisms
[259, 263]
Thiocolchicoside
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strae_HTML.gif
Leukaemia, lymphoma, and squamous cell carcinoma
Blocks the receptor stimulator NF-kB ligand
↓NF-kB signalling cascade
↓Cancer-induced bone metastasis
[308, 309]
Artemisinin
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Straf_HTML.gif
Breast cancer
Deactivates cancer-related fibroblasts and decreases CAFs mediating growth and metastases by suppressing TGF-β signalling
[315]
Artesunate
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strag_HTML.gif
Hepatocellular carcinoma
↑Pro-apoptotic proteins
↑caspases
↓MYC oncogene
↓Anti-apoptotic proteins
[272, 273]
Dihydroartemisinin
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strah_HTML.gif
Ovarian cancer
↓Cancer cell development
↓Metastases by addressing the platelet-derived growth factor receptor-alpha (PDGFR)
[316]
Mebendazole
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strai_HTML.gif
Melanoma
↑Apoptotic cell death
↑Caspases
↑Bcl-2
↓Repressor of apoptosis X-linked blocker of apoptosis (XIAP)
[71, 72]
Chloroquine
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Straj_HTML.gif
Metastatic tumors
Par-4-dependent suppression, mediates p53- and Rab8b-based Par-4 production to promote tumour cell death
[279]
Pyrimethamine
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Strak_HTML.gif
Non-small cell lung cancer
↓EMT
↓Invasion
↓Cancer cells growth, ↓Metastases by interacting with dihydrofolate reductase and thymidine phosphorylase
[96, 97]
Quinacrine
https://static-content.springer.com/image/art%3A10.1186%2Fs40001-023-01275-4/MediaObjects/40001_2023_1275_Stral_HTML.gif
Renal cancer
Facilitates chromatin transcription (FACT) protein complex, which seems to be trapped on chromatin and induces CK2-induced phosphorylation of p53, responsible for quinacrine-mediated p53 transcription
[299, 300]
Symbols: symbols: ↑increase, ↓decrease
EMT epithelial-to-mesenchymal transition, Erk ½ extracellular signal-regulated kinase 1/2, HDACs histone deacetylase inhibitors, M Phase mitosis, m RNA messenger ribonucleic acid, MAPK mitogen-activated protein kinase, MCF-7 michigan cancer foundation-7, miRNA microRNAs, MMP-2 matrix metalloproteinases-2, MYC master regulator of cell cycle entry and proliferative metabolism, NF-kB nuclear factor kappa-light-chain-enhancer of activated B cells, P4HA2 collagen prolyl-4-hydroxylase α subunit 2, PARP poly ADP ribose polymerase, PCNA proliferating cell nuclear antigen, PDGFR platelet-derived growth factors, PI3K phosphoinositide 3-kinases, ROS reactive oxygen species, SOD2 superoxide dismutase 2, STAT-3 signal transducer and activator of transcription 3, TGFBR2 transforming growth factor-beta, TRAIL TNF-related apoptosis-inducing ligand, VEGF vascular endothelial growth factor, XIAP X-linked inhibitor of apoptosis protein, YAP1 yes-associated protein 1

Azithromycin

Azithromycin is a macrolide antibacterial drug. The proliferative potential of cancer cells has been inhibited by this drug. In colon carcinoma cells, ex-vivo and in-vivo, it increases the antineoplastic effectiveness of TNF-α-related apoptosis-inducing ligand (TRAIL) by suppressing autophagy and increasing DR4/5 [155]. Azithromycin also reduces angiogenesis in pulmonary carcinoma by inhibiting vascular endothelial growth factor receptor 2-induced focal adhesion and the PI3K/AKT signalling cascade [156].

Doxycycline

Doxycycline is a tetracycline antibiotic that is used for treating different infections. Some tetracyclines were reported to suppress angiogenesis in the early 1990s [157], and doxycycline was eventually found to have antiproliferative activity in bone and prostate carcinoma and mesothelioma cells [158160]. It has also been shown to induce apoptosis in pancreatic islets [161, 162] and myeloid cells [163]. Matrix metalloproteinases (MMPs) are inhibited by tetracyclines [164]. Doxycycline therapy inhibits penetration by downregulating MMP-2 and MMP-9 levels in myeloid cells [165] and colon carcinoma [166]. Doxycycline has been investigated as a suppressor of tumour progression, because MMPs are assumed to play a crucial role in cancer infiltration and progression. Doxycycline has been shown to reduce different tumours and it seems to be beneficial for breast cancer patients at risk for osteolytic bone metastasis. [167]. The same investigators also demonstrated that zoledronic acid, a medicine used to decrease the incidence of bone fractures in patients with osteosarcoma or osteoporosis, might be useful in combination with doxycycline [168]. MMP-2/9 suppression has also been shown to reduce metastasis in preclinical trials of prostate cancer [169] and squamous skin carcinoma [170]. Doxycycline can reduce EMT-marker transcription in pulmonary and hepatic carcinoma cells, reversing their pro-metastatic character [171, 172]. Doxycycline therapy reduced clonogenic potential and decreased the expression level of stem cell markers in hepatic carcinoma cells enriched with stem-related characteristics. Doxycycline decreased proliferation markers Ki67 and PCNA in the in vivo xenograft mouse model. [173]. Doxycycline was combined with interferon-alpha (IF-α) treatment in a phase II clinical study of renal cancer metastases. VEGF levels were measured to see if there was an antiangiogenic effect. The combined therapy proved to be ineffective in patients with renal carcinoma metastases, despite modest early reduction of VEGF expression in some patients [174]. The efficacy of doxycycline therapy in combination with bone-targeting medicines was evaluated in a recent phase II study in females with breast carcinoma metastases. In this trial, doxycycline was shown to have a negligible effect but it was associated with severe harmful effects [175].

Ionophore antibiotics

Ionophore antibiotics have shown antitumor effects against cancers of the colon, and prostate, as well as endometrial, blood, cerebral, and bone malignancies. Ionophore antibiotics salinomycin and nigericin specifically attack CSCs and it seem that they are slightly more effective than paclitaxel. Cell migration, metastases, and the GTPase K-Ras cascade are all targeted by these drugs. Salinomycin inhibited the hedgehog and WNT/-β catenin pathways, resulting in decreased tumour size of metastatic breast cancer [176]. In lymphomas, salinomycin combined with doxorubicin was reported to have a synergistic effect [176]. Rapamycin, an antimicrobial drug, has an antiproliferative effect by blocking cell cycle progression due to its effect on CDK proteins and mTOR signalling. In vivo investigations have shown that it can also prevent cancer and malignancies caused by the Epstein–Barr virus [177]. Rapamycin has chemotherapeutic potential in different types of human cancers, and it was found to synergize with erlotinib in NSCLC and paediatric glioma [177, 178].

Anthracycline drugs

Anthracycline drugs have also been intensively studied as antimalignant drugs. Doxorubicin, idarubicin, mitoxantrone, daunorubicin and epirubicin are often used anthracycline medicines that have been extensively investigated in solid and blood malignancies [179181]. Garg et al. found that doxorubicin and selinexor promoted the death of thyroid cancer and AML cells when used together [180, 181]. Duocarmycin is an antibiotic that works at low doses. It attaches to the DNA molecule causing DNA to be alkylated, which causes the cell cycle arrest. It is one of the most successful medications used to treat endometrial, breast, bladder, hepatic, thyroid, and pulmonary malignancies [182]. Wang et al. analyzed 124 patients from January 1996 to July 2018 to analyze the effects of chemotherapeutic treatment with gemcitabine and anthracycline (epirubicin and pirarubicin). They analyzed the probability of tumour’s recurrence and therapy failures. Gemcitabine had a lower recurrence percentage and therapy failure rates than anthracycline antimicrobials, suggesting that this approach should be explored for patients who cannot be treated with BCG [183]. Landomycin E is an angucycline antibiotic produced by Streptomyces globisporus which caused cell death in T-cell leukaemia cells by rapidly generating hydrogen peroxide and activating caspases [184]. NAC, in combination with doxorubicin, demonstrated decreased adverse effects on nephrons, somewhat enhanced cytotoxic effects of T-cells, and it slightly enhanced their survivability. NAC, in combination with landomycin, on the other hand, significantly enhanced the lifespan of the rodents while also having some tissue-protective effects. As a result, NAC in combination with landomycin appears to be more effective than doxorubicin. Landomycin seems to be more powerful than doxorubicin at low concentrations in ex-vivo and in-vivo melanoma with fewer adverse effects. However, adverse effects such as cardiovascular events and mucositis suggested the need for other antibacterial drugs. Idarubicin, a doxorubicin analogue, was created. In acute myelogenous leukaemia, idarubicin showed increased lipophilicity and anti-malignant efficacy. In acute myeloid leukaemia clarubicin, an anthracycline antibiotic derived from Streptomyces galilaeus, suppresses RNA production. Amrubicin has chemotherapeutic efficacy against SCLC, lymphoma cells and bladder carcinoma and got marketing authorization in Japan. Zorubicin which is a benzoylhydrazone derivative of the well-known anthracycline antineoplastic antibiotic daunorubicin is at the moment in the confirmatory phase of clinical studies for breast cancer and leukaemia.

Antifungals

Itraconazole

Itraconazole is an antifungal drug that works by inhibiting 14-α-lanosterol demethylase (14-LDM), a crucial enzyme in cholesterol production. Itraconazole also decreases angiogenesis in endothelial cells by reducing endothelial growth [185] and suppresses VEGFR-2 levels and multiple cellular pathways in endothelial cells [186]. Treatment with itraconazole of human vascular endothelial cells (HUVECs) inhibited movement and tube creation by decreasing the phosphorylation of growth factor receptors [187]. Itraconazole therapy of pulmonary carcinoma xenografts reduced angiogenesis and regression in this study. Its antitumor activity may be mediated by different pathways together with its antiangiogenic effects. In glioblastoma cells, removing lipids from the plasma membrane reduced Akt-1 action, which inhibited its downstream target mTOR, causing death and decreased tumor growth [188] (Fig. 3). It has been hypothesized that itraconazole might be a Hedgehog antagonist. In mice treated with itraconazole the proliferation of two hedgehog-based tumor types, a medulloblastoma and a basal cell cancer of the skin, was reduced in vivo [189]. Research on pleural mesothelioma cells came up with similar results [190]. Itraconazole was to a certain extent successful in a randomized clinical study with metastatic castration-resistant prostate carcinoma, with an extended PSA progression-free life, and suppression of Hedgehog signalling [191]. Itraconazole was also used to treat 19 patients with basal cell malignancy in a limited phase II study. Hedgehog signalling was decreased which was followed by slower tumor growth and tumor regression. In a minor phase II trial, combining itraconazole with conventional anticancer therapy for lung cancers improved both progression-free and ultimate survival. The authors of this study speculated that this result could be attributed to the antiangiogenic effects of itraconazole [192]. Nevertheless, there might be some problems with itraconazole treatment of malignant diseases. According to some studies, antifungal medications may affect the effects of other anticancer drugs, particularly antibodies, such as rituximab [193].

Rapamycin

Rapamycin, also called “sirolimus,” is a drug that was first discovered for its potent antifungal effects [194]. Because of its unique immunosuppressive effects, rapamycin has been routinely used to prevent rejection after organ transplantation. Rapamycin has relatively recently been identified as an mTOR inhibitor that may be used to treat Kras Pten endocrine ductal adenocarcinoma, resulting in inhibition of proliferation and tumor size shrinkage [195]. By transcriptional activation of Atg7 and DAPK, rapamycin was also used to activate cellular autophagy and increase the chemotherapeutic effects of dihydro-artemisinin in breast tumor cells [196]. Due to the increased expression of p73, rapamycin improves the susceptibility of ER-positive breast cancer cells to tamoxifen [197].

Griseofulvin

Griseofulvin causes apoptotic cell death in lymphoma and leukemia cells. [198]. It affects microtubule assembly in MCF-7 cells causing apoptotic death and cell cycle arrest, and it has a stimulatory effect together with vinblastine [199]. The same chemotherapeutic effects have been observed in colorectal and cervical cancers [200, 201]. Centrosome clumping has also been associated with the production of micronuclei in prostate carcinoma. In pulmonary and prostate cancer, combining radiation with griseofulvin therapy showed synergistic anticancer effects. The sulfonyl group substitution analogues of griseofulvin have antiproliferative effects on oral cancer cells and cytotoxic activity on breast cancer cells [202].

Clotrimazole

Clotrimazole inhibits glioblastoma cell invasion and metastasis. It inhibited actin polymerization and promoted glycolytic inflow in breast, colon, and pulmonary cancer cells [203]. Kadavakollu et al. [204] evaluated many potential anti-cancer pathways. In prostate and cervical cancers, as well as lymphoid malignancies, ruthenium combined with other drugs, had stronger cytotoxic activity than the monotherapy with individual drugs [205]. A combination of imatinib and clotrimazole inhibited the glycolysis pathway more effectively and boosted NO and VEGF production in breast cancer cells [206].

Ciclopirox

Ciclopirox (CPX), an antifungal drug, induced ageing in p53 deficient HeLa cells by a mechanism unrelated to mTOR [207]. Due to degrading CDC–CDK, downregulating Bcl-xL, and activating the caspase-dependent cascade, it causes apoptotic cell death in breast and colorectal cancer, and rhabdomyosarcoma cells [208, 209]. Prolonged exposure to this drug caused p53-independent caspase stimulation and cell death [207]. It has also been demonstrated that it suppresses HPV genetic mutations. By activating oxygen radicals, caspase-3, and lowering Bcl-xL levels, CPX was significantly more effective than gemcitabine in endocrine carcinoma. Nevertheless, when it came to triggering apoptotic pathways, the combination of these drugs was significantly more efficacious than each of these medicines alone [210]. According to ex-vivo and in-vivo studies in colorectal cancer, CPX causes autophagy through the depletion of DJ-1 and the formation of oxygen radicals [211]. According to Ahmad et al. ethacrynic acid and CPX had an additive anticancer effect in hepatocellular carcinoma. However, low concentrations of CPX were harmful not just to cancer cells but also to healthy cells [212].

Nannocystin A

Nannocystin A showed chemotherapeutic effects on colorectal and breast cancer cells. Nannocystin A has been shown to target eukaryotic elongation factor 1 in proteome studies [213, 214]. These findings support the repositioning of an antifungal drug with anticancer potential. In future, additional clinical testing is needed to confirm this.

NSAIDS/anti-inflammatory drugs

Aspirin

Aspirin is a drug that has effects on cyclooxygenase (COX) isoenzymes 1 and 2 and is widely used in the treatment and prevention of myocardial infarction in patients with coronary heart disease. COX-1 is important for platelet synthesis of thromboxane A2, which results in platelet aggregation and adhesion to cells, including malignant cells. Platelets covering tumor cells prevent the immune system from recognizing these cells, favoring the development and spread of cancer. COX-2 is important in the production of prostaglandin E2, which significantly stimulates the growth of tumor cells [215217]. Recent pharmacological studies have analyzed the potential of aspirin as a therapeutic approach in spontaneous or chemically provoked tumors [218220]. Many clinical trials have found that taking aspirin after a diagnosis is associated with a better prognosis in patients with colorectal cancer [221224]. However, because most of these studies were retrospective and the patient recruitment was not homogenous, there is a lot of contradictory information. To determine the function of aspirin as a potential anticancer therapy, prospective trials are required. Several clinical trials are now in progress, the majority of which are looking at the effect of aspirin in preventing relaps of the illness.

Ibuprofen

Ibuprofen is a non-selective cyclooxygenase inhibitor. It slows down the development of prostate carcinoma [225]. It also has a radio-sensitizing effect ex-vivo but at larger doses than those that have been documented to suppress eicosanoid production, indicating that other pathways are included [226, 227]. Anti-angiogenesis, initiation of apoptosis, and decrease of cellular proliferation were found to have antitumor effects on gastric adenocarcinoma cells ex-vivo, along with modulating the expression levels of the cancer-related genes Akt, PCNA, Bax, P53, and Bcl2 [228] (Fig. 3). It has been shown that TNF-α upregulated metastatic melanoma cell migration in vitro and that this could be reduced by ibuprofen both in solution and delivered from a hydrogel. Although this might be attributed to the induction of apoptotic cell death, the mechanism of this is still not completely explained [229, 230]. Chemosensitivity could also be modulated by ibuprofen. Ibuprofen therapy reduced the amounts of Hsp70, a heat shock protein associated with apoptotic tolerance in lung carcinoma cells. Following ibuprofen therapy, blocking Hsp70 and inducing apoptosis improved responsiveness to the anticancer drug, cisplatin [231].

Naproxen

Naproxen is a propionic-acid analogue that reduces cell growth, provokes programmed cell death, and restricts metastasis [232]. This drug is a non-selective cyclooxygenase inhibitor. Ex-vivo and in-vivo chemotherapeutic effects have been documented in breast, leukemic, bladder, colorectal, and osteosarcoma cells. Naproxen causes the death of bladder carcinoma cells ex-vivo by targeting PI3K [233, 234]. A combination of cholesterol-lowering drug atorvastatin and naproxen effectively suppressed colon adenocarcinomas in experimental animals in-vivo [235]. A combination of calcitriol and naproxen has been tested in phase II clinical trials for preventing the relapse of prostate carcinoma. Such a combined therapy has been well-tolerated, with 19% of enrolled participants having a reduction in PSA doubling time (PSADT) and 67% having a prolongation of PSADT when compared with baseline [236].

Diclofenac

Diclofenac is an acetic acid derivative with a modest affinity for cyclooxygenase-2. It has been shown that this drug has beneficial effects on several malignant tumors including fibrosarcoma, hepatoma, colorectal, endometrial, and endocrine carcinoma. The effect of diclofenac (3%) and calcitriol was shown on different carcinoma cancer cell types, such as endometrial, breast, cerebral, colorectal, endocrine, non-small cell lung cancer, hepatic, and showed a higher cytotoxic effect in cells from chronic lymphocytic leukemia than in normal lymphocytes[237, 238]. Furthermore, growth rates and degree of vasculature were significantly decreased in experiments on rats with fibrosarcoma and hepatoma [239]. Diclofenac also has an anticancer effect in colon carcinoma [240, 241]. Diclofenac also reduced tumor growth in a mouse model of pancreatic cancer [242], as well as in ovarian cancer [243]. In vitro evidence suggests that diclofenac therapy induces apoptotic cell death by inhibiting antioxidant SOD2, resulting in a greater proportion of free radicals [244]. Despite the increasing amount of data supporting the anticancer effects of diclofenac, there are at the moment no active clinical studies evaluating the effects of diclofenac as a chemotherapeutic agent. A phase II clinical study for basal cell carcinoma, on the other hand, was just completed. Diclofenac was tested as a stand-alone treatment and in combination with calcitriol. The study found that diclofenac applied topically was more successful than combination therapy, with complete histologic tumor regression in 64.3% [245].

Celecoxib

Celecoxib, a specific COX-2 blocker, has chemotherapeutic effects in different types of cancer. In randomized controlled studies celecoxib combined with chemotherapeutic treatment has beneficial effects on breast cancer, progressive pulmonary carcinoma, and transitory bladder cancer [246249]. Through miRNA-145/TGFBR2/Smad3 axis, celecoxib suppresses the epithelial-to-mesenchymal shift in cancer bladder cells [250]. Celecoxib decreases liver cancer cells proliferation and metastasis by addressing PNO1 and reduces AKT/c-Met-induced hepatocarcinogenesis by inhibiting COX-2/Akt/FASN pathway [249, 251, 252]. Celecoxib has effects on proline metabolism, generating an upregulation in proapoptotic markers (PRODH/POX, PPAR), lowering HIF-1 levels, and triggering squamous skin carcinoma programmed cell death. In human oral cancer cells, combination therapy with celecoxib and calyculin-A suppresses epithelial–mesenchymal shift [253, 254].

Indomethacin

Indomethacin is an antinociceptive non-steroidal anti-inflammatory drug (NSAID) widely used in the management of rheumatoid diseases [255]. It has been noted that patients who were treated for a long period with NSAIDs had a decreased chance of acquiring cancer which was confirmed in several clinical studies [255]. Furthermore, there is a growing number of publications suggesting that indomethacin and indomethacin-dependent prodrugs have chemo-preventive effects against different malignant diseases by inhibiting COX-1/2-associated angiogenesis [256, 257]. Indomethacin inhibits cancer cell progression by competing with calcium-related signalling and the creation of focal interactions [258]. A COX-independent mode of activity for indomethacin's antiproliferative effect has been discovered in several studies, as indicated by cell growth suppression in indomethacin-treated colonic cancer cells that would not exhibit COX-1/2. [259261]. Lin et al. relatively recently proposed that the chemotherapeutic activity of indomethacin might be due to MAPK-associated pathway suppression [262]. They used computational scanning to perform this drug repurposing using a current drug repository. Indomethacin was proven to have a stronger association with ShcPTB by interacting with the phosphotyrosine binding (PTB) region of adaptor protein Shc (ShcPTB) as a readout. It seems that indomethacin competes against active EGFR by interacting with ShcPTB without disrupting the ERK-binding region, preventing EGFR from recruiting Shc and inducing abnormal signalling as a consequence of ERK production. Indomethacin suppresses cancer cell proliferation by disturbing PKC–p38–DRP1 axis-based mitochondrial dynamics or downregulating Wnt/β-catenin signalling to effectively target MAPK mechanisms [263]. NSAIDs, such as indomethacin, are currently seriously being considered as potential anti-carcinogenic medicines [264267]. The therapeutic use of indomethacin offers a lot of potentials particularly if data would be collected enabling a better knowledge of the processes concerning its antiproliferative effects.

Antimalarial drugs

Artemisinins

A drug development initiative for the management of malaria led to the innovation of artemisinins. Artemisinins are plant extracts that have been used in Chinese traditional medicine for centuries. 
When used as a treatment for malaria, artemisinins trigger the production of free radicals in infected erythrocytes, eradicating the plasmodium parasite [268]. The effects of artemisinins are attributed to the interaction of endoperoxide moiety with the Fe-containing heme groups in the affected RBCs [269]. Artesunate is by far the most researched artemisinin which could be used for cancer medication repurposing. Ex-vivo and in-vivo data indicate that it might have antiangiogenic effects, the effects on the formation of free radicals, and the modification of antimicrobial resistance in a wide range of cancers [270]. Artesunate has antiproliferative and pro-apoptotic effects on lymphoma and myeloma cells [271], as well as on hepatocellular cancer cells [272, 273]. An increase in the expression of pro-apoptotic proteins, such as caspase-3, a reduction in the MYC oncogene, and a reduction of many anti-apoptotic proteins are all possible explanations for why this drug might have anti-malignant effects. The chemotherapeutic efficacy of artesunate in hepatocellular cancer is increased when combined with sorafenib [272] and gemcitabine [273]. Antiangiogenic effects of artesunate have been documented in renal cancer and hepatocellular carcinoma, with decreased tumor development in vivo, lower vessel number, and decreased vascular endothelial growth factor [274]. 
Dihydroartemisinin (DHA) is an analogue of artemisinin that inhibits leukemia cell proliferation by inducing autophagy and programmed cell death which is reactive oxygen species-dependent [275]. Decreased expression of the protein transferrin receptor 1 (TfR1) and cell cycle arrest were two factors that contributed to the effect of DHA. DHA therapy showed a significant reduction in iron in hepatoma and breast cancer cells due to the downregulation of TfR1, a protein that plays a significant role in iron absorption [276]. In leukemic cells, the TfR1 level was reduced [277]. TfR1 level was required for DHA responsiveness in different papillomavirus-infected cells and cervical cancer cells. In an in-vivo papilloma model, DHA therapy also suppressed tumour development [278].

Chloroquine

Chloroquine (CQ) is an antimalarial drug which was developed in the 1930s became the most widely used synthetic antimalarial drug during the 1960s and 1970s until the development of newer antimalarials in the whole world. Chloroquine has relatively recently been found to have potential anticancer effects. Chloroquine triggers tumor apoptosis by p53 and Rab8b-dependent Par-4 release, resulting in Par-4-dependent suppression of metastatic tumor development [279]. By reverting tumour-based macrophages to the M1 phenotype, chloroquine has effects on anticancer immune system response [280]. Chloroquine inhibits extrinsic neutrophil entrapment which decreases hypercoagulability in pancreatic malignant tumors [281]. Chloroquine suppresses tumour-associated Kv10.1 gates and reduces MDAMB-231 breast carcinoma cell motility ex-vivo [282]. In colon cancer, the combination of temsirolimus and chloroquine improves radiosensitivity [283]. Chloroquine sensitizes human T-cell blood cancer with oncogenic NOTCH1 mutations to secretase reduction [284]. Chloroquine is a derivative of quinoline similar to that of clioquinol. Many studies have reported its antitumor effects both in-vivo and in vitro. It has a synergistic effect in combination with paclitaxel and can stop the growth of breast malignant tumors [285]. Chloroquine, in combination with gemcitabine, seems to be able to eradicate tumor cells in xenograft models [149].

Hydroxychloroquine

Hydroxychloroquine is an analogue of chloroquine which has the same therapeutic effects as chloroquine although with less systemic toxicity. Hydroxychloroquine blocks intracellular lysosomal activities and improves the anticancer activities of breast cancer and glioblastoma [286, 287]. Hydroxychloroquine is an autophagy blocker that significantly increases the chemotherapeutic activity of bevacizumab on glioblastoma by suppressing of autophagy [288]. Ex-vivo and in-vivo, hydroxychloroquine increase the chemotherapeutic potential of the anti-angiogenesis drug BC001, suppressing the development of gastric carcinoma [289]. Pulmonary cancer cells are suppressed by hydroxychloroquine due to the increased chemo-sensitizing effect and effects on the change of M2-TAMs to M1-related macrophages, which improves the CD8+ T cell immunological reaction [290].

Quinacrine

Quinacrine was first identified in the 1920s as an antimalarial drug [291]. It is also used as an antibiotic, and a pleural sclerosing substance to manage giardiasis—a protozoal infection of the intestinal tract, certain types of lupus erythematosus and rheumatoid arthritis [292294]. Quinacrine has also been used in clinical studies for the management of Creutzfeldt–Jakob disease including a new variant of CJD which is linked to contamination of food by the bovine spongiform encephalopathy (BSE). Quinacrine seems to be very suitable for repurposing for cancer therapy [295297]. Earlier studies have shown that quinacrine has beneficial effects on different malignancies and that these effects are mediated by p53 activation. Quinacrine promotes p53 expression in renal cancer using suggesting that simultaneous inhibition of NF-kappaB and activation of p53 by a single small molecule can have anti-cancer effects [298]. It seems that quinacrine's cytotoxicity is associated with elevated p53 levels. It also seems that the facilitates chromatin transcription (FACT) protein complex, which seems to be trapped on chromatin and induces CK2-induced phosphorylation of p53, is responsible for quinacrine-mediated p53 transcription [299, 300]. Conversely, there are indications that the down-regulation of p53 increases the quinacrine effect in MCF-7 cells when compared with normal cells [301]. Nonetheless, the results of some studies suggest that quinacrine cytotoxicity on cancer cells is influenced by the p53 expression, at least to a certain extent [302, 303]. Studies with quinacrine as a chemotherapeutic drug are published recently quite often [32]. For example, in a Phase, I and Phase II clinical study, Fox Chase Cancer Center researchers combined quinacrine with capecitabine to treat colon carcinoma (NCT01844076). In the Phase I clinical study, quinacrine was combined with erlotinib for the management of recurrent or delayed pulmonary cancer (NCT01839955). Overall, quinacrine is very promising as an anticancer treatment, and the effects of this drug might be associated with the stimulation of p53, a crucial growth inhibitor that is dysregulated in many malignant diseases [32].

Atovaquone

In recent years, Atovaquone, a well-studied molecule known for its role as a non-oncological and anti-malarial drug, has garnered significant attention in the field of cancer therapy [304]. With its established safety profile and extensive clinical use, Atovaquone has emerged as a potential candidate for repurposing in the treatment of cancer [305]. Atovaquone, primarily used as an antiparasitic agent against malaria, has demonstrated promising anticancer properties in preclinical studies. Multiple investigations have shown its ability to inhibit cancer cell growth and induce apoptosis in various cancer types, including lung, breast, colon, and prostate cancers. These findings highlight the potential of Atovaquone as an effective anticancer agent [305].
Mechanistically, Atovaquone exerts its anticancer effects through multiple pathways. It has been shown to disrupt mitochondrial function, leading to energy depletion and apoptosis in cancer cells [304]. In addition, Atovaquone has demonstrated the ability to inhibit specific signaling pathways involved in cancer cell proliferation and survival, such as the PI3K/AKT pathway [306]. Moreover, the favorable safety profile of Atovaquone, established through its extensive use as an anti-malarial agent, further supports its potential for repurposing in cancer therapy. The well-tolerated nature of Atovaquone could potentially minimize adverse effects commonly associated with traditional chemotherapy agents, offering a more favorable treatment option for cancer patients [304, 306]. Although the repurposing of Atovaquone for cancer therapy is still in its early stages, ongoing preclinical and clinical studies are actively investigating its efficacy and safety in cancer treatment [304]. These studies aim to evaluate the optimal dosage, combination strategies, and patient selection criteria for Atovaquone-based therapies. Preliminary results from these studies have shown promising outcomes, warranting further investigation and clinical trials. Atovaquone, an anti-malarial drug, has emerged as a well-studied molecule with a high safety profile; its potential as an anticancer agent has been supported by pharmacological preclinical evidence demonstrating its ability to inhibit cancer cell growth and induce apoptosis [304, 306]. Ongoing research and clinical studies will shed more light on the efficacy and safety of Atovaquone in cancer therapy, paving the way for its potential inclusion as a repurposed drug in the treatment armamentarium against cancer.

Myorelaxant agents

Thiocolchicoside

Thiocolchicoside is a chemically synthesized colchicoside produced from Gloriosa superba (Liliaceae) that has been authorized in Europe (EMA) only as an add-on treatment for painful muscle contractures (permanent tightening of the muscle tissue) [307]. Surprisingly, thiocolchicoside has lately been mentioned in several publications to have anticancer effects. Reuter et al., for instance, found that thiocolchicoside has anticancer effects in different malignant diseases including leukemia, lymphoma, and squamous cell carcinoma. Thiocolchicoside inhibited NF-B and COX-2 stimulation by promoting ubiquitination deterioration of IB, a major suppressor of the NF-B signalling cascade controlling IKK status and p65 nuclear translocation [308]. It seems that thiocolchicoside can block the receptor stimulator of the NF-kB ligand and the NF-B signalling cascade, which inhibits cancer-induced bone metastasis [309]. Different pharmaceutical firms market thiocolchicoside as a myorelaxant with anti-inflammatory and analgesic effects and advertise its use as a nociceptive medication [310]. The effects of thiocolchicoside, when used in the treatment of lower back pain, have been validated in several clinical trials [311, 312]. Despite the limited clinical trials exploring its anticancer effects, thiocolchicoside, a half-century-old medication, might be useful in cancer treatment through drug repurposing. However, this has to be proven in clinical trials, because until now no such evidence does exist.

Conclusion

Human cancers are different diseases and, therefore, need different treatment approaches and options. There has been a significant development in discovering new drugs for different types of malignant diseases during the last two decades. However, due to acquired resistance to existing medicines, a considerable number of cancer patients are incurable, which causes frustration for scientists and physicians. The budgets of many countries for treating human cancers are limited, and they cannot afford the current chemotherapy treatments which are more and more expensive. As a result, drug repurposing has been identified as one of the most promising ways to find novel anticancer therapies that are cheaper and can be faster to obtain marketing authorization. Academics, scientists, and pharmaceutical businesses all recognize the value of drug repurposing in dealing with the rising burden of human cancers. Different types of drugs that can have anti-cancer cell effects have been discussed in this review. By targeting the well-studied mechanisms implicated in carcinogenesis, these drugs have been shown to limit cellular growth, metastasis, and invasion or induce cell cycle arrest and apoptosis. Clinical trials are currently being performed with repurposed drugs based on their preclinical anti-cancer efficacy. Some of them have been approved by the FDA for the treatment of human malignant diseases, such as raloxifene which has been approved for breast cancer, and thalidomide, which is used to treat multiple myeloma. A meta-analysis of medications including metformin, statins, and aspirin demonstrated their association with a lower risk of cancer, and these treatments may be licensed for cancer treatment in the near future. Scientists can now predict a treatment's efficacy, mode of action, and safety in different diseases, including cancer, thanks to advances in pharmacogenomics and high-throughput drug screening methods. Drug repurposing brings up a whole new world of research into existing drugs, potentially allowing more prompt and cheaper therapy for malignant diseases.
In conclusion, this comprehensive review explores the repurposing of non-oncology drugs for cancer therapy, focusing on elucidating mechanisms, evaluating efficacy, and exploring clinical prospects. One notable finding from our analysis is the observation of higher IC50 values associated with the repurposed compounds during in vitro studies. The higher IC50 values suggest that the repurposed compounds may exhibit reduced potency in inhibiting cancer cell growth compared to traditional anticancer agents. While this finding poses challenges, it also presents opportunities for future investigations. Understanding the underlying reasons for these elevated IC50 values is crucial for optimizing the therapeutic potential of repurposed compounds in cancer treatment. Several factors could contribute to the observed higher IC50 values, including differences in target specificity, altered pharmacokinetics, or complex interactions within the cancer microenvironment. Addressing these issues requires a multi-faceted approach involving in-depth mechanistic studies, refinement of drug formulations, and innovative combination strategies. To overcome the limitations posed by higher IC50 values, further research should focus on enhancing the effectiveness of repurposed compounds through various strategies. These may include identifying synergistic drug combinations, optimizing drug delivery systems to enhance bioavailability, or exploring novel formulations to improve target specificity. In addition, the use of advanced preclinical models that better recapitulate the complexities of human cancer biology could provide valuable insights into the efficacy of repurposed compounds. While higher IC50 values observed in vitro pose challenges, it is important to consider that repurposed drugs have the advantage of established safety profiles, known pharmacokinetics, and potentially reduced development timelines. By addressing the issue of higher IC50 values and leveraging the strengths of repurposed compounds, we can advance the field of cancer therapy by potentially identifying new treatment options that are both effective and safe. In summary, the observation of higher IC50 values for repurposed compounds during in vitro studies highlights the need for further investigation and optimization. By addressing these challenges head-on and capitalizing on the unique opportunities provided by repurposed drugs, we can accelerate the development of innovative cancer therapies and improve patient outcomes. This discrepancy raises important considerations for further research and clinical translation.

Acknowledgements

The authors acknowledge University of Kashmir for providing necessary facilities to complete this work.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors wish to confirm that there are no known conflicts of interest associated with this publication and there has been no significant financial support for this work that could have influenced its outcome.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Ozkan G, Günal-Köroğlu D, Karadag A, Capanoglu E, Cardoso SM, Al-Omari B, Calina D, Sharifi-Rad J, Cho WC. A mechanistic updated overview on lycopene as potential anticancer agent. Biomed Pharmacother. 2023;1(161):114428. Ozkan G, Günal-Köroğlu D, Karadag A, Capanoglu E, Cardoso SM, Al-Omari B, Calina D, Sharifi-Rad J, Cho WC. A mechanistic updated overview on lycopene as potential anticancer agent. Biomed Pharmacother. 2023;1(161):114428.
2.
Zurück zum Zitat Sharma R, Abbasi-Kangevari M, Abd-Rabu R, Abidi H, Abu-Gharbieh E, Acuna JM, Adhikari S, Advani SM, Afzal MS, Meybodi MA, Ahinkorah BO. Global, regional, and national burden of colorectal cancer and its risk factors, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Gastroenterol Hepatol. 2022;7(7):627–47. Sharma R, Abbasi-Kangevari M, Abd-Rabu R, Abidi H, Abu-Gharbieh E, Acuna JM, Adhikari S, Advani SM, Afzal MS, Meybodi MA, Ahinkorah BO. Global, regional, and national burden of colorectal cancer and its risk factors, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Gastroenterol Hepatol. 2022;7(7):627–47.
3.
Zurück zum Zitat Mohi-Ud-Din R, Mir RH, Wani TU, Alsharif KF, Alam W, Albrakati A, Saso L, Khan H. The regulation of endoplasmic reticulum stress in Cancer: special focuses on luteolin patents. Molecules. 2022;27(8):2471.PubMedPubMedCentral Mohi-Ud-Din R, Mir RH, Wani TU, Alsharif KF, Alam W, Albrakati A, Saso L, Khan H. The regulation of endoplasmic reticulum stress in Cancer: special focuses on luteolin patents. Molecules. 2022;27(8):2471.PubMedPubMedCentral
4.
Zurück zum Zitat Iqbal MJ, Javed Z, Herrera-Bravo J, Sadia H, Anum F, Raza S, Tahir A, Shahwani MN, Sharifi-Rad J, Calina D, Cho WC. Biosensing chips for cancer diagnosis and treatment: a new wave towards clinical innovation. Cancer Cell Int. 2022;22(1):1–6. Iqbal MJ, Javed Z, Herrera-Bravo J, Sadia H, Anum F, Raza S, Tahir A, Shahwani MN, Sharifi-Rad J, Calina D, Cho WC. Biosensing chips for cancer diagnosis and treatment: a new wave towards clinical innovation. Cancer Cell Int. 2022;22(1):1–6.
5.
Zurück zum Zitat Ianoși SL, Batani A, Ilie MA, Tampa M, Georgescu SR, Zurac S, Boda D, Ianosi NG, Neagoe D, Calina D, Tutunaru C. Non-invasive imaging techniques for the in vivo diagnosis of Bowen’s disease: three case reports. Oncol Lett. 2019;17(5):4094–101.PubMedPubMedCentral Ianoși SL, Batani A, Ilie MA, Tampa M, Georgescu SR, Zurac S, Boda D, Ianosi NG, Neagoe D, Calina D, Tutunaru C. Non-invasive imaging techniques for the in vivo diagnosis of Bowen’s disease: three case reports. Oncol Lett. 2019;17(5):4094–101.PubMedPubMedCentral
6.
Zurück zum Zitat Jain D, Chaudhary P, Varshney N, Bin Razzak KS, Verma D, Khan Zahra TR, Janmeda P, Sharifi-Rad J, Daştan SD, Mahmud S, Docea AO. Tobacco smoking and liver cancer risk: potential avenues for carcinogenesis. J Oncol. 2021;10:2021. Jain D, Chaudhary P, Varshney N, Bin Razzak KS, Verma D, Khan Zahra TR, Janmeda P, Sharifi-Rad J, Daştan SD, Mahmud S, Docea AO. Tobacco smoking and liver cancer risk: potential avenues for carcinogenesis. J Oncol. 2021;10:2021.
7.
Zurück zum Zitat Mir RH, Mir PA, Mohi-Ud-Din R, Sabreen S, Maqbool M, Shah AJ, Shenmar K, Raza SN, Pottoo FH. A comprehensive review on journey of pyrrole scaffold against multiple therapeutic targets. Anti-Cancer Agent Med Chem. 2022;22(19):3291–303. Mir RH, Mir PA, Mohi-Ud-Din R, Sabreen S, Maqbool M, Shah AJ, Shenmar K, Raza SN, Pottoo FH. A comprehensive review on journey of pyrrole scaffold against multiple therapeutic targets. Anti-Cancer Agent Med Chem. 2022;22(19):3291–303.
8.
Zurück zum Zitat Mir RH, Mohi-ud-din R, Wani TU, Dar MO, Shah AJ, Lone B, Pooja C, Masoodi MH. Indole: a privileged heterocyclic moiety in the management of cancer. Curr Org Chem. 2021;25(6):724–36. Mir RH, Mohi-ud-din R, Wani TU, Dar MO, Shah AJ, Lone B, Pooja C, Masoodi MH. Indole: a privileged heterocyclic moiety in the management of cancer. Curr Org Chem. 2021;25(6):724–36.
9.
Zurück zum Zitat Mohi-Ud-Din R, Mir RH, Sawhney G, Dar MA, Bhat ZA. Possible pathways of hepatotoxicity caused by chemical agents. Curr Drug Metab. 2019;20(11):867–79.PubMed Mohi-Ud-Din R, Mir RH, Sawhney G, Dar MA, Bhat ZA. Possible pathways of hepatotoxicity caused by chemical agents. Curr Drug Metab. 2019;20(11):867–79.PubMed
10.
Zurück zum Zitat Dhyani P, Quispe C, Sharma E, Bahukhandi A, Sati P, Attri DC, Szopa A, Sharifi-Rad J, Docea AO, Mardare I, Calina D. Anticancer potential of alkaloids: a key emphasis to colchicine, vinblastine, vincristine, vindesine, vinorelbine and vincamine. Cancer Cell Int. 2022;22(1):1–20. Dhyani P, Quispe C, Sharma E, Bahukhandi A, Sati P, Attri DC, Szopa A, Sharifi-Rad J, Docea AO, Mardare I, Calina D. Anticancer potential of alkaloids: a key emphasis to colchicine, vinblastine, vincristine, vindesine, vinorelbine and vincamine. Cancer Cell Int. 2022;22(1):1–20.
11.
Zurück zum Zitat Garzoli S, Alarcón-Zapata P, Seitimova G, Alarcón-Zapata B, Martorell M, Sharopov F, Fokou PV, Dize D, Yamthe LR, Les F, Cásedas G. Natural essential oils as a new therapeutic tool in colorectal cancer. Cancer Cell Int. 2022;22(1):407.PubMedPubMedCentral Garzoli S, Alarcón-Zapata P, Seitimova G, Alarcón-Zapata B, Martorell M, Sharopov F, Fokou PV, Dize D, Yamthe LR, Les F, Cásedas G. Natural essential oils as a new therapeutic tool in colorectal cancer. Cancer Cell Int. 2022;22(1):407.PubMedPubMedCentral
12.
Zurück zum Zitat Mir PA, Mohi-Ud-Din R, Banday N, Maqbool M, Raza SN, Farooq S, Afzal S, Mir RH. Anticancer potential of thymoquinone: a novel bioactive natural compound from Nigella sativa L. Anti-Cancer Agent Med Chem. 2022;22(20):3401–15. Mir PA, Mohi-Ud-Din R, Banday N, Maqbool M, Raza SN, Farooq S, Afzal S, Mir RH. Anticancer potential of thymoquinone: a novel bioactive natural compound from Nigella sativa L. Anti-Cancer Agent Med Chem. 2022;22(20):3401–15.
13.
Zurück zum Zitat Mohi-Ud-Din R, Mir RH, Sabreen S, Jan R, Pottoo FH, Singh IP. Recent insights into therapeutic potential of plant-derived flavonoids against cancer. Anti-Cancer Agent Med Chem. 2022;22(20):3343–69. Mohi-Ud-Din R, Mir RH, Sabreen S, Jan R, Pottoo FH, Singh IP. Recent insights into therapeutic potential of plant-derived flavonoids against cancer. Anti-Cancer Agent Med Chem. 2022;22(20):3343–69.
14.
Zurück zum Zitat Bhat IA, Kabeer SW, Reza MI, Mir RH, Dar MO. AdipoRon: a novel insulin sensitizer in various complications and the underlying mechanisms: a review. Curr Mol Pharmacol. 2020;13(2):94–107.PubMed Bhat IA, Kabeer SW, Reza MI, Mir RH, Dar MO. AdipoRon: a novel insulin sensitizer in various complications and the underlying mechanisms: a review. Curr Mol Pharmacol. 2020;13(2):94–107.PubMed
15.
Zurück zum Zitat Almatroodi SA, Alsahli MA, Almatroudi A, Rahmani AH. Garlic and its active compounds: a potential candidate in the prevention of cancer by modulating various cell signalling pathways. Anti-Cancer Agent Med Chem. 2019;19(11):1314–24. Almatroodi SA, Alsahli MA, Almatroudi A, Rahmani AH. Garlic and its active compounds: a potential candidate in the prevention of cancer by modulating various cell signalling pathways. Anti-Cancer Agent Med Chem. 2019;19(11):1314–24.
16.
Zurück zum Zitat Wani TU, Mohi-Ud-Din R, Mir RH, Itoo AM, Mir KB, Fazli AA, Pottoo FH. Exosomes harnessed as nanocarriers for cancer therapy-current status and potential for future clinical applications. Curr Mol Med. 2021;21(9):707–23.PubMed Wani TU, Mohi-Ud-Din R, Mir RH, Itoo AM, Mir KB, Fazli AA, Pottoo FH. Exosomes harnessed as nanocarriers for cancer therapy-current status and potential for future clinical applications. Curr Mol Med. 2021;21(9):707–23.PubMed
17.
Zurück zum Zitat Hassan R, Mohi-Ud-Din R, Dar MO, Shah AJ, Mir PA, Shaikh M, Pottoo FH. Bioactive heterocyclic compounds as potential therapeutics in the treatment of gliomas: a review. Anti-Cancer Agent Med Chem. 2022;22(3):551–65. Hassan R, Mohi-Ud-Din R, Dar MO, Shah AJ, Mir PA, Shaikh M, Pottoo FH. Bioactive heterocyclic compounds as potential therapeutics in the treatment of gliomas: a review. Anti-Cancer Agent Med Chem. 2022;22(3):551–65.
18.
Zurück zum Zitat Mohi-ud-Din R, Mir RH, Mir PA, Banday N, Shah AJ, Sawhney G, Bhat MM, Batiha GE, Pottoo FH. Dysfunction of ABC transporters at the surface of BBB: potential implications in intractable epilepsy and applications of nanotechnology enabled drug delivery. Curr Drug Metab. 2022;23(9):735–56.PubMed Mohi-ud-Din R, Mir RH, Mir PA, Banday N, Shah AJ, Sawhney G, Bhat MM, Batiha GE, Pottoo FH. Dysfunction of ABC transporters at the surface of BBB: potential implications in intractable epilepsy and applications of nanotechnology enabled drug delivery. Curr Drug Metab. 2022;23(9):735–56.PubMed
19.
Zurück zum Zitat Bertolini F, Sukhatme VP, Bouche G. Drug repurposing in oncology—patient and health systems opportunities. Nat Rev Clin Oncol. 2015;12(12):732–42.PubMed Bertolini F, Sukhatme VP, Bouche G. Drug repurposing in oncology—patient and health systems opportunities. Nat Rev Clin Oncol. 2015;12(12):732–42.PubMed
20.
Zurück zum Zitat Xue H, Li J, Xie H, Wang Y. Review of drug repositioning approaches and resources. Int J Biol Sci. 2018;14(10):1232.PubMedPubMedCentral Xue H, Li J, Xie H, Wang Y. Review of drug repositioning approaches and resources. Int J Biol Sci. 2018;14(10):1232.PubMedPubMedCentral
21.
Zurück zum Zitat Taheri Y, Joković N, Vitorović J, Grundmann O, Maroyi A, Calina D. The burden of the serious and difficult-to-treat infections and a new antibiotic available: cefiderocol. Front Pharmacol. 2021;14(11):578823. Taheri Y, Joković N, Vitorović J, Grundmann O, Maroyi A, Calina D. The burden of the serious and difficult-to-treat infections and a new antibiotic available: cefiderocol. Front Pharmacol. 2021;14(11):578823.
22.
Zurück zum Zitat Mir RH, Mir PA, Shah AJ, Banday N, Sabreen S, Maqbool M, Jan R, Shafi N, Masoodi MH. Curcumin as a privileged scaffold molecule for various biological targets in drug development. Stud Nat Prod Chem. 2022;1(73):405–34. Mir RH, Mir PA, Shah AJ, Banday N, Sabreen S, Maqbool M, Jan R, Shafi N, Masoodi MH. Curcumin as a privileged scaffold molecule for various biological targets in drug development. Stud Nat Prod Chem. 2022;1(73):405–34.
23.
Zurück zum Zitat Mir RH, Mir PA, Uppal J, Chawla A, Patel M, Bardakci F, Adnan M, Mohi-Ud-Din R. Evolution of natural product scaffolds as potential proteasome inhibitors in developing cancer therapeutics. Metabolites. 2023;13(4):509.PubMedPubMedCentral Mir RH, Mir PA, Uppal J, Chawla A, Patel M, Bardakci F, Adnan M, Mohi-Ud-Din R. Evolution of natural product scaffolds as potential proteasome inhibitors in developing cancer therapeutics. Metabolites. 2023;13(4):509.PubMedPubMedCentral
24.
Zurück zum Zitat Sadeghi HM, Adeli I, Calina D, Docea AO, Mousavi T, Daniali M, Nikfar S, Tsatsakis A, Abdollahi M. Polycystic ovary syndrome: a comprehensive review of pathogenesis, management, and drug repurposing. Int J Mol Sci. 2022;23(2):583.PubMedPubMedCentral Sadeghi HM, Adeli I, Calina D, Docea AO, Mousavi T, Daniali M, Nikfar S, Tsatsakis A, Abdollahi M. Polycystic ovary syndrome: a comprehensive review of pathogenesis, management, and drug repurposing. Int J Mol Sci. 2022;23(2):583.PubMedPubMedCentral
25.
Zurück zum Zitat Maxmen A. Busting the billion-dollar myth: how to slash the cost of drug development. Nature. 2016;536(7617):388.PubMed Maxmen A. Busting the billion-dollar myth: how to slash the cost of drug development. Nature. 2016;536(7617):388.PubMed
26.
Zurück zum Zitat Reichel A, Lienau P. Pharmacokinetics in drug discovery: an exposure-centred approach to optimising and predicting drug efficacy and safety. In: Nielsch U, Fuhrmann U, Jaroch S, editors. New approaches to drug discovery. Berlin: Springer; 2016. p. 235–60. Reichel A, Lienau P. Pharmacokinetics in drug discovery: an exposure-centred approach to optimising and predicting drug efficacy and safety. In: Nielsch U, Fuhrmann U, Jaroch S, editors. New approaches to drug discovery. Berlin: Springer; 2016. p. 235–60.
27.
Zurück zum Zitat Rudrapal M, Khairnar SJ, Jadhav AG. Drug repurposing (DR): an emerging approach in drug discovery. Drug Repurp-Hypothesis Mol Aspect Ther Appl. 2020;13:10. Rudrapal M, Khairnar SJ, Jadhav AG. Drug repurposing (DR): an emerging approach in drug discovery. Drug Repurp-Hypothesis Mol Aspect Ther Appl. 2020;13:10.
28.
Zurück zum Zitat Deore AB, Dhumane JR, Wagh R, Sonawane R. The stages of drug discovery and development process. Asian J Pharm Res Dev. 2019;7(6):62–7. Deore AB, Dhumane JR, Wagh R, Sonawane R. The stages of drug discovery and development process. Asian J Pharm Res Dev. 2019;7(6):62–7.
29.
Zurück zum Zitat Ávalos-Moreno M, López-Tejada A, Blaya-Cánovas JL, Cara-Lupiañez FE, González-González A, Lorente JA, Sánchez-Rovira P, Granados-Principal S. Drug repurposing for triple-negative breast cancer. J Personal Med. 2020;10(4):200. Ávalos-Moreno M, López-Tejada A, Blaya-Cánovas JL, Cara-Lupiañez FE, González-González A, Lorente JA, Sánchez-Rovira P, Granados-Principal S. Drug repurposing for triple-negative breast cancer. J Personal Med. 2020;10(4):200.
30.
Zurück zum Zitat Fogel DB. Factors associated with clinical trials that fail and opportunities for improving the likelihood of success: a review. Contempor Clin Trials Commun. 2018;1(11):156–64. Fogel DB. Factors associated with clinical trials that fail and opportunities for improving the likelihood of success: a review. Contempor Clin Trials Commun. 2018;1(11):156–64.
31.
Zurück zum Zitat Shim JS, Liu JO. Recent advances in drug repositioning for the discovery of new anticancer drugs. Int J Biol Sci. 2014;10(7):654.PubMedPubMedCentral Shim JS, Liu JO. Recent advances in drug repositioning for the discovery of new anticancer drugs. Int J Biol Sci. 2014;10(7):654.PubMedPubMedCentral
32.
Zurück zum Zitat Zhang Z, Zhou L, Xie N, Nice EC, Zhang T, Cui Y, Huang C. Overcoming cancer therapeutic bottleneck by drug repurposing. Signal Transduct Target Ther. 2020;5(1):113.PubMedPubMedCentral Zhang Z, Zhou L, Xie N, Nice EC, Zhang T, Cui Y, Huang C. Overcoming cancer therapeutic bottleneck by drug repurposing. Signal Transduct Target Ther. 2020;5(1):113.PubMedPubMedCentral
33.
Zurück zum Zitat Hernández-Lemus E, Martínez-García M. Pathway-based drug-repurposing schemes in cancer: the role of translational bioinformatics. Front Oncol. 2021;14(10):605680. Hernández-Lemus E, Martínez-García M. Pathway-based drug-repurposing schemes in cancer: the role of translational bioinformatics. Front Oncol. 2021;14(10):605680.
34.
Zurück zum Zitat Sahoo BM, Ravi Kumar BV, Sruti J, Mahapatra MK, Banik BK, Borah P. Drug repurposing strategy (DRS): emerging approach to identify potential therapeutics for treatment of novel coronavirus infection. Front Mol Biosci. 2021;26(8):628144. Sahoo BM, Ravi Kumar BV, Sruti J, Mahapatra MK, Banik BK, Borah P. Drug repurposing strategy (DRS): emerging approach to identify potential therapeutics for treatment of novel coronavirus infection. Front Mol Biosci. 2021;26(8):628144.
36.
Zurück zum Zitat Ko Y. Computational drug repositioning: current progress and challenges. Appl Sci. 2020;10(15):5076. Ko Y. Computational drug repositioning: current progress and challenges. Appl Sci. 2020;10(15):5076.
37.
Zurück zum Zitat Plenge RM, Scolnick EM, Altshuler D. Validating therapeutic targets through human genetics. Nat Rev Drug Discov. 2013;12(8):581–94.PubMed Plenge RM, Scolnick EM, Altshuler D. Validating therapeutic targets through human genetics. Nat Rev Drug Discov. 2013;12(8):581–94.PubMed
38.
Zurück zum Zitat Sonaye HV, Sheikh RY, Doifode CA. Drug repurposing: iron in the fire for older drugs. Biomed Pharmacother. 2021;1(141):111638. Sonaye HV, Sheikh RY, Doifode CA. Drug repurposing: iron in the fire for older drugs. Biomed Pharmacother. 2021;1(141):111638.
39.
Zurück zum Zitat Kim TW. Drug repositioning approaches for the discovery of new therapeutics for Alzheimer’s disease. Neurotherapeutics. 2015;12:132–42.PubMed Kim TW. Drug repositioning approaches for the discovery of new therapeutics for Alzheimer’s disease. Neurotherapeutics. 2015;12:132–42.PubMed
40.
Zurück zum Zitat Ciallella JR, Reaume AG. In vivo phenotypic screening: clinical proof of concept for a drug repositioning approach. Drug Discov Today Technol. 2017;1(23):45–52. Ciallella JR, Reaume AG. In vivo phenotypic screening: clinical proof of concept for a drug repositioning approach. Drug Discov Today Technol. 2017;1(23):45–52.
41.
Zurück zum Zitat Ayyar P, Subramanian U. Repurposing–second life for drugs. Pharmacia. 2022;69(1):51–9. Ayyar P, Subramanian U. Repurposing–second life for drugs. Pharmacia. 2022;69(1):51–9.
42.
Zurück zum Zitat Parvathaneni V, Kulkarni NS, Muth A, Gupta V. Drug repurposing: a promising tool to accelerate the drug discovery process. Drug Discov Today. 2019;24(10):2076–85.PubMed Parvathaneni V, Kulkarni NS, Muth A, Gupta V. Drug repurposing: a promising tool to accelerate the drug discovery process. Drug Discov Today. 2019;24(10):2076–85.PubMed
43.
Zurück zum Zitat Reddy AS, Zhang S. Polypharmacology: drug discovery for the future. Expert Rev Clin Pharmacol. 2013;6(1):41–7.PubMed Reddy AS, Zhang S. Polypharmacology: drug discovery for the future. Expert Rev Clin Pharmacol. 2013;6(1):41–7.PubMed
44.
Zurück zum Zitat Zhang W, Bai Y, Wang Y, Xiao W. Polypharmacology in drug discovery: a review from systems pharmacology perspective. Curr Pharm Des. 2016;22(21):3171–81.PubMed Zhang W, Bai Y, Wang Y, Xiao W. Polypharmacology in drug discovery: a review from systems pharmacology perspective. Curr Pharm Des. 2016;22(21):3171–81.PubMed
45.
Zurück zum Zitat Gns HS, Saraswathy GR, Murahari M, Krishnamurthy M. An update on drug repurposing: re-written saga of the drug’s fate. Biomed Pharmacother. 2019;1(110):700–16. Gns HS, Saraswathy GR, Murahari M, Krishnamurthy M. An update on drug repurposing: re-written saga of the drug’s fate. Biomed Pharmacother. 2019;1(110):700–16.
46.
Zurück zum Zitat Galvin BD, Li Z, Villemaine E, Poole CB, Chapman MS, Pollastri MP, Wyatt PG, Carlow CK. A target repurposing approach identifies N-myristoyltransferase as a new candidate drug target in filarial nematodes. PLoS Negl Trop Dis. 2014;8(9):e3145.PubMedPubMedCentral Galvin BD, Li Z, Villemaine E, Poole CB, Chapman MS, Pollastri MP, Wyatt PG, Carlow CK. A target repurposing approach identifies N-myristoyltransferase as a new candidate drug target in filarial nematodes. PLoS Negl Trop Dis. 2014;8(9):e3145.PubMedPubMedCentral
47.
Zurück zum Zitat Mejía-Pedroza RA, Espinal-Enríquez J, Hernández-Lemus E. Pathway-based drug repositioning for breast cancer molecular subtypes. Front Pharmacol. 2018;15(9):905. Mejía-Pedroza RA, Espinal-Enríquez J, Hernández-Lemus E. Pathway-based drug repositioning for breast cancer molecular subtypes. Front Pharmacol. 2018;15(9):905.
48.
Zurück zum Zitat Jin G, Wong ST. Toward better drug repositioning: prioritizing and integrating existing methods into efficient pipelines. Drug Discov Today. 2014;19(5):637–44.PubMed Jin G, Wong ST. Toward better drug repositioning: prioritizing and integrating existing methods into efficient pipelines. Drug Discov Today. 2014;19(5):637–44.PubMed
49.
Zurück zum Zitat Kotelnikova E, Yuryev A, Mazo I, Daraselia N. Computational approaches for drug repositioning and combination therapy design. J Bioinform Comput Biol. 2010;8(03):593–606.PubMed Kotelnikova E, Yuryev A, Mazo I, Daraselia N. Computational approaches for drug repositioning and combination therapy design. J Bioinform Comput Biol. 2010;8(03):593–606.PubMed
50.
Zurück zum Zitat Iorio F, Saez-Rodriguez J, Bernardo DD. Network based elucidation of drug response: from modulators to targets. BMC Syst Biol. 2013;7(1):1–9. Iorio F, Saez-Rodriguez J, Bernardo DD. Network based elucidation of drug response: from modulators to targets. BMC Syst Biol. 2013;7(1):1–9.
51.
Zurück zum Zitat Austin CP, Brady LS, Insel TR, Collins FS. NIH molecular libraries initiative. Science. 2004;306(5699):1138–9.PubMed Austin CP, Brady LS, Insel TR, Collins FS. NIH molecular libraries initiative. Science. 2004;306(5699):1138–9.PubMed
52.
Zurück zum Zitat Skrabanek L, Saini HK, Bader GD, Enright AJ. Computational prediction of protein–protein interactions. Mol Biotechnol. 2008;38:1–7.PubMed Skrabanek L, Saini HK, Bader GD, Enright AJ. Computational prediction of protein–protein interactions. Mol Biotechnol. 2008;38:1–7.PubMed
53.
Zurück zum Zitat Laudisi F, Marônek M, Di Grazia A, Monteleone G, Stolfi C. Repositioning of anthelmintic drugs for the treatment of cancers of the digestive system. Int J Mol Sci. 2020;21(14):4957.PubMedPubMedCentral Laudisi F, Marônek M, Di Grazia A, Monteleone G, Stolfi C. Repositioning of anthelmintic drugs for the treatment of cancers of the digestive system. Int J Mol Sci. 2020;21(14):4957.PubMedPubMedCentral
54.
Zurück zum Zitat Armando RG, Mengual Gómez DL, Gomez DE. New drugs are not enough-drug repositioning in oncology: An update. Int J Oncol. 2020;56(3):651–84.PubMedPubMedCentral Armando RG, Mengual Gómez DL, Gomez DE. New drugs are not enough-drug repositioning in oncology: An update. Int J Oncol. 2020;56(3):651–84.PubMedPubMedCentral
55.
Zurück zum Zitat Hou ZJ, Luo X, Zhang W, Peng F, Cui B, Wu SJ, Zheng FM, Xu J, Xu LZ, Long ZJ, Wang XT. Flubendazole, FDA-approved anthelmintic, targets breast cancer stem-like cells. Oncotarget. 2015;6(8):6326.PubMedPubMedCentral Hou ZJ, Luo X, Zhang W, Peng F, Cui B, Wu SJ, Zheng FM, Xu J, Xu LZ, Long ZJ, Wang XT. Flubendazole, FDA-approved anthelmintic, targets breast cancer stem-like cells. Oncotarget. 2015;6(8):6326.PubMedPubMedCentral
56.
Zurück zum Zitat Oh E, Kim YJ, An H, Sung D, Cho TM, Farrand L, Jang S, Seo JH, Kim JY. Flubendazole elicits anti-metastatic effects in triple-negative breast cancer via STAT3 inhibition. Int J Cancer. 2018;143(8):1978–93.PubMed Oh E, Kim YJ, An H, Sung D, Cho TM, Farrand L, Jang S, Seo JH, Kim JY. Flubendazole elicits anti-metastatic effects in triple-negative breast cancer via STAT3 inhibition. Int J Cancer. 2018;143(8):1978–93.PubMed
57.
Zurück zum Zitat Kim YJ, Sung D, Oh E, Cho Y, Cho TM, Farrand L, Seo JH, Kim JY. Flubendazole overcomes trastuzumab resistance by targeting cancer stem-like properties and HER2 signaling in HER2-positive breast cancer. Cancer Lett. 2018;1(412):118–30. Kim YJ, Sung D, Oh E, Cho Y, Cho TM, Farrand L, Seo JH, Kim JY. Flubendazole overcomes trastuzumab resistance by targeting cancer stem-like properties and HER2 signaling in HER2-positive breast cancer. Cancer Lett. 2018;1(412):118–30.
58.
Zurück zum Zitat Lin S, Yang L, Yao Y, Xu L, Xiang Y, Zhao H, Wang L, Zuo Z, Huang X, Zhao C. Flubendazole demonstrates valid antitumor effects by inhibiting STAT3 and activating autophagy. J Exp Clin Cancer Res. 2019;38:1–3. Lin S, Yang L, Yao Y, Xu L, Xiang Y, Zhao H, Wang L, Zuo Z, Huang X, Zhao C. Flubendazole demonstrates valid antitumor effects by inhibiting STAT3 and activating autophagy. J Exp Clin Cancer Res. 2019;38:1–3.
59.
Zurück zum Zitat Li Y, Acharya G, Elahy M, Xin H, Khachigian LM. The anthelmintic flubendazole blocks human melanoma growth and metastasis and suppresses programmed cell death protein-1 and myeloid-derived suppressor cell accumulation. Cancer Lett. 2019;10(459):268–76. Li Y, Acharya G, Elahy M, Xin H, Khachigian LM. The anthelmintic flubendazole blocks human melanoma growth and metastasis and suppresses programmed cell death protein-1 and myeloid-derived suppressor cell accumulation. Cancer Lett. 2019;10(459):268–76.
60.
Zurück zum Zitat Laclette JP, Guerra G, Zetina C. Inhibition of tubulin polymerization by mebendazole. Biochem Biophys Res Commun. 1980;92(2):417–23.PubMed Laclette JP, Guerra G, Zetina C. Inhibition of tubulin polymerization by mebendazole. Biochem Biophys Res Commun. 1980;92(2):417–23.PubMed
61.
Zurück zum Zitat Guerini AE, Triggiani L, Maddalo M, Bonù ML, Frassine F, Baiguini A, Alghisi A, Tomasini D, Borghetti P, Pasinetti N, Bresciani R. Mebendazole as a candidate for drug repurposing in oncology: An extensive review of current literature. Cancers. 2019;11(9):1284.PubMedPubMedCentral Guerini AE, Triggiani L, Maddalo M, Bonù ML, Frassine F, Baiguini A, Alghisi A, Tomasini D, Borghetti P, Pasinetti N, Bresciani R. Mebendazole as a candidate for drug repurposing in oncology: An extensive review of current literature. Cancers. 2019;11(9):1284.PubMedPubMedCentral
62.
Zurück zum Zitat Bai RY, Staedtke V, Aprhys CM, Gallia GL, Riggins GJ. Antiparasitic mebendazole shows survival benefit in 2 preclinical models of glioblastoma multiforme. Neuro Oncol. 2011;13(9):974–82.PubMedPubMedCentral Bai RY, Staedtke V, Aprhys CM, Gallia GL, Riggins GJ. Antiparasitic mebendazole shows survival benefit in 2 preclinical models of glioblastoma multiforme. Neuro Oncol. 2011;13(9):974–82.PubMedPubMedCentral
63.
Zurück zum Zitat Rushworth LK, Hewit K, Munnings-Tomes S, Somani S, James D, Shanks E, Dufès C, Straube A, Patel R, Leung HY. Repurposing screen identifies mebendazole as a clinical candidate to synergise with docetaxel for prostate cancer treatment. Br J Cancer. 2020;122(4):517–27.PubMed Rushworth LK, Hewit K, Munnings-Tomes S, Somani S, James D, Shanks E, Dufès C, Straube A, Patel R, Leung HY. Repurposing screen identifies mebendazole as a clinical candidate to synergise with docetaxel for prostate cancer treatment. Br J Cancer. 2020;122(4):517–27.PubMed
64.
Zurück zum Zitat Zhang Z, Ji J, Liu H. Drug repurposing in oncology: Current evidence and future direction. Curr Med Chem. 2021;28(11):2175–94.PubMed Zhang Z, Ji J, Liu H. Drug repurposing in oncology: Current evidence and future direction. Curr Med Chem. 2021;28(11):2175–94.PubMed
65.
Zurück zum Zitat Williamson T, Mendes TB, Joe N, Cerutti JM, Riggins GJ. Mebendazole inhibits tumor growth and prevents lung metastasis in models of advanced thyroid cancer. Endocr Relat Cancer. 2020;27(3):123–36.PubMed Williamson T, Mendes TB, Joe N, Cerutti JM, Riggins GJ. Mebendazole inhibits tumor growth and prevents lung metastasis in models of advanced thyroid cancer. Endocr Relat Cancer. 2020;27(3):123–36.PubMed
66.
Zurück zum Zitat Pantziarka P, Bouche G, Meheus L, Sukhatme V, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)—mebendazole as an anti-cancer agent. Ecancermedicalscience. 2014;8:443.PubMedPubMedCentral Pantziarka P, Bouche G, Meheus L, Sukhatme V, Sukhatme VP. Repurposing Drugs in Oncology (ReDO)—mebendazole as an anti-cancer agent. Ecancermedicalscience. 2014;8:443.PubMedPubMedCentral
67.
Zurück zum Zitat Mukhopadhyay T, Sasaki JI, Ramesh R, Roth JA. Mebendazole elicits a potent antitumor effect on human cancer cell lines both in vitro and in vivo. Clin Cancer Res. 2002;8(9):2963–9.PubMed Mukhopadhyay T, Sasaki JI, Ramesh R, Roth JA. Mebendazole elicits a potent antitumor effect on human cancer cell lines both in vitro and in vivo. Clin Cancer Res. 2002;8(9):2963–9.PubMed
68.
Zurück zum Zitat Sawanyawisuth K, Williamson T, Wongkham S, Riggins GJ. Effect of the antiparasitic drug mebendazole on cholangiocarcinoma growth. Southeast Asian J Trop Med Public Health. 2014;45(6):1264.PubMed Sawanyawisuth K, Williamson T, Wongkham S, Riggins GJ. Effect of the antiparasitic drug mebendazole on cholangiocarcinoma growth. Southeast Asian J Trop Med Public Health. 2014;45(6):1264.PubMed
69.
Zurück zum Zitat Sasaki JI, Ramesh R, Chada S, Gomyo Y, Roth JA, Mukhopadhyay T. The anthelmintic drug mebendazole induces mitotic arrest and apoptosis by depolymerizing tubulin in non-small cell lung cancer cells. Mol Cancer Ther. 2002;1(13):1201–9.PubMed Sasaki JI, Ramesh R, Chada S, Gomyo Y, Roth JA, Mukhopadhyay T. The anthelmintic drug mebendazole induces mitotic arrest and apoptosis by depolymerizing tubulin in non-small cell lung cancer cells. Mol Cancer Ther. 2002;1(13):1201–9.PubMed
70.
Zurück zum Zitat Nygren P, Fryknäs M, Ågerup B, Larsson R. Repositioning of the anthelmintic drug mebendazole for the treatment for colon cancer. J Cancer Res Clin Oncol. 2013;139:2133–40.PubMedPubMedCentral Nygren P, Fryknäs M, Ågerup B, Larsson R. Repositioning of the anthelmintic drug mebendazole for the treatment for colon cancer. J Cancer Res Clin Oncol. 2013;139:2133–40.PubMedPubMedCentral
71.
Zurück zum Zitat Doudican N, Rodriguez A, Osman I, Orlow SJ. Mebendazole induces apoptosis via Bcl-2 inactivation in chemoresistant melanoma cells. Mol Cancer Res. 2008;6(8):1308–15.PubMed Doudican N, Rodriguez A, Osman I, Orlow SJ. Mebendazole induces apoptosis via Bcl-2 inactivation in chemoresistant melanoma cells. Mol Cancer Res. 2008;6(8):1308–15.PubMed
72.
Zurück zum Zitat Doudican NA, Byron SA, Pollock PM, Orlow SJ. XIAP downregulation accompanies mebendazole growth inhibition in melanoma xenografts. Anticancer Drugs. 2013;24(2):181–8.PubMed Doudican NA, Byron SA, Pollock PM, Orlow SJ. XIAP downregulation accompanies mebendazole growth inhibition in melanoma xenografts. Anticancer Drugs. 2013;24(2):181–8.PubMed
73.
Zurück zum Zitat Bodhinayake I, Symons M, Boockvar JA. Repurposing mebendazole for the treatment of medulloblastoma. Neurosurgery. 2015;76(2):N15–6.PubMed Bodhinayake I, Symons M, Boockvar JA. Repurposing mebendazole for the treatment of medulloblastoma. Neurosurgery. 2015;76(2):N15–6.PubMed
74.
Zurück zum Zitat Larsen AR, Bai RY, Chung JH, Borodovsky A, Rudin CM, Riggins GJ, Bunz F. Repurposing the antihelmintic mebendazole as a hedgehog inhibitor. Mol Cancer Ther. 2015;14(1):3–13.PubMed Larsen AR, Bai RY, Chung JH, Borodovsky A, Rudin CM, Riggins GJ, Bunz F. Repurposing the antihelmintic mebendazole as a hedgehog inhibitor. Mol Cancer Ther. 2015;14(1):3–13.PubMed
75.
Zurück zum Zitat Nygren P, Larsson R. Drug repositioning from bench to bedside: tumour remission by the antihelmintic drug mebendazole in refractory metastatic colon cancer. Acta Oncol. 2014;53(3):427–8.PubMed Nygren P, Larsson R. Drug repositioning from bench to bedside: tumour remission by the antihelmintic drug mebendazole in refractory metastatic colon cancer. Acta Oncol. 2014;53(3):427–8.PubMed
76.
Zurück zum Zitat Dobrosotskaya IY, Hammer GD, Schteingart DE, Maturen KE, Worden FP. Mebendazole monotherapy and long-term disease control in metastatic adrenocortical carcinoma. Endocr Pract. 2011;17(3):e59-62.PubMed Dobrosotskaya IY, Hammer GD, Schteingart DE, Maturen KE, Worden FP. Mebendazole monotherapy and long-term disease control in metastatic adrenocortical carcinoma. Endocr Pract. 2011;17(3):e59-62.PubMed
77.
Zurück zum Zitat Giovanelli A, Silva CL, Medeiros L, Vasconcellos MC. The molluscicidal activity of niclosamide (Bayluscide WP70®) on Melanoides tuberculata (Thiaridae), a snail associated with habitats of Biomphalaria glabrata (Planorbidae). Mem Inst Oswaldo Cruz. 2002;97:743–5.PubMed Giovanelli A, Silva CL, Medeiros L, Vasconcellos MC. The molluscicidal activity of niclosamide (Bayluscide WP70®) on Melanoides tuberculata (Thiaridae), a snail associated with habitats of Biomphalaria glabrata (Planorbidae). Mem Inst Oswaldo Cruz. 2002;97:743–5.PubMed
78.
Zurück zum Zitat Lu L, Dong J, Wang L, Xia Q, Zhang D, Kim H, Yin T, Fan S, Shen Q. Activation of STAT3 and Bcl-2 and reduction of reactive oxygen species (ROS) promote radioresistance in breast cancer and overcome of radioresistance with niclosamide. Oncogene. 2018;37(39):5292–304.PubMed Lu L, Dong J, Wang L, Xia Q, Zhang D, Kim H, Yin T, Fan S, Shen Q. Activation of STAT3 and Bcl-2 and reduction of reactive oxygen species (ROS) promote radioresistance in breast cancer and overcome of radioresistance with niclosamide. Oncogene. 2018;37(39):5292–304.PubMed
79.
Zurück zum Zitat Pampori NA, Singh G, Srivastava VM. Cotugnia digonopora: carbohydrate metabolism and effect of anthelmintics on immature worms. J Helminthol. 1984;58(1):39–47.PubMed Pampori NA, Singh G, Srivastava VM. Cotugnia digonopora: carbohydrate metabolism and effect of anthelmintics on immature worms. J Helminthol. 1984;58(1):39–47.PubMed
80.
Zurück zum Zitat Liao Z, Nan G, Yan Z, Zeng L, Deng Y, Ye J, Zhang Z, Qiao M, Li R, Denduluri S, Wang J. The anthelmintic drug niclosamide inhibits the proliferative activity of human osteosarcoma cells by targeting multiple signal pathways. Curr Cancer Drug Targets. 2015;15(8):726–38.PubMed Liao Z, Nan G, Yan Z, Zeng L, Deng Y, Ye J, Zhang Z, Qiao M, Li R, Denduluri S, Wang J. The anthelmintic drug niclosamide inhibits the proliferative activity of human osteosarcoma cells by targeting multiple signal pathways. Curr Cancer Drug Targets. 2015;15(8):726–38.PubMed
81.
Zurück zum Zitat Pan JX, Ding K, Wang CY. Niclosamide, an old antihelminthic agent, demonstrates antitumor activity by blocking multiple signaling pathways of cancer stem cells. Chin J Cancer. 2012;31(4):178.PubMedPubMedCentral Pan JX, Ding K, Wang CY. Niclosamide, an old antihelminthic agent, demonstrates antitumor activity by blocking multiple signaling pathways of cancer stem cells. Chin J Cancer. 2012;31(4):178.PubMedPubMedCentral
82.
Zurück zum Zitat Sack U, Walther W, Scudiero D, Selby M, Kobelt D, Lemm M, Fichtner I, Schlag PM, Shoemaker RH, Stein U. Novel effect of antihelminthic Niclosamide on S100A4-mediated metastatic progression in colon cancer. J Natl Cancer Inst. 2011;103(13):1018–36.PubMed Sack U, Walther W, Scudiero D, Selby M, Kobelt D, Lemm M, Fichtner I, Schlag PM, Shoemaker RH, Stein U. Novel effect of antihelminthic Niclosamide on S100A4-mediated metastatic progression in colon cancer. J Natl Cancer Inst. 2011;103(13):1018–36.PubMed
83.
Zurück zum Zitat Ye T, Xiong Y, Yan Y, Xia Y, Song X, Liu L, Li D, Wang N, Zhang L, Zhu Y, Zeng J. The anthelmintic drug niclosamide induces apoptosis, impairs metastasis and reduces immunosuppressive cells in breast cancer model. PLoS ONE. 2014;9(1):e85887.PubMedPubMedCentral Ye T, Xiong Y, Yan Y, Xia Y, Song X, Liu L, Li D, Wang N, Zhang L, Zhu Y, Zeng J. The anthelmintic drug niclosamide induces apoptosis, impairs metastasis and reduces immunosuppressive cells in breast cancer model. PLoS ONE. 2014;9(1):e85887.PubMedPubMedCentral
84.
Zurück zum Zitat Chen H, Yang Z, Ding C, Chu L, Zhang Y, Terry K, Liu H, Shen Q, Zhou J. Discovery of O-alkylamino-tethered niclosamide derivatives as potent and orally bioavailable anticancer agents. ACS Med Chem Lett. 2013;4(2):180–5.PubMedPubMedCentral Chen H, Yang Z, Ding C, Chu L, Zhang Y, Terry K, Liu H, Shen Q, Zhou J. Discovery of O-alkylamino-tethered niclosamide derivatives as potent and orally bioavailable anticancer agents. ACS Med Chem Lett. 2013;4(2):180–5.PubMedPubMedCentral
85.
Zurück zum Zitat Ma R, Ma ZG, Gao JL, Tai Y, Li LJ, Zhu HB, Li L, Dong DL, Sun ZJ. Injectable pegylated niclosamide (polyethylene glycol-modified niclosamide) for cancer therapy. J Biomed Mater Res, Part A. 2020;108(1):30–8. Ma R, Ma ZG, Gao JL, Tai Y, Li LJ, Zhu HB, Li L, Dong DL, Sun ZJ. Injectable pegylated niclosamide (polyethylene glycol-modified niclosamide) for cancer therapy. J Biomed Mater Res, Part A. 2020;108(1):30–8.
86.
Zurück zum Zitat Lin CK, Bai MY, Hu TM, Wang YC, Chao TK, Weng SJ, Huang RL, Su PH, Lai HC. Preclinical evaluation of a nanoformulated antihelminthic, niclosamide, in ovarian cancer. Oncotarget. 2016;7(8):8993.PubMedPubMedCentral Lin CK, Bai MY, Hu TM, Wang YC, Chao TK, Weng SJ, Huang RL, Su PH, Lai HC. Preclinical evaluation of a nanoformulated antihelminthic, niclosamide, in ovarian cancer. Oncotarget. 2016;7(8):8993.PubMedPubMedCentral
87.
Zurück zum Zitat Samy AL, Bakthavachalam V, Vudutha M, Vinjamuri S, Chinnapaka S, Munirathinam G. Eprinomectin, a novel semi-synthetic macrocylic lactone is cytotoxic to PC3 metastatic prostate cancer cells via inducing apoptosis. Toxicol Appl Pharmacol. 2020;15(401):115071. Samy AL, Bakthavachalam V, Vudutha M, Vinjamuri S, Chinnapaka S, Munirathinam G. Eprinomectin, a novel semi-synthetic macrocylic lactone is cytotoxic to PC3 metastatic prostate cancer cells via inducing apoptosis. Toxicol Appl Pharmacol. 2020;15(401):115071.
88.
Zurück zum Zitat Melotti A, Mas C, Kuciak M, Lorente-Trigos A, Ruiz i Altaba A. The river blindness drug I vermectin and related macrocyclic lactones inhibit WNT-TCF pathway responses in human cancer. EMBO Mol Med. 2014;6(10):1263–78.PubMedPubMedCentral Melotti A, Mas C, Kuciak M, Lorente-Trigos A, Ruiz i Altaba A. The river blindness drug I vermectin and related macrocyclic lactones inhibit WNT-TCF pathway responses in human cancer. EMBO Mol Med. 2014;6(10):1263–78.PubMedPubMedCentral
89.
Zurück zum Zitat Drinyaev VA, Mosin VA, Kruglyak EB, Novik TS, Sterlina TS, Ermakova NV, Kublik LN, Levitman MK, Shaposhnikova VV, Korystov YN. Antitumor effect of avermectins. Eur J Pharmacol. 2004;501(1–3):19–23.PubMed Drinyaev VA, Mosin VA, Kruglyak EB, Novik TS, Sterlina TS, Ermakova NV, Kublik LN, Levitman MK, Shaposhnikova VV, Korystov YN. Antitumor effect of avermectins. Eur J Pharmacol. 2004;501(1–3):19–23.PubMed
90.
Zurück zum Zitat Kirtonia A, Gala K, Fernandes SG, Pandya G, Pandey AK, Sethi G, Khattar E, Garg M. Repurposing of drugs: An attractive pharmacological strategy for cancer therapeutics. Semin Cancer Biol. 2021;68:258–78.PubMed Kirtonia A, Gala K, Fernandes SG, Pandya G, Pandey AK, Sethi G, Khattar E, Garg M. Repurposing of drugs: An attractive pharmacological strategy for cancer therapeutics. Semin Cancer Biol. 2021;68:258–78.PubMed
91.
Zurück zum Zitat Wang K, Gao W, Dou Q, Chen H, Li Q, Nice EC, Huang C. Ivermectin induces PAK1-mediated cytostatic autophagy in breast cancer. Autophagy. 2016;12(12):2498–9.PubMedPubMedCentral Wang K, Gao W, Dou Q, Chen H, Li Q, Nice EC, Huang C. Ivermectin induces PAK1-mediated cytostatic autophagy in breast cancer. Autophagy. 2016;12(12):2498–9.PubMedPubMedCentral
92.
Zurück zum Zitat Nambara S, Masuda T, Nishio M, Kuramitsu S, Tobo T, Ogawa Y, Hu Q, Iguchi T, Kuroda Y, Ito S, Eguchi H. Antitumor effects of the antiparasitic agent ivermectin via inhibition of Yes-associated protein 1 expression in gastric cancer. Oncotarget. 2017;8(64):107666.PubMedPubMedCentral Nambara S, Masuda T, Nishio M, Kuramitsu S, Tobo T, Ogawa Y, Hu Q, Iguchi T, Kuroda Y, Ito S, Eguchi H. Antitumor effects of the antiparasitic agent ivermectin via inhibition of Yes-associated protein 1 expression in gastric cancer. Oncotarget. 2017;8(64):107666.PubMedPubMedCentral
93.
Zurück zum Zitat Dominguez-Gomez G, Chavez-Blanco A, Medina-Franco JL, Saldivar-Gonzalez F, Flores-Torrontegui Y, Juarez M, Díaz-Chávez J, Gonzalez-Fierro A, Dueñas-González A. Ivermectin as an inhibitor of cancer stem-like cells. Mol Med Rep. 2018;17(2):3397–403.PubMed Dominguez-Gomez G, Chavez-Blanco A, Medina-Franco JL, Saldivar-Gonzalez F, Flores-Torrontegui Y, Juarez M, Díaz-Chávez J, Gonzalez-Fierro A, Dueñas-González A. Ivermectin as an inhibitor of cancer stem-like cells. Mol Med Rep. 2018;17(2):3397–403.PubMed
94.
Zurück zum Zitat Alavi SE, Shahmabadi HE. Anthelmintics for drug repurposing: Opportunities and challenges. Saudi Pharm J. 2021;29(5):434–45.PubMedPubMedCentral Alavi SE, Shahmabadi HE. Anthelmintics for drug repurposing: Opportunities and challenges. Saudi Pharm J. 2021;29(5):434–45.PubMedPubMedCentral
95.
Zurück zum Zitat Cao B, Li J, Zhu J, Shen M, Han K, Zhang Z, Yu Y, Wang Y, Wu D, Chen S, Sun A. The antiparasitic clioquinol induces apoptosis in leukemia and myeloma cells by inhibiting histone deacetylase activity. J Biol Chem. 2013;288(47):34181–9.PubMedPubMedCentral Cao B, Li J, Zhu J, Shen M, Han K, Zhang Z, Yu Y, Wang Y, Wu D, Chen S, Sun A. The antiparasitic clioquinol induces apoptosis in leukemia and myeloma cells by inhibiting histone deacetylase activity. J Biol Chem. 2013;288(47):34181–9.PubMedPubMedCentral
96.
Zurück zum Zitat Lin MX, Lin SH, Lin CC, Yang CC, Yuan SY. In vitro and in vivo antitumor effects of pyrimethamine on non-small cell lung cancers. Anticancer Res. 2018;38(6):3435–45.PubMed Lin MX, Lin SH, Lin CC, Yang CC, Yuan SY. In vitro and in vivo antitumor effects of pyrimethamine on non-small cell lung cancers. Anticancer Res. 2018;38(6):3435–45.PubMed
97.
Zurück zum Zitat Liu H, Qin Y, Zhai D, Zhang Q, Gu J, Tang Y, Yang J, Li K, Yang L, Chen S, Zhong W. Antimalarial drug pyrimethamine plays a dual role in antitumor proliferation and metastasis through targeting DHFR and TP. Mol Cancer Ther. 2019;18(3):541–55.PubMed Liu H, Qin Y, Zhai D, Zhang Q, Gu J, Tang Y, Yang J, Li K, Yang L, Chen S, Zhong W. Antimalarial drug pyrimethamine plays a dual role in antitumor proliferation and metastasis through targeting DHFR and TP. Mol Cancer Ther. 2019;18(3):541–55.PubMed
98.
Zurück zum Zitat Khan MW, Saadalla A, Ewida AH, Al-Katranji K, Al-Saoudi G, Giaccone ZT, Gounari F, Zhang M, Frank DA, Khazaie K. The STAT3 inhibitor pyrimethamine displays anti-cancer and immune stimulatory effects in murine models of breast cancer. Cancer Immunol Immunother. 2018;67:13–23.PubMed Khan MW, Saadalla A, Ewida AH, Al-Katranji K, Al-Saoudi G, Giaccone ZT, Gounari F, Zhang M, Frank DA, Khazaie K. The STAT3 inhibitor pyrimethamine displays anti-cancer and immune stimulatory effects in murine models of breast cancer. Cancer Immunol Immunother. 2018;67:13–23.PubMed
99.
Zurück zum Zitat Liu Y, Zhou H, Yi T, Wang H. Pyrimethamine exerts significant antitumor effects on human ovarian cancer cells both in vitro and in vivo. Anticancer Drugs. 2019;30(6):571–8.PubMed Liu Y, Zhou H, Yi T, Wang H. Pyrimethamine exerts significant antitumor effects on human ovarian cancer cells both in vitro and in vivo. Anticancer Drugs. 2019;30(6):571–8.PubMed
100.
Zurück zum Zitat Seaberg EC, Wiley D, Martínez-Maza O, Chmiel JS, Kingsley L, Tang Y, Margolick JB, Jacobson LP, Multicenter AIDS Cohort Study (MACS). Cancer incidence in the multicenter AIDS Cohort Study before and during the HAART era: 1984 to 2007. Cancer. 2010;116(23):5507–16.PubMed Seaberg EC, Wiley D, Martínez-Maza O, Chmiel JS, Kingsley L, Tang Y, Margolick JB, Jacobson LP, Multicenter AIDS Cohort Study (MACS). Cancer incidence in the multicenter AIDS Cohort Study before and during the HAART era: 1984 to 2007. Cancer. 2010;116(23):5507–16.PubMed
101.
Zurück zum Zitat Kumar S, Bryant CS, Chamala S, Qazi A, Seward S, Pal J, Steffes CP, Weaver DW, Morris R, Malone JM, Shammas MA. Ritonavir blocks AKT signaling, activates apoptosis and inhibits migration and invasion in ovarian cancer cells. Mol Cancer. 2009;8(1):1–2. Kumar S, Bryant CS, Chamala S, Qazi A, Seward S, Pal J, Steffes CP, Weaver DW, Morris R, Malone JM, Shammas MA. Ritonavir blocks AKT signaling, activates apoptosis and inhibits migration and invasion in ovarian cancer cells. Mol Cancer. 2009;8(1):1–2.
102.
Zurück zum Zitat Batchu RB, Gruzdyn OV, Bryant CS, Qazi AM, Kumar S, Chamala S, Kung ST, Sanka RS, Puttagunta US, Weaver DW, Gruber SA. Ritonavir-mediated induction of apoptosis in pancreatic cancer occurs via the RB/E2F-1 and AKT pathways. Pharmaceuticals. 2014;7(1):46–57.PubMedPubMedCentral Batchu RB, Gruzdyn OV, Bryant CS, Qazi AM, Kumar S, Chamala S, Kung ST, Sanka RS, Puttagunta US, Weaver DW, Gruber SA. Ritonavir-mediated induction of apoptosis in pancreatic cancer occurs via the RB/E2F-1 and AKT pathways. Pharmaceuticals. 2014;7(1):46–57.PubMedPubMedCentral
103.
Zurück zum Zitat Sleire L, Førde HE, Netland IA, Leiss L, Skeie BS, Enger PØ. Drug repurposing in cancer. Pharmacol Res. 2017;1(124):74–91. Sleire L, Førde HE, Netland IA, Leiss L, Skeie BS, Enger PØ. Drug repurposing in cancer. Pharmacol Res. 2017;1(124):74–91.
104.
Zurück zum Zitat Kast RE, Ramiro S, Lladó S, Toro S, Coveñas R, Muñoz M. Antitumor action of temozolomide, ritonavir and aprepitant against human glioma cells. J Neurooncol. 2016;126:425–31.PubMed Kast RE, Ramiro S, Lladó S, Toro S, Coveñas R, Muñoz M. Antitumor action of temozolomide, ritonavir and aprepitant against human glioma cells. J Neurooncol. 2016;126:425–31.PubMed
105.
Zurück zum Zitat Sato A, Asano T, Ito K, Asano T. Ritonavir interacts with bortezomib to enhance protein ubiquitination and histone acetylation synergistically in renal cancer cells. Urology. 2012;79(4):966-e13. Sato A, Asano T, Ito K, Asano T. Ritonavir interacts with bortezomib to enhance protein ubiquitination and histone acetylation synergistically in renal cancer cells. Urology. 2012;79(4):966-e13.
106.
Zurück zum Zitat Adekola KU, Dalva Aydemir S, Ma S, Zhou Z, Rosen ST, Shanmugam M. Investigating and targeting chronic lymphocytic leukemia metabolism with the human immunodeficiency virus protease inhibitor ritonavir and metformin. Leuk Lymphoma. 2015;56(2):450–9.PubMed Adekola KU, Dalva Aydemir S, Ma S, Zhou Z, Rosen ST, Shanmugam M. Investigating and targeting chronic lymphocytic leukemia metabolism with the human immunodeficiency virus protease inhibitor ritonavir and metformin. Leuk Lymphoma. 2015;56(2):450–9.PubMed
107.
Zurück zum Zitat Popović-Djordjević J, Quispe C, Giordo R, Kostić A, Stanković JS, Fokou PV, Carbone K, Martorell M, Kumar M, Pintus G, Sharifi-Rad J. Natural products and synthetic analogues against HIV: a perspective to develop new potential anti-HIV drugs. Eur J Med Chem. 2022;5(233):114217. Popović-Djordjević J, Quispe C, Giordo R, Kostić A, Stanković JS, Fokou PV, Carbone K, Martorell M, Kumar M, Pintus G, Sharifi-Rad J. Natural products and synthetic analogues against HIV: a perspective to develop new potential anti-HIV drugs. Eur J Med Chem. 2022;5(233):114217.
108.
Zurück zum Zitat Bono C, Karlin L, Harel S, Mouly E, Labaume S, Galicier L, Apcher S, Sauvageon H, Fermand JP, Bories JC, Arnulf B. The human immunodeficiency virus-1 protease inhibitor nelfinavir impairs proteasome activity and inhibits the proliferation of multiple myeloma cells in vitro and in vivo. Haematologica. 2012;97(7):1101.PubMedPubMedCentral Bono C, Karlin L, Harel S, Mouly E, Labaume S, Galicier L, Apcher S, Sauvageon H, Fermand JP, Bories JC, Arnulf B. The human immunodeficiency virus-1 protease inhibitor nelfinavir impairs proteasome activity and inhibits the proliferation of multiple myeloma cells in vitro and in vivo. Haematologica. 2012;97(7):1101.PubMedPubMedCentral
109.
Zurück zum Zitat Pore N, Gupta AK, Cerniglia GJ, Jiang Z, Bernhard EJ, Evans SM, Koch CJ, Hahn SM, Maity A. Nelfinavir down-regulates hypoxia-inducible factor 1α and VEGF expression and increases tumor oxygenation: implications for radiotherapy. Can Res. 2006;66(18):9252–9. Pore N, Gupta AK, Cerniglia GJ, Jiang Z, Bernhard EJ, Evans SM, Koch CJ, Hahn SM, Maity A. Nelfinavir down-regulates hypoxia-inducible factor 1α and VEGF expression and increases tumor oxygenation: implications for radiotherapy. Can Res. 2006;66(18):9252–9.
110.
Zurück zum Zitat Gills JJ, LoPiccolo J, Tsurutani J, Shoemaker RH, Best CJ, Abu-Asab MS, Borojerdi J, Warfel NA, Gardner ER, Danish M, Hollander MC. Nelfinavir, A lead HIV protease inhibitor, is a broad-spectrum, anticancer agent that induces endoplasmic reticulum stress, autophagy, and apoptosis in vitro and in vivo. Clin Cancer Res. 2007;13(17):5183–94.PubMed Gills JJ, LoPiccolo J, Tsurutani J, Shoemaker RH, Best CJ, Abu-Asab MS, Borojerdi J, Warfel NA, Gardner ER, Danish M, Hollander MC. Nelfinavir, A lead HIV protease inhibitor, is a broad-spectrum, anticancer agent that induces endoplasmic reticulum stress, autophagy, and apoptosis in vitro and in vivo. Clin Cancer Res. 2007;13(17):5183–94.PubMed
111.
Zurück zum Zitat Brüning A, Burger P, Vogel M, Rahmeh M, Gingelmaier A, Friese K, Lenhard M, Burges A. Nelfinavir induces the unfolded protein response in ovarian cancer cells, resulting in ER vacuolization, cell cycle retardation and apoptosis. Cancer Biol Ther. 2009;8(3):226–32.PubMed Brüning A, Burger P, Vogel M, Rahmeh M, Gingelmaier A, Friese K, Lenhard M, Burges A. Nelfinavir induces the unfolded protein response in ovarian cancer cells, resulting in ER vacuolization, cell cycle retardation and apoptosis. Cancer Biol Ther. 2009;8(3):226–32.PubMed
112.
Zurück zum Zitat Guan M, Fousek K, Jiang C, Guo S, Synold T, Xi B, Shih CC, Chow WA. Nelfinavir induces liposarcoma apoptosis through inhibition of regulated intramembrane proteolysis of SREBP-1 and ATF6. Clin Cancer Res. 2011;17(7):1796–806.PubMed Guan M, Fousek K, Jiang C, Guo S, Synold T, Xi B, Shih CC, Chow WA. Nelfinavir induces liposarcoma apoptosis through inhibition of regulated intramembrane proteolysis of SREBP-1 and ATF6. Clin Cancer Res. 2011;17(7):1796–806.PubMed
113.
Zurück zum Zitat Cho HY, Thomas S, Golden EB, Gaffney KJ, Hofman FM, Chen TC, Louie SG, Petasis NA, Schönthal AH. Enhanced killing of chemo-resistant breast cancer cells via controlled aggravation of ER stress. Cancer Lett. 2009;282(1):87–97.PubMed Cho HY, Thomas S, Golden EB, Gaffney KJ, Hofman FM, Chen TC, Louie SG, Petasis NA, Schönthal AH. Enhanced killing of chemo-resistant breast cancer cells via controlled aggravation of ER stress. Cancer Lett. 2009;282(1):87–97.PubMed
114.
Zurück zum Zitat Guan M, Fousek K, Chow WA. Nelfinavir inhibits regulated intramembrane proteolysis of sterol regulatory element binding protein-1 and activating transcription factor 6 in castration-resistant prostate cancer. FEBS J. 2012;279(13):2399–411.PubMed Guan M, Fousek K, Chow WA. Nelfinavir inhibits regulated intramembrane proteolysis of sterol regulatory element binding protein-1 and activating transcription factor 6 in castration-resistant prostate cancer. FEBS J. 2012;279(13):2399–411.PubMed
115.
Zurück zum Zitat Guan M, Su L, Yuan YC, Li H, Chow WA. Nelfinavir and nelfinavir analogs block site-2 protease cleavage to inhibit castration-resistant prostate cancer. Sci Rep. 2015;5(1):9698.PubMedPubMedCentral Guan M, Su L, Yuan YC, Li H, Chow WA. Nelfinavir and nelfinavir analogs block site-2 protease cleavage to inhibit castration-resistant prostate cancer. Sci Rep. 2015;5(1):9698.PubMedPubMedCentral
116.
Zurück zum Zitat Thomas S, Sharma N, Golden EB, Cho H, Agarwal P, Gaffney KJ, Petasis NA, Chen TC, Hofman FM, Louie SG, Schönthal AH. Preferential killing of triple-negative breast cancer cells in vitro and in vivo when pharmacological aggravators of endoplasmic reticulum stress are combined with autophagy inhibitors. Cancer Lett. 2012;325(1):63–71.PubMed Thomas S, Sharma N, Golden EB, Cho H, Agarwal P, Gaffney KJ, Petasis NA, Chen TC, Hofman FM, Louie SG, Schönthal AH. Preferential killing of triple-negative breast cancer cells in vitro and in vivo when pharmacological aggravators of endoplasmic reticulum stress are combined with autophagy inhibitors. Cancer Lett. 2012;325(1):63–71.PubMed
117.
Zurück zum Zitat Pyrko P, Kardosh A, Wang W, Xiong W, Schönthal AH, Chen TC. HIV-1 protease inhibitors nelfinavir and atazanavir induce malignant glioma death by triggering endoplasmic reticulum stress. Can Res. 2007;67(22):10920–8. Pyrko P, Kardosh A, Wang W, Xiong W, Schönthal AH, Chen TC. HIV-1 protease inhibitors nelfinavir and atazanavir induce malignant glioma death by triggering endoplasmic reticulum stress. Can Res. 2007;67(22):10920–8.
118.
Zurück zum Zitat Tian X, Ye J, Alonso-Basanta M, Hahn SM, Koumenis C, Dorsey JF. Modulation of CCAAT/enhancer binding protein homologous protein (CHOP)-dependent DR5 expression by nelfinavir sensitizes glioblastoma multiforme cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). J Biol Chem. 2011;286(33):29408–16.PubMedPubMedCentral Tian X, Ye J, Alonso-Basanta M, Hahn SM, Koumenis C, Dorsey JF. Modulation of CCAAT/enhancer binding protein homologous protein (CHOP)-dependent DR5 expression by nelfinavir sensitizes glioblastoma multiforme cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). J Biol Chem. 2011;286(33):29408–16.PubMedPubMedCentral
119.
Zurück zum Zitat Xiang T, Du L, Pham P, Zhu B, Jiang S. Nelfinavir, an HIV protease inhibitor, induces apoptosis and cell cycle arrest in human cervical cancer cells via the ROS-dependent mitochondrial pathway. Cancer Lett. 2015;364(1):79–88.PubMed Xiang T, Du L, Pham P, Zhu B, Jiang S. Nelfinavir, an HIV protease inhibitor, induces apoptosis and cell cycle arrest in human cervical cancer cells via the ROS-dependent mitochondrial pathway. Cancer Lett. 2015;364(1):79–88.PubMed
120.
Zurück zum Zitat Pan J, Mott M, Xi B, Hepner E, Guan M, Fousek K, Magnusson R, Tinsley R, Valdes F, Frankel P, Synold T. Phase I study of nelfinavir in liposarcoma. Cancer Chemother Pharmacol. 2012;70:791–9.PubMedPubMedCentral Pan J, Mott M, Xi B, Hepner E, Guan M, Fousek K, Magnusson R, Tinsley R, Valdes F, Frankel P, Synold T. Phase I study of nelfinavir in liposarcoma. Cancer Chemother Pharmacol. 2012;70:791–9.PubMedPubMedCentral
121.
Zurück zum Zitat Hoover AC, Milhem MM, Anderson CM, Sun W, Smith BJ, Hoffman HT, Buatti JM. Efficacy of nelfinavir as monotherapy in refractory adenoid cystic carcinoma: results of a phase II clinical trial. Head Neck. 2015;37(5):722–6.PubMed Hoover AC, Milhem MM, Anderson CM, Sun W, Smith BJ, Hoffman HT, Buatti JM. Efficacy of nelfinavir as monotherapy in refractory adenoid cystic carcinoma: results of a phase II clinical trial. Head Neck. 2015;37(5):722–6.PubMed
122.
Zurück zum Zitat Elion GB. Mechanism of action and selectivity of acyclovir. Am J Med. 1982;73(1):7–13.PubMed Elion GB. Mechanism of action and selectivity of acyclovir. Am J Med. 1982;73(1):7–13.PubMed
123.
Zurück zum Zitat Assouline S, Culjkovic B, Cocolakis E, Rousseau C, Beslu N, Amri A, Caplan S, Leber B, Roy DC, Miller WH Jr, Borden KL. Molecular targeting of the oncogene eIF4E in acute myeloid leukemia (AML): a proof-of-principle clinical trial with ribavirin. Blood. 2009;114(2):257–60.PubMed Assouline S, Culjkovic B, Cocolakis E, Rousseau C, Beslu N, Amri A, Caplan S, Leber B, Roy DC, Miller WH Jr, Borden KL. Molecular targeting of the oncogene eIF4E in acute myeloid leukemia (AML): a proof-of-principle clinical trial with ribavirin. Blood. 2009;114(2):257–60.PubMed
124.
Zurück zum Zitat Kentsis A, Topisirovic I, Culjkovic B, Shao L, Borden KL. Ribavirin suppresses eIF4E-mediated oncogenic transformation by physical mimicry of the 7-methyl guanosine mRNA cap. Proc Natl Acad Sci. 2004;101(52):18105–10.PubMedPubMedCentral Kentsis A, Topisirovic I, Culjkovic B, Shao L, Borden KL. Ribavirin suppresses eIF4E-mediated oncogenic transformation by physical mimicry of the 7-methyl guanosine mRNA cap. Proc Natl Acad Sci. 2004;101(52):18105–10.PubMedPubMedCentral
125.
Zurück zum Zitat Shaimerdenova M, Karapina O, Mektepbayeva D, Alibek K, Akilbekova D. The effects of antiviral treatment on breast cancer cell line. Infect Agent Cancer. 2017;12:1. Shaimerdenova M, Karapina O, Mektepbayeva D, Alibek K, Akilbekova D. The effects of antiviral treatment on breast cancer cell line. Infect Agent Cancer. 2017;12:1.
126.
Zurück zum Zitat Sidwell RW, Huffman JH, Khare GP, Allen LB, Witkowski JT, Robins RK. Broad-spectrum antiviral activity of virazole: 1-β-D-ribofuranosyl-1, 2, 4-triazole-3-carboxamide. Science. 1972;177(4050):705–6.PubMed Sidwell RW, Huffman JH, Khare GP, Allen LB, Witkowski JT, Robins RK. Broad-spectrum antiviral activity of virazole: 1-β-D-ribofuranosyl-1, 2, 4-triazole-3-carboxamide. Science. 1972;177(4050):705–6.PubMed
127.
Zurück zum Zitat Crotty S, Maag D, Arnold JJ, Zhong W, Lau JY, Hong Z, Andino R, Cameron CE. The broad-spectrum antiviral ribonucleoside ribavirin is an RNA virus mutagen. Nat Med. 2000;6(12):1375–9.PubMed Crotty S, Maag D, Arnold JJ, Zhong W, Lau JY, Hong Z, Andino R, Cameron CE. The broad-spectrum antiviral ribonucleoside ribavirin is an RNA virus mutagen. Nat Med. 2000;6(12):1375–9.PubMed
128.
Zurück zum Zitat Tam RC, Lau JY, Hong Z. Mechanisms of action of ribavirin in antiviral therapies. Antiviral Chem Chemother. 2001;12(5):261–72. Tam RC, Lau JY, Hong Z. Mechanisms of action of ribavirin in antiviral therapies. Antiviral Chem Chemother. 2001;12(5):261–72.
129.
Zurück zum Zitat Von der Haar T, Gross JD, Wagner G, McCarthy JE. The mRNA cap-binding protein eIF4E in post-transcriptional gene expression. Nat Struct Mol Biol. 2004;11(6):503–11.PubMed Von der Haar T, Gross JD, Wagner G, McCarthy JE. The mRNA cap-binding protein eIF4E in post-transcriptional gene expression. Nat Struct Mol Biol. 2004;11(6):503–11.PubMed
130.
Zurück zum Zitat Graff JR, Zimmer SG. Translational control and metastatic progression: enhanced activity of the mRNA cap-binding protein eIF-4E selectively enhances translation of metastasis-related mRNAs. Clin Exp Metas. 2003;20:265–73. Graff JR, Zimmer SG. Translational control and metastatic progression: enhanced activity of the mRNA cap-binding protein eIF-4E selectively enhances translation of metastasis-related mRNAs. Clin Exp Metas. 2003;20:265–73.
131.
Zurück zum Zitat Drach JC, Thomas MA, Barnett JW, Smith SH, Shipman C Jr. Tritiated thymidine incorporation does not measure DNA synthesis in ribavirin-treated human cells. Science. 1981;212(4494):549–51.PubMed Drach JC, Thomas MA, Barnett JW, Smith SH, Shipman C Jr. Tritiated thymidine incorporation does not measure DNA synthesis in ribavirin-treated human cells. Science. 1981;212(4494):549–51.PubMed
132.
Zurück zum Zitat Zimmermann T, Stingele K, Hartmann M, Haas J, von Einsiedel R, Wildemann B. Successful treatment of aids related PML with HAART and cidofovir. Eur J Med Res. 2001;6(5):190–62.PubMed Zimmermann T, Stingele K, Hartmann M, Haas J, von Einsiedel R, Wildemann B. Successful treatment of aids related PML with HAART and cidofovir. Eur J Med Res. 2001;6(5):190–62.PubMed
133.
Zurück zum Zitat Bronson JJ, Ho HT, De Boeck HI, Woods K, Ghazzouli IS, Martin JC, Hitchcock MJ. Biochemical pharmacology of acyclic nucleotide analogues. Ann NY Acad Sci. 1990;1(616):398–407. Bronson JJ, Ho HT, De Boeck HI, Woods K, Ghazzouli IS, Martin JC, Hitchcock MJ. Biochemical pharmacology of acyclic nucleotide analogues. Ann NY Acad Sci. 1990;1(616):398–407.
134.
Zurück zum Zitat De Clercq E. Therapeutic potential of Cidofovir (HPMPC, Vistide) for the treatment of DNA virus (ie herpes-, papova-, pox-and adenovirus) infections. Verhandelingen-Koninklijke Academie voor Geneeskunde Van Belgie. 1996;58(1):19–47.PubMed De Clercq E. Therapeutic potential of Cidofovir (HPMPC, Vistide) for the treatment of DNA virus (ie herpes-, papova-, pox-and adenovirus) infections. Verhandelingen-Koninklijke Academie voor Geneeskunde Van Belgie. 1996;58(1):19–47.PubMed
135.
Zurück zum Zitat Andrei G, Snoeck R, Piette J, Delvenne P, De Clercq E. Inhibiting effects of cidofovir (HPMPC) on the growth of the human cervical carcinoma (SiHa) xenografts in athymic nude mice. Oncol Res Featur Preclin Clin Cancer Ther. 1998;10(10):533–9. Andrei G, Snoeck R, Piette J, Delvenne P, De Clercq E. Inhibiting effects of cidofovir (HPMPC) on the growth of the human cervical carcinoma (SiHa) xenografts in athymic nude mice. Oncol Res Featur Preclin Clin Cancer Ther. 1998;10(10):533–9.
136.
Zurück zum Zitat Liekens S, Andrei G, Vandeputte M, De Clercq E, Neyts J. Potent inhibition of hemangioma formation in rats by the acyclic nucleoside phosphonate analogue cidofovir. Can Res. 1998;58(12):2562–7. Liekens S, Andrei G, Vandeputte M, De Clercq E, Neyts J. Potent inhibition of hemangioma formation in rats by the acyclic nucleoside phosphonate analogue cidofovir. Can Res. 1998;58(12):2562–7.
137.
Zurück zum Zitat Liekens S, Verbeken E, De Clercq E, Neyts J. Potent inhibition of hemangiosarcoma development in mice by cidofovir. Int J Cancer. 2001;92(2):161–7.PubMed Liekens S, Verbeken E, De Clercq E, Neyts J. Potent inhibition of hemangiosarcoma development in mice by cidofovir. Int J Cancer. 2001;92(2):161–7.PubMed
138.
Zurück zum Zitat Neyts J, Sadler R, De Clercq E, Raab-Traub N, Pagano JS. The antiviral agent cidofovir [(S)-1-(3-hydroxy-2-phosphonyl-methoxypropyl) cytosine] has pronounced activity against nasopharyngeal carcinoma grown in nude mice. Can Res. 1998;58(3):384–8. Neyts J, Sadler R, De Clercq E, Raab-Traub N, Pagano JS. The antiviral agent cidofovir [(S)-1-(3-hydroxy-2-phosphonyl-methoxypropyl) cytosine] has pronounced activity against nasopharyngeal carcinoma grown in nude mice. Can Res. 1998;58(3):384–8.
139.
Zurück zum Zitat Murono S, Raab-Traub N, Pagano JS. Prevention and inhibition of nasopharyngeal carcinoma growth by antiviral phosphonated nucleoside analogs. Can Res. 2001;61(21):7875–7. Murono S, Raab-Traub N, Pagano JS. Prevention and inhibition of nasopharyngeal carcinoma growth by antiviral phosphonated nucleoside analogs. Can Res. 2001;61(21):7875–7.
140.
Zurück zum Zitat Liekens S, Gijsbers S, Vanstreels E, Daelemans D, De Clercq E, Hatse S. The nucleotide analog cidofovir suppresses basic fibroblast growth factor (FGF2) expression and signaling and induces apoptosis in FGF2-overexpressing endothelial cells. Mol Pharmacol. 2007;71(3):695–703.PubMed Liekens S, Gijsbers S, Vanstreels E, Daelemans D, De Clercq E, Hatse S. The nucleotide analog cidofovir suppresses basic fibroblast growth factor (FGF2) expression and signaling and induces apoptosis in FGF2-overexpressing endothelial cells. Mol Pharmacol. 2007;71(3):695–703.PubMed
141.
Zurück zum Zitat Liekens S, Neyts J, De Clercq E, Verbeken E, Ribatti D, Presta M. Inhibition of fibroblast growth factor-2-induced vascular tumor formation by the acyclic nucleoside phosphonate cidofovir. Can Res. 2001;61(13):5057–64. Liekens S, Neyts J, De Clercq E, Verbeken E, Ribatti D, Presta M. Inhibition of fibroblast growth factor-2-induced vascular tumor formation by the acyclic nucleoside phosphonate cidofovir. Can Res. 2001;61(13):5057–64.
142.
Zurück zum Zitat Yan K, He LJ, Cheng W, Ji ZZ, Zhao BX, Hui XL, Cao SS, Chen B, He L, Lang SH, Miao Y. Inhibiting gastric cancer-associated angiogenesis by CIAPIN1 siRNA. Cancer Biol Ther. 2009;8(11):1058–63.PubMed Yan K, He LJ, Cheng W, Ji ZZ, Zhao BX, Hui XL, Cao SS, Chen B, He L, Lang SH, Miao Y. Inhibiting gastric cancer-associated angiogenesis by CIAPIN1 siRNA. Cancer Biol Ther. 2009;8(11):1058–63.PubMed
143.
Zurück zum Zitat Hadaczek P, Ozawa T, Soroceanu L, Yoshida Y, Matlaf L, Singer E, Fiallos E, James CD, Cobbs CS. Cidofovir: a novel antitumor agent for glioblastoma. Clin Cancer Res. 2013;19(23):6473–83.PubMedPubMedCentral Hadaczek P, Ozawa T, Soroceanu L, Yoshida Y, Matlaf L, Singer E, Fiallos E, James CD, Cobbs CS. Cidofovir: a novel antitumor agent for glioblastoma. Clin Cancer Res. 2013;19(23):6473–83.PubMedPubMedCentral
144.
Zurück zum Zitat Zhang L, Dratver MB, Yazal T, Dong K, Nguyen A, Yu G, Dao A, Dratver MB, Duhachek-Muggy S, Bhat K, Alli C. Mebendazole potentiates radiation therapy in triple-negative breast cancer. Int J Radiat Oncol Biol Phys. 2019;103(1):195–207.PubMed Zhang L, Dratver MB, Yazal T, Dong K, Nguyen A, Yu G, Dao A, Dratver MB, Duhachek-Muggy S, Bhat K, Alli C. Mebendazole potentiates radiation therapy in triple-negative breast cancer. Int J Radiat Oncol Biol Phys. 2019;103(1):195–207.PubMed
145.
Zurück zum Zitat Chen L, Wang L, Shen H, Lin H, Li D. Anthelminthic drug niclosamide sensitizes the responsiveness of cervical cancer cells to paclitaxel via oxidative stress-mediated mTOR inhibition. Biochem Biophys Res Commun. 2017;484(2):416–21.PubMed Chen L, Wang L, Shen H, Lin H, Li D. Anthelminthic drug niclosamide sensitizes the responsiveness of cervical cancer cells to paclitaxel via oxidative stress-mediated mTOR inhibition. Biochem Biophys Res Commun. 2017;484(2):416–21.PubMed
146.
Zurück zum Zitat King ML, Lindberg ME, Stodden GR, Okuda H, Ebers SD, Johnson A, Montag A, Lengyel E, MacLean Ii JA, Hayashi K. WNT7A/β-catenin signaling induces FGF1 and influences sensitivity to niclosamide in ovarian cancer. Oncogene. 2015;34(26):3452–62.PubMed King ML, Lindberg ME, Stodden GR, Okuda H, Ebers SD, Johnson A, Montag A, Lengyel E, MacLean Ii JA, Hayashi K. WNT7A/β-catenin signaling induces FGF1 and influences sensitivity to niclosamide in ovarian cancer. Oncogene. 2015;34(26):3452–62.PubMed
147.
Zurück zum Zitat Tang M, Hu X, Wang Y, Yao X, Zhang W, Yu C, Cheng F, Li J, Fang Q. Ivermectin, a potential anticancer drug derived from an antiparasitic drug. Pharmacol Res. 2021;1(163):105207. Tang M, Hu X, Wang Y, Yao X, Zhang W, Yu C, Cheng F, Li J, Fang Q. Ivermectin, a potential anticancer drug derived from an antiparasitic drug. Pharmacol Res. 2021;1(163):105207.
148.
Zurück zum Zitat Darwish WM, Bayoumi NA, El-Kolaly MT. Laser-responsive liposome for selective tumor targeting of nitazoxanide nanoparticles. Eur J Pharm Sci. 2018;1(111):526–33. Darwish WM, Bayoumi NA, El-Kolaly MT. Laser-responsive liposome for selective tumor targeting of nitazoxanide nanoparticles. Eur J Pharm Sci. 2018;1(111):526–33.
149.
Zurück zum Zitat Balic A, Sørensen MD, Trabulo SM, Sainz B Jr, Cioffi M, Vieira CR, Miranda-Lorenzo I, Hidalgo M, Kleeff J, Erkan M, Heeschen C. Chloroquine targets pancreatic cancer stem cells via inhibition of CXCR4 and hedgehog signaling. Mol Cancer Ther. 2014;13(7):1758–71.PubMed Balic A, Sørensen MD, Trabulo SM, Sainz B Jr, Cioffi M, Vieira CR, Miranda-Lorenzo I, Hidalgo M, Kleeff J, Erkan M, Heeschen C. Chloroquine targets pancreatic cancer stem cells via inhibition of CXCR4 and hedgehog signaling. Mol Cancer Ther. 2014;13(7):1758–71.PubMed
150.
Zurück zum Zitat Dalva-Aydemir S, Bajpai R, Martinez M, Adekola KU, Kandela I, Wei C, Singhal S, Koblinski JE, Raje NS, Rosen ST, Shanmugam M. Targeting the metabolic plasticity of multiple myeloma with FDA-approved ritonavir and metformin. Clin Cancer Res. 2015;21(5):1161–71.PubMed Dalva-Aydemir S, Bajpai R, Martinez M, Adekola KU, Kandela I, Wei C, Singhal S, Koblinski JE, Raje NS, Rosen ST, Shanmugam M. Targeting the metabolic plasticity of multiple myeloma with FDA-approved ritonavir and metformin. Clin Cancer Res. 2015;21(5):1161–71.PubMed
151.
Zurück zum Zitat Johnson CE, Hunt DK, Wiltshire M, Herbert TP, Sampson JR, Errington RJ, Davies DM, Tee AR. Endoplasmic reticulum stress and cell death in mTORC1-overactive cells is induced by nelfinavir and enhanced by chloroquine. Mol Oncol. 2015;9(3):675–88.PubMed Johnson CE, Hunt DK, Wiltshire M, Herbert TP, Sampson JR, Errington RJ, Davies DM, Tee AR. Endoplasmic reticulum stress and cell death in mTORC1-overactive cells is induced by nelfinavir and enhanced by chloroquine. Mol Oncol. 2015;9(3):675–88.PubMed
152.
Zurück zum Zitat Tan H, He L, Cheng Z. Inhibition of eIF4E signaling by ribavirin selectively targets lung cancer and angiogenesis. Biochem Biophys Res Commun. 2020;529(3):519–25.PubMed Tan H, He L, Cheng Z. Inhibition of eIF4E signaling by ribavirin selectively targets lung cancer and angiogenesis. Biochem Biophys Res Commun. 2020;529(3):519–25.PubMed
153.
Zurück zum Zitat Verhees F, Legemaate D, Demers I, Jacobs R, Haakma WE, Rousch M, Kremer B, Speel EJ. The antiviral agent cidofovir induces DNA damage and mitotic catastrophe in HPV-positive and-negative head and neck squamous cell carcinomas in vitro. Cancers. 2019;11(7):919.PubMedPubMedCentral Verhees F, Legemaate D, Demers I, Jacobs R, Haakma WE, Rousch M, Kremer B, Speel EJ. The antiviral agent cidofovir induces DNA damage and mitotic catastrophe in HPV-positive and-negative head and neck squamous cell carcinomas in vitro. Cancers. 2019;11(7):919.PubMedPubMedCentral
154.
Zurück zum Zitat Im EJ, Lee CH, Moon PG, Rangaswamy GG, Lee B, Lee JM, Lee JC, Jee JG, Bae JS, Kwon TK, Kang KW. Sulfisoxazole inhibits the secretion of small extracellular vesicles by targeting the endothelin receptor A. Nat Commun. 2019;10(1):1387.PubMedPubMedCentral Im EJ, Lee CH, Moon PG, Rangaswamy GG, Lee B, Lee JM, Lee JC, Jee JG, Bae JS, Kwon TK, Kang KW. Sulfisoxazole inhibits the secretion of small extracellular vesicles by targeting the endothelin receptor A. Nat Commun. 2019;10(1):1387.PubMedPubMedCentral
155.
Zurück zum Zitat Qiao X, Wang X, Shang Y, Li Y, Chen SZ. Azithromycin enhances anticancer activity of TRAIL by inhibiting autophagy and up-regulating the protein levels of DR4/5 in colon cancer cells in vitro and in vivo. Cancer Commun. 2018;38:1–3. Qiao X, Wang X, Shang Y, Li Y, Chen SZ. Azithromycin enhances anticancer activity of TRAIL by inhibiting autophagy and up-regulating the protein levels of DR4/5 in colon cancer cells in vitro and in vivo. Cancer Commun. 2018;38:1–3.
156.
Zurück zum Zitat Li F, Huang J, Ji D, Meng Q, Wang C, Chen S, Wang X, Zhu Z, Jiang C, Shi Y, Liu S. Azithromycin effectively inhibits tumor angiogenesis by suppressing vascular endothelial growth factor receptor 2-mediated signaling pathways in lung cancer. Oncol Lett. 2017;14(1):89–96.PubMedPubMedCentral Li F, Huang J, Ji D, Meng Q, Wang C, Chen S, Wang X, Zhu Z, Jiang C, Shi Y, Liu S. Azithromycin effectively inhibits tumor angiogenesis by suppressing vascular endothelial growth factor receptor 2-mediated signaling pathways in lung cancer. Oncol Lett. 2017;14(1):89–96.PubMedPubMedCentral
157.
Zurück zum Zitat Tamargo RJ, Bok RA, Brem H. Angiogenesis inhibition by minocycline. Can Res. 1991;51(2):672–5. Tamargo RJ, Bok RA, Brem H. Angiogenesis inhibition by minocycline. Can Res. 1991;51(2):672–5.
158.
Zurück zum Zitat Fife RS, Rougraff BT, Proctor C, Sledge GW Jr. Inhibition of proliferation and induction of apoptosis by doxycycline in cultured human osteosarcoma cells. J Lab Clin Med. 1997;130(5):530–4.PubMed Fife RS, Rougraff BT, Proctor C, Sledge GW Jr. Inhibition of proliferation and induction of apoptosis by doxycycline in cultured human osteosarcoma cells. J Lab Clin Med. 1997;130(5):530–4.PubMed
159.
Zurück zum Zitat Fife RS, Sledge GW Jr, Roth BJ, Proctor C. Effects of doxycycline on human prostate cancer cells in vitro. Cancer Lett. 1998;127(1–2):37–41.PubMed Fife RS, Sledge GW Jr, Roth BJ, Proctor C. Effects of doxycycline on human prostate cancer cells in vitro. Cancer Lett. 1998;127(1–2):37–41.PubMed
160.
Zurück zum Zitat Rubins JB, Charboneau D, Alter MD, Bitterman PB, Kratzke RA. Inhibition of mesothelioma cell growth in vitro by doxycycline. J Lab Clin Med. 2001;138(2):101–6.PubMed Rubins JB, Charboneau D, Alter MD, Bitterman PB, Kratzke RA. Inhibition of mesothelioma cell growth in vitro by doxycycline. J Lab Clin Med. 2001;138(2):101–6.PubMed
161.
Zurück zum Zitat Mouratidis PX, Colston KW, Dalgleish AG. Doxycycline induces caspase-dependent apoptosis in human pancreatic cancer cells. Int J Cancer. 2007;120(4):743–52.PubMed Mouratidis PX, Colston KW, Dalgleish AG. Doxycycline induces caspase-dependent apoptosis in human pancreatic cancer cells. Int J Cancer. 2007;120(4):743–52.PubMed
162.
Zurück zum Zitat Son K, Fujioka S, Iida T, Furukawa K, Fujita T, Yamada H, Chiao PJ, Yanaga K. Doxycycline induces apoptosis in PANC-1 pancreatic cancer cells. Anticancer Res. 2009;29(10):3995–4003.PubMed Son K, Fujioka S, Iida T, Furukawa K, Fujita T, Yamada H, Chiao PJ, Yanaga K. Doxycycline induces apoptosis in PANC-1 pancreatic cancer cells. Anticancer Res. 2009;29(10):3995–4003.PubMed
163.
Zurück zum Zitat Song H, Fares M, Maguire KR, Sidén Å, Potacova Z. Cytotoxic effects of tetracycline analogues (doxycycline, minocycline and COL-3) in acute myeloid leukemia HL-60 cells. PLoS ONE. 2014;9(12):e114457.PubMedPubMedCentral Song H, Fares M, Maguire KR, Sidén Å, Potacova Z. Cytotoxic effects of tetracycline analogues (doxycycline, minocycline and COL-3) in acute myeloid leukemia HL-60 cells. PLoS ONE. 2014;9(12):e114457.PubMedPubMedCentral
164.
Zurück zum Zitat Golub LM, Ramamurthy NS, McNamara TF, Greenwald RA, Rifkin BR. Tetracyclines inhibit connective tissue breakdown: new therapeutic implications for an old family of drugs. Crit Rev Oral Biol Med. 1991;2(3):297–321.PubMed Golub LM, Ramamurthy NS, McNamara TF, Greenwald RA, Rifkin BR. Tetracyclines inhibit connective tissue breakdown: new therapeutic implications for an old family of drugs. Crit Rev Oral Biol Med. 1991;2(3):297–321.PubMed
165.
Zurück zum Zitat Iwasaki H, Inoue H, Mitsuke Y, Badran A, Ikegaya S, Ueda T. Doxycycline induces apoptosis by way of caspase-3 activation with inhibition of matrix metalloproteinase in human T-lymphoblastic leukemia CCRF-CEM cells. J Lab Clin Med. 2002;140(6):382–6.PubMed Iwasaki H, Inoue H, Mitsuke Y, Badran A, Ikegaya S, Ueda T. Doxycycline induces apoptosis by way of caspase-3 activation with inhibition of matrix metalloproteinase in human T-lymphoblastic leukemia CCRF-CEM cells. J Lab Clin Med. 2002;140(6):382–6.PubMed
166.
Zurück zum Zitat Onoda T, Ono T, Dhar DK, Yamanoi A, Fujii T, Nagasue N. Doxycycline inhibits cell proliferation and invasive potential: combination therapy with cyclooxygenase-2 inhibitor in human colorectal cancer cells. J Lab Clin Med. 2004;143(4):207–16.PubMed Onoda T, Ono T, Dhar DK, Yamanoi A, Fujii T, Nagasue N. Doxycycline inhibits cell proliferation and invasive potential: combination therapy with cyclooxygenase-2 inhibitor in human colorectal cancer cells. J Lab Clin Med. 2004;143(4):207–16.PubMed
167.
Zurück zum Zitat Duivenvoorden WC, Popovic SV, Lhoták S, Seidlitz E, Hirte HW, Tozer RG, Singh G. Doxycycline decreases tumor burden in a bone metastasis model of human breast cancer. Can Res. 2002;62(6):1588–91. Duivenvoorden WC, Popovic SV, Lhoták S, Seidlitz E, Hirte HW, Tozer RG, Singh G. Doxycycline decreases tumor burden in a bone metastasis model of human breast cancer. Can Res. 2002;62(6):1588–91.
168.
Zurück zum Zitat Duivenvoorden WC, Vukmirović-Popović S, Kalina M, Seidlitz E, Singh G. Effect of zoledronic acid on the doxycycline-induced decrease in tumour burden in a bone metastasis model of human breast cancer. Br J Cancer. 2007;96(10):1526–31.PubMedPubMedCentral Duivenvoorden WC, Vukmirović-Popović S, Kalina M, Seidlitz E, Singh G. Effect of zoledronic acid on the doxycycline-induced decrease in tumour burden in a bone metastasis model of human breast cancer. Br J Cancer. 2007;96(10):1526–31.PubMedPubMedCentral
169.
Zurück zum Zitat Lokeshwar BL, Selzer MG, Zhu BQ, Block NL, Golub LM. Inhibition of cell proliferation, invasion, tumor growth and metastasis by an oral non-antimicrobial tetracycline analog (COL-3) in a metastatic prostate cancer model. Int J Cancer. 2002;98(2):297–309.PubMed Lokeshwar BL, Selzer MG, Zhu BQ, Block NL, Golub LM. Inhibition of cell proliferation, invasion, tumor growth and metastasis by an oral non-antimicrobial tetracycline analog (COL-3) in a metastatic prostate cancer model. Int J Cancer. 2002;98(2):297–309.PubMed
170.
Zurück zum Zitat Shen LC, Chen YK, Lin LM, Shaw SY. Anti-invasion and anti-tumor growth effect of doxycycline treatment for human oral squamous-cell carcinoma–in vitro and in vivo studies. Oral Oncol. 2010;46(3):178–84.PubMed Shen LC, Chen YK, Lin LM, Shaw SY. Anti-invasion and anti-tumor growth effect of doxycycline treatment for human oral squamous-cell carcinoma–in vitro and in vivo studies. Oral Oncol. 2010;46(3):178–84.PubMed
171.
Zurück zum Zitat Qin Y, Zhang Q, Lee S, Zhong WL, Liu YR, Liu HJ, Zhao D, Chen S, Xiao T, Meng J, Jing XS. Doxycycline reverses epithelial-to-mesenchymal transition and suppresses the proliferation and metastasis of lung cancer cells. Oncotarget. 2015;6(38):40667.PubMedPubMedCentral Qin Y, Zhang Q, Lee S, Zhong WL, Liu YR, Liu HJ, Zhao D, Chen S, Xiao T, Meng J, Jing XS. Doxycycline reverses epithelial-to-mesenchymal transition and suppresses the proliferation and metastasis of lung cancer cells. Oncotarget. 2015;6(38):40667.PubMedPubMedCentral
172.
Zurück zum Zitat Meng J, Sun B, Zhao X, Zhang D, Zhao X, Gu Q, Dong X, Zhao N, Liu P, Liu Y. Doxycycline as an inhibitor of the epithelial-to-mesenchymal transition and vasculogenic mimicry in hepatocellular carcinoma. Mol Cancer Ther. 2014;13(12):3107–22.PubMed Meng J, Sun B, Zhao X, Zhang D, Zhao X, Gu Q, Dong X, Zhao N, Liu P, Liu Y. Doxycycline as an inhibitor of the epithelial-to-mesenchymal transition and vasculogenic mimicry in hepatocellular carcinoma. Mol Cancer Ther. 2014;13(12):3107–22.PubMed
173.
Zurück zum Zitat Yang B, Lu Y, Zhang AI, Zhou A, Zhang L, Zhang L, Gao L, Zang Y, Tang X, Sun L. Doxycycline induces apoptosis and inhibits proliferation and invasion of human cervical carcinoma stem cells. PLoS ONE. 2015;10(6):e0129138.PubMedPubMedCentral Yang B, Lu Y, Zhang AI, Zhou A, Zhang L, Zhang L, Gao L, Zang Y, Tang X, Sun L. Doxycycline induces apoptosis and inhibits proliferation and invasion of human cervical carcinoma stem cells. PLoS ONE. 2015;10(6):e0129138.PubMedPubMedCentral
174.
Zurück zum Zitat Huie M, Oettel K, Van Ummersen L, Kim KM, Zhang Y, Staab MJ, Horvath D, Marnocha R, Douglas J, Drezen A, Alberti D. Phase II study of interferon-alpha and doxycycline for advanced renal cell carcinoma. Invest New Drugs. 2006;24:255–60.PubMed Huie M, Oettel K, Van Ummersen L, Kim KM, Zhang Y, Staab MJ, Horvath D, Marnocha R, Douglas J, Drezen A, Alberti D. Phase II study of interferon-alpha and doxycycline for advanced renal cell carcinoma. Invest New Drugs. 2006;24:255–60.PubMed
175.
Zurück zum Zitat Addison CL, Simos D, Wang Z, Pond G, Smith S, Robertson S, Mazzarello S, Singh G, Vandermeer L, Fernandes R, Iyengar A. A phase 2 trial exploring the clinical and correlative effects of combining doxycycline with bone-targeted therapy in patients with metastatic breast cancer. J Bone Oncol. 2016;5(4):173–9.PubMedPubMedCentral Addison CL, Simos D, Wang Z, Pond G, Smith S, Robertson S, Mazzarello S, Singh G, Vandermeer L, Fernandes R, Iyengar A. A phase 2 trial exploring the clinical and correlative effects of combining doxycycline with bone-targeted therapy in patients with metastatic breast cancer. J Bone Oncol. 2016;5(4):173–9.PubMedPubMedCentral
176.
Zurück zum Zitat Boesch M, Sopper S, Wolf D. Ionophore antibiotics as cancer stem cell-selective drugs: open questions. Oncologist. 2016;21(11):1291–3.PubMedPubMedCentral Boesch M, Sopper S, Wolf D. Ionophore antibiotics as cancer stem cell-selective drugs: open questions. Oncologist. 2016;21(11):1291–3.PubMedPubMedCentral
177.
Zurück zum Zitat Vaysberg M, Balatoni CE, Nepomuceno RR, Krams SM, Martinez OM. Rapamycin inhibits proliferation of epstein-barr virus-positive B-cell lymphomas through modulation of cell-cycle protein expression. Transplantation. 2007;83(8):1114–21.PubMed Vaysberg M, Balatoni CE, Nepomuceno RR, Krams SM, Martinez OM. Rapamycin inhibits proliferation of epstein-barr virus-positive B-cell lymphomas through modulation of cell-cycle protein expression. Transplantation. 2007;83(8):1114–21.PubMed
179.
Zurück zum Zitat Young RC, Ozols RF, Myers CE. The anthracycline antineoplastic drugs. N Engl J Med. 1981;305(3):139–53.PubMed Young RC, Ozols RF, Myers CE. The anthracycline antineoplastic drugs. N Engl J Med. 1981;305(3):139–53.PubMed
180.
Zurück zum Zitat Garg M, Kanojia D, Mayakonda A, Ganesan TS, Sadhanandhan B, Suresh S, Nagare RP, Said JW, Doan NB, Ding LW, Baloglu E. Selinexor (KPT-330) has antitumor activity against anaplastic thyroid carcinoma in vitro and in vivo and enhances sensitivity to doxorubicin. Sci Rep. 2017;7(1):9749.PubMedPubMedCentral Garg M, Kanojia D, Mayakonda A, Ganesan TS, Sadhanandhan B, Suresh S, Nagare RP, Said JW, Doan NB, Ding LW, Baloglu E. Selinexor (KPT-330) has antitumor activity against anaplastic thyroid carcinoma in vitro and in vivo and enhances sensitivity to doxorubicin. Sci Rep. 2017;7(1):9749.PubMedPubMedCentral
181.
Zurück zum Zitat Garg M, Nagata Y, Kanojia D, Mayakonda A, Yoshida K, Haridas Keloth S, Zang ZJ, Okuno Y, Shiraishi Y, Chiba K, Tanaka H. Profiling of somatic mutations in acute myeloid leukemia with FLT3-ITD at diagnosis and relapse. Blood J Am Soc Hematol. 2015;126(22):2491–501. Garg M, Nagata Y, Kanojia D, Mayakonda A, Yoshida K, Haridas Keloth S, Zang ZJ, Okuno Y, Shiraishi Y, Chiba K, Tanaka H. Profiling of somatic mutations in acute myeloid leukemia with FLT3-ITD at diagnosis and relapse. Blood J Am Soc Hematol. 2015;126(22):2491–501.
182.
Zurück zum Zitat Boyle KE, Boger DL, Wroe A, Vazquez M. Duocarmycin SA, a potent antitumor antibiotic, sensitizes glioblastoma cells to proton radiation. Bioorg Med Chem Lett. 2018;28(16):2688–92.PubMedPubMedCentral Boyle KE, Boger DL, Wroe A, Vazquez M. Duocarmycin SA, a potent antitumor antibiotic, sensitizes glioblastoma cells to proton radiation. Bioorg Med Chem Lett. 2018;28(16):2688–92.PubMedPubMedCentral
183.
Zurück zum Zitat Wang TW, Yuan H, Diao WL, Yang R, Zhao XZ, Guo HQ. Comparison of gemcitabine and anthracycline antibiotics in prevention of superficial bladder cancer recurrence. BMC Urol. 2019;19:1–5. Wang TW, Yuan H, Diao WL, Yang R, Zhao XZ, Guo HQ. Comparison of gemcitabine and anthracycline antibiotics in prevention of superficial bladder cancer recurrence. BMC Urol. 2019;19:1–5.
184.
Zurück zum Zitat Panchuk RR, Lehka LV, Terenzi A, Matselyukh BP, Rohr J, Jha AK, Downey T, Kril IJ, Herbacek I, van Schoonhoven S, Heffeter P. Rapid generation of hydrogen peroxide contributes to the complex cell death induction by the angucycline antibiotic landomycin E. Free Radical Biol Med. 2017;1(106):134–47. Panchuk RR, Lehka LV, Terenzi A, Matselyukh BP, Rohr J, Jha AK, Downey T, Kril IJ, Herbacek I, van Schoonhoven S, Heffeter P. Rapid generation of hydrogen peroxide contributes to the complex cell death induction by the angucycline antibiotic landomycin E. Free Radical Biol Med. 2017;1(106):134–47.
185.
Zurück zum Zitat Chong CR, Xu J, Lu J, Bhat S, Sullivan DJ Jr, Liu JO. Inhibition of angiogenesis by the antifungal drug itraconazole. ACS Chem Biol. 2007;2(4):263–70.PubMed Chong CR, Xu J, Lu J, Bhat S, Sullivan DJ Jr, Liu JO. Inhibition of angiogenesis by the antifungal drug itraconazole. ACS Chem Biol. 2007;2(4):263–70.PubMed
186.
Zurück zum Zitat Nacev BA, Grassi P, Dell A, Haslam SM, Liu JO. The antifungal drug itraconazole inhibits vascular endothelial growth factor receptor 2 (VEGFR2) glycosylation, trafficking, and signaling in endothelial cells. J Biol Chem. 2011;286(51):44045–56.PubMedPubMedCentral Nacev BA, Grassi P, Dell A, Haslam SM, Liu JO. The antifungal drug itraconazole inhibits vascular endothelial growth factor receptor 2 (VEGFR2) glycosylation, trafficking, and signaling in endothelial cells. J Biol Chem. 2011;286(51):44045–56.PubMedPubMedCentral
187.
Zurück zum Zitat Aftab BT, Dobromilskaya I, Liu JO, Rudin CM. Itraconazole inhibits angiogenesis and tumor growth in non–small cell lung cancer. Can Res. 2011;71(21):6764–72. Aftab BT, Dobromilskaya I, Liu JO, Rudin CM. Itraconazole inhibits angiogenesis and tumor growth in non–small cell lung cancer. Can Res. 2011;71(21):6764–72.
188.
Zurück zum Zitat Liu R, Li J, Zhang T, Zou L, Chen Y, Wang K, Lei Y, Yuan K, Li Y, Lan J, Cheng L. Itraconazole suppresses the growth of glioblastoma through induction of autophagy: involvement of abnormal cholesterol trafficking. Autophagy. 2014;10(7):1241–55.PubMedPubMedCentral Liu R, Li J, Zhang T, Zou L, Chen Y, Wang K, Lei Y, Yuan K, Li Y, Lan J, Cheng L. Itraconazole suppresses the growth of glioblastoma through induction of autophagy: involvement of abnormal cholesterol trafficking. Autophagy. 2014;10(7):1241–55.PubMedPubMedCentral
189.
Zurück zum Zitat Kim J, Tang JY, Gong R, Kim J, Lee JJ, Clemons KV, Chong CR, Chang KS, Fereshteh M, Gardner D, Reya T. Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell. 2010;17(4):388–99.PubMedPubMedCentral Kim J, Tang JY, Gong R, Kim J, Lee JJ, Clemons KV, Chong CR, Chang KS, Fereshteh M, Gardner D, Reya T. Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell. 2010;17(4):388–99.PubMedPubMedCentral
190.
Zurück zum Zitat You M, Varona-Santos J, Singh S, Robbins DJ, Savaraj N, Nguyen DM. Targeting of the Hedgehog signal transduction pathway suppresses survival of malignant pleural mesothelioma cells in vitro. J Thorac Cardiovasc Surg. 2014;147(1):508–16.PubMed You M, Varona-Santos J, Singh S, Robbins DJ, Savaraj N, Nguyen DM. Targeting of the Hedgehog signal transduction pathway suppresses survival of malignant pleural mesothelioma cells in vitro. J Thorac Cardiovasc Surg. 2014;147(1):508–16.PubMed
191.
Zurück zum Zitat Antonarakis ES, Heath EI, Smith DC, Rathkopf D, Blackford AL, Danila DC, King S, Frost A, Ajiboye AS, Zhao M, Mendonca J. Repurposing itraconazole as a treatment for advanced prostate cancer: a noncomparative randomized phase II trial in men with metastatic castration-resistant prostate cancer. Oncologist. 2013;18(2):163–73.PubMedPubMedCentral Antonarakis ES, Heath EI, Smith DC, Rathkopf D, Blackford AL, Danila DC, King S, Frost A, Ajiboye AS, Zhao M, Mendonca J. Repurposing itraconazole as a treatment for advanced prostate cancer: a noncomparative randomized phase II trial in men with metastatic castration-resistant prostate cancer. Oncologist. 2013;18(2):163–73.PubMedPubMedCentral
192.
Zurück zum Zitat Rudin CM, Brahmer JR, Juergens RA, Hann CL, Ettinger DS, Sebree R, Smith R, Aftab BT, Huang P, Liu JO. Phase 2 study of pemetrexed and itraconazole as second-line therapy for metastatic nonsquamous non–small-cell lung cancer. J Thorac Oncol. 2013;8(5):619–23.PubMedPubMedCentral Rudin CM, Brahmer JR, Juergens RA, Hann CL, Ettinger DS, Sebree R, Smith R, Aftab BT, Huang P, Liu JO. Phase 2 study of pemetrexed and itraconazole as second-line therapy for metastatic nonsquamous non–small-cell lung cancer. J Thorac Oncol. 2013;8(5):619–23.PubMedPubMedCentral
193.
Zurück zum Zitat Ringshausen I, Feuerstacke Y, Krainz P, den Hollander J, Hermann K, Buck A, Peschel C, zum Meyer Bueschenfelde C. Antifungal therapy with itraconazole impairs the anti-lymphoma effects of rituximab by inhibiting recruitment of CD20 to cell surface lipid rafts. Cancer Res. 2010;70(11):4292–6.PubMed Ringshausen I, Feuerstacke Y, Krainz P, den Hollander J, Hermann K, Buck A, Peschel C, zum Meyer Bueschenfelde C. Antifungal therapy with itraconazole impairs the anti-lymphoma effects of rituximab by inhibiting recruitment of CD20 to cell surface lipid rafts. Cancer Res. 2010;70(11):4292–6.PubMed
194.
Zurück zum Zitat Ali ES, Mitra K, Akter S, Ramproshad S, Mondal B, Khan IN, Islam MT, Sharifi-Rad J, Calina D, Cho WC. Recent advances and limitations of mTOR inhibitors in the treatment of cancer. Cancer Cell Int. 2022;22(1):1–6. Ali ES, Mitra K, Akter S, Ramproshad S, Mondal B, Khan IN, Islam MT, Sharifi-Rad J, Calina D, Cho WC. Recent advances and limitations of mTOR inhibitors in the treatment of cancer. Cancer Cell Int. 2022;22(1):1–6.
195.
Zurück zum Zitat Morran DC, Wu J, Jamieson NB, Mrowinska A, Kalna G, Karim SA, Au AY, Scarlett CJ, Chang DK, Pajak MZ, Oien KA. Targeting mTOR dependency in pancreatic cancer. Gut. 2014;63(9):1481–9.PubMed Morran DC, Wu J, Jamieson NB, Mrowinska A, Kalna G, Karim SA, Au AY, Scarlett CJ, Chang DK, Pajak MZ, Oien KA. Targeting mTOR dependency in pancreatic cancer. Gut. 2014;63(9):1481–9.PubMed
196.
Zurück zum Zitat Liu Q, Zhou X, Li C, Zhang X, Li CL. Rapamycin promotes the anticancer action of dihydroartemisinin in breast cancer MDA-MB-231 cells by regulating expression of Atg7 and DAPK. Oncol Lett. 2018;15(4):5781–6.PubMedPubMedCentral Liu Q, Zhou X, Li C, Zhang X, Li CL. Rapamycin promotes the anticancer action of dihydroartemisinin in breast cancer MDA-MB-231 cells by regulating expression of Atg7 and DAPK. Oncol Lett. 2018;15(4):5781–6.PubMedPubMedCentral
197.
Zurück zum Zitat Zhu L, Li XX, Shi L, Wu J, Qian JY, Xia TS, Zhou WB, Sun X, Zhou XJ, Wei JF, Ding Q. Rapamycin enhances the sensitivity of ER-positive breast cancer cells to tamoxifen by upregulating p73 expression. Oncol Rep. 2019;41(1):455–64.PubMed Zhu L, Li XX, Shi L, Wu J, Qian JY, Xia TS, Zhou WB, Sun X, Zhou XJ, Wei JF, Ding Q. Rapamycin enhances the sensitivity of ER-positive breast cancer cells to tamoxifen by upregulating p73 expression. Oncol Rep. 2019;41(1):455–64.PubMed
198.
Zurück zum Zitat Schmeel LC, Schmeel FC, Kim Y, Blaum-Feder S, Schmidt-Wolf IG. Griseofulvin efficiently induces apoptosis in in vitro treatment of lymphoma and multiple myeloma. Anticancer Res. 2017;37(5):2289–95.PubMed Schmeel LC, Schmeel FC, Kim Y, Blaum-Feder S, Schmidt-Wolf IG. Griseofulvin efficiently induces apoptosis in in vitro treatment of lymphoma and multiple myeloma. Anticancer Res. 2017;37(5):2289–95.PubMed
199.
Zurück zum Zitat Rathinasamy K, Jindal B, Asthana J, Singh P, Balaji PV, Panda D. Griseofulvin stabilizes microtubule dynamics, activates p53 and inhibits the proliferation of MCF-7 cells synergistically with vinblastine. BMC Cancer. 2010;10:1–3. Rathinasamy K, Jindal B, Asthana J, Singh P, Balaji PV, Panda D. Griseofulvin stabilizes microtubule dynamics, activates p53 and inhibits the proliferation of MCF-7 cells synergistically with vinblastine. BMC Cancer. 2010;10:1–3.
200.
Zurück zum Zitat Ho YS, Duh JS, Jeng JH, Wang YJ, Liang YC, Lin CH, Tseng CJ, Yu CF, Chen RJ, Lin JK. Griseofulvin potentiates antitumorigenesis effects of nocodazole through induction of apoptosis and G2/M cell cycle arrest in human colorectal cancer cells. Int J Cancer. 2001;91(3):393–401.PubMed Ho YS, Duh JS, Jeng JH, Wang YJ, Liang YC, Lin CH, Tseng CJ, Yu CF, Chen RJ, Lin JK. Griseofulvin potentiates antitumorigenesis effects of nocodazole through induction of apoptosis and G2/M cell cycle arrest in human colorectal cancer cells. Int J Cancer. 2001;91(3):393–401.PubMed
201.
Zurück zum Zitat Panda D, Rathinasamy K, Santra MK, Wilson L. Kinetic suppression of microtubule dynamic instability by griseofulvin: implications for its possible use in the treatment of cancer. Proc Natl Acad Sci. 2005;102(28):9878–83.PubMedPubMedCentral Panda D, Rathinasamy K, Santra MK, Wilson L. Kinetic suppression of microtubule dynamic instability by griseofulvin: implications for its possible use in the treatment of cancer. Proc Natl Acad Sci. 2005;102(28):9878–83.PubMedPubMedCentral
202.
Zurück zum Zitat Liéby-Muller F, Le Baliner QH, Grisoni S, Fournier E, Guilbaud N, Marion F. Synthesis and activities towards resistant cancer cells of sulfone and sulfoxide griseofulvin derivatives. Bioorg Med Chem Lett. 2015;25(10):2078–81.PubMed Liéby-Muller F, Le Baliner QH, Grisoni S, Fournier E, Guilbaud N, Marion F. Synthesis and activities towards resistant cancer cells of sulfone and sulfoxide griseofulvin derivatives. Bioorg Med Chem Lett. 2015;25(10):2078–81.PubMed
203.
Zurück zum Zitat Furtado CM, Marcondes MC, Sola-Penna M, de Souza ML, Zancan P. Clotrimazole preferentially inhibits human breast cancer cell proliferation, viability and glycolysis. PLoS ONE. 2012;7(2):e30462.PubMedPubMedCentral Furtado CM, Marcondes MC, Sola-Penna M, de Souza ML, Zancan P. Clotrimazole preferentially inhibits human breast cancer cell proliferation, viability and glycolysis. PLoS ONE. 2012;7(2):e30462.PubMedPubMedCentral
204.
Zurück zum Zitat Kadavakollu S, Stailey C, Kunapareddy CS, White S. Clotrimazole as a cancer drug: a short review. Med Chem. 2014;4(11):722. Kadavakollu S, Stailey C, Kunapareddy CS, White S. Clotrimazole as a cancer drug: a short review. Med Chem. 2014;4(11):722.
205.
Zurück zum Zitat Robles-Escajeda E, Martínez A, Varela-Ramirez A, Sánchez-Delgado RA, Aguilera RJ. Analysis of the cytotoxic effects of ruthenium–ketoconazole and ruthenium–clotrimazole complexes on cancer cells. Cell Biol Toxicol. 2013;29:431–43.PubMedPubMedCentral Robles-Escajeda E, Martínez A, Varela-Ramirez A, Sánchez-Delgado RA, Aguilera RJ. Analysis of the cytotoxic effects of ruthenium–ketoconazole and ruthenium–clotrimazole complexes on cancer cells. Cell Biol Toxicol. 2013;29:431–43.PubMedPubMedCentral
206.
Zurück zum Zitat Motawi TM, Sadik NA, Fahim SA, Shouman SA. Combination of imatinib and clotrimazole enhances cell growth inhibition in T47D breast cancer cells. Chem Biol Interact. 2015;25(233):147–56. Motawi TM, Sadik NA, Fahim SA, Shouman SA. Combination of imatinib and clotrimazole enhances cell growth inhibition in T47D breast cancer cells. Chem Biol Interact. 2015;25(233):147–56.
207.
Zurück zum Zitat Braun JA, Herrmann AL, Blase JI, Frensemeier K, Bulkescher J, Scheffner M, Galy B, Hoppe-Seyler K, Hoppe-Seyler F. Effects of the antifungal agent ciclopirox in HPV-positive cancer cells: Repression of viral E6/E7 oncogene expression and induction of senescence and apoptosis. Int J Cancer. 2020;146(2):461–74.PubMed Braun JA, Herrmann AL, Blase JI, Frensemeier K, Bulkescher J, Scheffner M, Galy B, Hoppe-Seyler K, Hoppe-Seyler F. Effects of the antifungal agent ciclopirox in HPV-positive cancer cells: Repression of viral E6/E7 oncogene expression and induction of senescence and apoptosis. Int J Cancer. 2020;146(2):461–74.PubMed
208.
Zurück zum Zitat Zhou H, Shen T, Luo Y, Liu L, Chen W, Xu B, Han X, Pang J, Rivera CA, Huang S. The antitumor activity of the fungicide ciclopirox. Int J Cancer. 2010;127(10):2467–77.PubMedPubMedCentral Zhou H, Shen T, Luo Y, Liu L, Chen W, Xu B, Han X, Pang J, Rivera CA, Huang S. The antitumor activity of the fungicide ciclopirox. Int J Cancer. 2010;127(10):2467–77.PubMedPubMedCentral
209.
Zurück zum Zitat Shen T, Shang C, Zhou H, Luo Y, Barzegar M, Odaka Y, Wu Y, Huang S. Ciclopirox inhibits cancer cell proliferation by suppression of Cdc25A. Genes Cancer. 2017;8(3–4):505.PubMedPubMedCentral Shen T, Shang C, Zhou H, Luo Y, Barzegar M, Odaka Y, Wu Y, Huang S. Ciclopirox inhibits cancer cell proliferation by suppression of Cdc25A. Genes Cancer. 2017;8(3–4):505.PubMedPubMedCentral
210.
Zurück zum Zitat Mihailidou C, Papakotoulas P, Papavassiliou AG, Karamouzis MV. Superior efficacy of the antifungal agent ciclopirox olamine over gemcitabine in pancreatic cancer models. Oncotarget. 2018;9(12):10360.PubMed Mihailidou C, Papakotoulas P, Papavassiliou AG, Karamouzis MV. Superior efficacy of the antifungal agent ciclopirox olamine over gemcitabine in pancreatic cancer models. Oncotarget. 2018;9(12):10360.PubMed
211.
Zurück zum Zitat Zhou J, Zhang L, Wang M, Zhou L, Feng X, Yu L, Lan J, Gao W, Zhang C, Bu Y, Huang C. CPX targeting DJ-1 triggers ROS-induced cell death and protective autophagy in colorectal cancer. Theranostics. 2019;9(19):5577.PubMedPubMedCentral Zhou J, Zhang L, Wang M, Zhou L, Feng X, Yu L, Lan J, Gao W, Zhang C, Bu Y, Huang C. CPX targeting DJ-1 triggers ROS-induced cell death and protective autophagy in colorectal cancer. Theranostics. 2019;9(19):5577.PubMedPubMedCentral
212.
Zurück zum Zitat Al-Dali AM, Weiher H, Schmidt-Wolf IG. Utilizing ethacrynic acid and ciclopirox olamine in liver cancer. Oncol Lett. 2018;16(5):6854–60.PubMedPubMedCentral Al-Dali AM, Weiher H, Schmidt-Wolf IG. Utilizing ethacrynic acid and ciclopirox olamine in liver cancer. Oncol Lett. 2018;16(5):6854–60.PubMedPubMedCentral
213.
Zurück zum Zitat Hoffmann H, Kogler H, Heyse W, Matter H, Caspers M, Schummer D, Klemke-Jahn C, Bauer A, Penarier G, Debussche L, Brönstrup M. Discovery, structure elucidation, and biological characterization of nannocystin A, a macrocyclic myxobacterial metabolite with potent antiproliferative properties. Angew Chem Int Ed. 2015;54(35):10145–8. Hoffmann H, Kogler H, Heyse W, Matter H, Caspers M, Schummer D, Klemke-Jahn C, Bauer A, Penarier G, Debussche L, Brönstrup M. Discovery, structure elucidation, and biological characterization of nannocystin A, a macrocyclic myxobacterial metabolite with potent antiproliferative properties. Angew Chem Int Ed. 2015;54(35):10145–8.
214.
Zurück zum Zitat Krastel P, Roggo S, Schirle M, Ross NT, Perruccio F, Aspesi P Jr, Aust T, Buntin K, Estoppey D, Liechty B, Mapa F. Nannocystin A: an elongation factor 1 inhibitor from myxobacteria with differential anti-cancer properties. Angew Chem Int Ed. 2015;54(35):10149–54. Krastel P, Roggo S, Schirle M, Ross NT, Perruccio F, Aspesi P Jr, Aust T, Buntin K, Estoppey D, Liechty B, Mapa F. Nannocystin A: an elongation factor 1 inhibitor from myxobacteria with differential anti-cancer properties. Angew Chem Int Ed. 2015;54(35):10149–54.
215.
Zurück zum Zitat Ishida J, Konishi M, Ebner N, Springer J. Repurposing of approved cardiovascular drugs. J Transl Med. 2016;14:1–5. Ishida J, Konishi M, Ebner N, Springer J. Repurposing of approved cardiovascular drugs. J Transl Med. 2016;14:1–5.
216.
Zurück zum Zitat Mir RH, Shah AJ, Mohi-Ud-Din R, Pottoo FH, Dar M, Jachak SM, Masoodi MH. Natural anti-inflammatory compounds as drug candidates in Alzheimer’s disease. Curr Med Chem. 2021;28(23):4799–825.PubMed Mir RH, Shah AJ, Mohi-Ud-Din R, Pottoo FH, Dar M, Jachak SM, Masoodi MH. Natural anti-inflammatory compounds as drug candidates in Alzheimer’s disease. Curr Med Chem. 2021;28(23):4799–825.PubMed
217.
Zurück zum Zitat Mir RH, Masoodi MH. Anti-inflammatory plant polyphenolics and cellular action mechanisms. Curr Bioact Compd. 2020;16(6):809–17. Mir RH, Masoodi MH. Anti-inflammatory plant polyphenolics and cellular action mechanisms. Curr Bioact Compd. 2020;16(6):809–17.
218.
Zurück zum Zitat Gasic G, Gasic T, Murphy S. Anti-metastatic effect of aspirin. Lancet. 1972;300(7783):932–3. Gasic G, Gasic T, Murphy S. Anti-metastatic effect of aspirin. Lancet. 1972;300(7783):932–3.
219.
Zurück zum Zitat Kolenich J, Mansour E, Flynn A. Haematological effects of aspirin. Lancet. 1972;300(7779):714. Kolenich J, Mansour E, Flynn A. Haematological effects of aspirin. Lancet. 1972;300(7779):714.
220.
Zurück zum Zitat Mir RH, Sawhney G, Verma R, Ahmad B, Kumar P, Ranjana S, Bhagat A, Madishetti S, Ahmed Z, Jachak SM, Choi S. Origanum vulgare L.: in vitro assessment of cytotoxicity, molecular docking studies, antioxidant and anti-inflammatory activity in LPS stimulated RAW 264.7 cells. Med Chem. 2021;17(9):983–93.PubMed Mir RH, Sawhney G, Verma R, Ahmad B, Kumar P, Ranjana S, Bhagat A, Madishetti S, Ahmed Z, Jachak SM, Choi S. Origanum vulgare L.: in vitro assessment of cytotoxicity, molecular docking studies, antioxidant and anti-inflammatory activity in LPS stimulated RAW 264.7 cells. Med Chem. 2021;17(9):983–93.PubMed
221.
Zurück zum Zitat Reimers MS, Bastiaannet E, van Herk-Sukel MP, Lemmens VE, van den Broek CB, van de Velde CJ, de Craen AJ, Liefers GJ. Aspirin use after diagnosis improves survival in older adults with colon cancer: a retrospective cohort study. J Am Geriatr Soc. 2012;60(12):2232–6.PubMed Reimers MS, Bastiaannet E, van Herk-Sukel MP, Lemmens VE, van den Broek CB, van de Velde CJ, de Craen AJ, Liefers GJ. Aspirin use after diagnosis improves survival in older adults with colon cancer: a retrospective cohort study. J Am Geriatr Soc. 2012;60(12):2232–6.PubMed
222.
Zurück zum Zitat McCowan C, Munro AJ, Donnan PT, Steele RJ. Use of aspirin post-diagnosis in a cohort of patients with colorectal cancer and its association with all-cause and colorectal cancer specific mortality. Eur J Cancer. 2013;49(5):1049–57.PubMed McCowan C, Munro AJ, Donnan PT, Steele RJ. Use of aspirin post-diagnosis in a cohort of patients with colorectal cancer and its association with all-cause and colorectal cancer specific mortality. Eur J Cancer. 2013;49(5):1049–57.PubMed
223.
Zurück zum Zitat Goh HH, Leong WQ, Chew MH, Pan YS, Tony LK, Chew L, Tan IB, Toh HC, Tang CL, Fu WP, Chia WK. Post-operative aspirin use and colorectal cancer-specific survival in patients with stage I-III colorectal cancer. Anticancer Res. 2014;34(12):7407–14.PubMed Goh HH, Leong WQ, Chew MH, Pan YS, Tony LK, Chew L, Tan IB, Toh HC, Tang CL, Fu WP, Chia WK. Post-operative aspirin use and colorectal cancer-specific survival in patients with stage I-III colorectal cancer. Anticancer Res. 2014;34(12):7407–14.PubMed
224.
Zurück zum Zitat Ahmad G, Hassan R, Dhiman N, Ali A. Assessment of anti-inflammatory activity of 3-acetylmyricadiol in LPSStimulated raw 264.7 macrophages. Comb Chem High Throughput Screen. 2022;25(1):204–10.PubMed Ahmad G, Hassan R, Dhiman N, Ali A. Assessment of anti-inflammatory activity of 3-acetylmyricadiol in LPSStimulated raw 264.7 macrophages. Comb Chem High Throughput Screen. 2022;25(1):204–10.PubMed
225.
Zurück zum Zitat Zhou Q, Zhao S, Gan L, Wang Z, Peng S, Li Q, Liu H, Liu X, Wang Z, Shi Q, Estill J. Use of non-steroidal anti-inflammatory drugs and adverse outcomes during the COVID-19 pandemic: a systematic review and meta-analysis. EClinicalMedicine. 2022;1:46. Zhou Q, Zhao S, Gan L, Wang Z, Peng S, Li Q, Liu H, Liu X, Wang Z, Shi Q, Estill J. Use of non-steroidal anti-inflammatory drugs and adverse outcomes during the COVID-19 pandemic: a systematic review and meta-analysis. EClinicalMedicine. 2022;1:46.
226.
Zurück zum Zitat Palayoor ST, Bump EA, Calderwood SK, Bartol S, Coleman CN. Combined antitumor effect of radiation and ibuprofen in human prostate carcinoma cells. Clin Cancer Res. 1998;4(3):763–71.PubMed Palayoor ST, Bump EA, Calderwood SK, Bartol S, Coleman CN. Combined antitumor effect of radiation and ibuprofen in human prostate carcinoma cells. Clin Cancer Res. 1998;4(3):763–71.PubMed
227.
Zurück zum Zitat Hassan Mir R, Godavari G, Siddiqui NA, Ahmad B, Mothana RA, Ullah R, Almarfadi OM, Jachak SM, Masoodi MH. Design, synthesis, molecular modelling, and biological evaluation of oleanolic acid-arylidene derivatives as potential anti-inflammatory agents. Drug Des Dev Ther. 2021;4:385–97. Hassan Mir R, Godavari G, Siddiqui NA, Ahmad B, Mothana RA, Ullah R, Almarfadi OM, Jachak SM, Masoodi MH. Design, synthesis, molecular modelling, and biological evaluation of oleanolic acid-arylidene derivatives as potential anti-inflammatory agents. Drug Des Dev Ther. 2021;4:385–97.
228.
Zurück zum Zitat Akrami H, Aminzadeh S, Fallahi H. Inhibitory effect of ibuprofen on tumor survival and angiogenesis in gastric cancer cell. Tumor Biol. 2015;36:3237–43. Akrami H, Aminzadeh S, Fallahi H. Inhibitory effect of ibuprofen on tumor survival and angiogenesis in gastric cancer cell. Tumor Biol. 2015;36:3237–43.
229.
Zurück zum Zitat Redpath M, Marques CM, Dibden C, Waddon A, Lalla R, MacNeil S. Ibuprofen and hydrogel-released ibuprofen in the reduction of inflammation-induced migration in melanoma cells. Br J Dermatol. 2009;161(1):25–33.PubMed Redpath M, Marques CM, Dibden C, Waddon A, Lalla R, MacNeil S. Ibuprofen and hydrogel-released ibuprofen in the reduction of inflammation-induced migration in melanoma cells. Br J Dermatol. 2009;161(1):25–33.PubMed
230.
Zurück zum Zitat Mir RH, Wani TU, Jan R, Shah AJ, Sabreen S, Mir PA, Rasool S, Masoodi MH, Bhat ZA. Nigella sativa as a therapeutic candidate for arthritis and related disorders. In: Khan A, Rehman M, editors. Black seeds (Nigella sativa). Amsterdam: Elsevier; 2022. p. 295–312. Mir RH, Wani TU, Jan R, Shah AJ, Sabreen S, Mir PA, Rasool S, Masoodi MH, Bhat ZA. Nigella sativa as a therapeutic candidate for arthritis and related disorders. In: Khan A, Rehman M, editors. Black seeds (Nigella sativa). Amsterdam: Elsevier; 2022. p. 295–312.
231.
Zurück zum Zitat Endo H, Yano M, Okumura Y, Kido H. Ibuprofen enhances the anticancer activity of cisplatin in lung cancer cells by inhibiting the heat shock protein 70. Cell Death Dis. 2014;5(1):e1027.PubMedPubMedCentral Endo H, Yano M, Okumura Y, Kido H. Ibuprofen enhances the anticancer activity of cisplatin in lung cancer cells by inhibiting the heat shock protein 70. Cell Death Dis. 2014;5(1):e1027.PubMedPubMedCentral
232.
Zurück zum Zitat Kolawole OR, Kashfi K. NSAIDs and cancer resolution: new paradigms beyond cyclooxygenase. Int J Mol Sci. 2022;23(3):1432.PubMedPubMedCentral Kolawole OR, Kashfi K. NSAIDs and cancer resolution: new paradigms beyond cyclooxygenase. Int J Mol Sci. 2022;23(3):1432.PubMedPubMedCentral
233.
Zurück zum Zitat Kim MS, Kim JE, Lim DY, Huang Z, Chen H, Langfald A, Lubet RA, Grubbs CJ, Dong Z, Bode AM. Naproxen induces cell-cycle arrest and apoptosis in human urinary bladder cancer cell lines and chemically induced cancers by targeting PI3K. Cancer Prev Res. 2014;7(2):236–45. Kim MS, Kim JE, Lim DY, Huang Z, Chen H, Langfald A, Lubet RA, Grubbs CJ, Dong Z, Bode AM. Naproxen induces cell-cycle arrest and apoptosis in human urinary bladder cancer cell lines and chemically induced cancers by targeting PI3K. Cancer Prev Res. 2014;7(2):236–45.
234.
Zurück zum Zitat Mir RH, Mir PA, Maqbool M, Banday N, Farooq S, Raza SN, Chawla PA. Therapeutic potential of plant-derived flavonoids against inflammation. In: Prasher P, Zacconi F, Dua K, Rathbone M, Withey J, editors. Recent developments in anti-inflammatory therapy. Cambridge: Academic Press; 2023. p. 279–93. Mir RH, Mir PA, Maqbool M, Banday N, Farooq S, Raza SN, Chawla PA. Therapeutic potential of plant-derived flavonoids against inflammation. In: Prasher P, Zacconi F, Dua K, Rathbone M, Withey J, editors. Recent developments in anti-inflammatory therapy. Cambridge: Academic Press; 2023. p. 279–93.
235.
Zurück zum Zitat Suh N, Reddy BS, DeCastro A, Paul S, Lee HJ, Smolarek AK, So JY, Simi B, Wang CX, Janakiram NB, Steele V. Combination of atorvastatin with sulindac or naproxen profoundly inhibits colonic adenocarcinomas by suppressing the p65/β-catenin/cyclin D1 signaling pathway in rats. Cancer Prev Res. 2011;4(11):1895–902. Suh N, Reddy BS, DeCastro A, Paul S, Lee HJ, Smolarek AK, So JY, Simi B, Wang CX, Janakiram NB, Steele V. Combination of atorvastatin with sulindac or naproxen profoundly inhibits colonic adenocarcinomas by suppressing the p65/β-catenin/cyclin D1 signaling pathway in rats. Cancer Prev Res. 2011;4(11):1895–902.
236.
Zurück zum Zitat Srinivas S, Feldman D. A phase II trial of calcitriol and naproxen in recurrent prostate cancer. Anticancer Res. 2009;29(9):3605–10.PubMed Srinivas S, Feldman D. A phase II trial of calcitriol and naproxen in recurrent prostate cancer. Anticancer Res. 2009;29(9):3605–10.PubMed
237.
Zurück zum Zitat Wickström M, Danielsson K, Rickardson L, Gullbo J, Nygren P, Isaksson A, Larsson R, Lövborg H. Pharmacological profiling of disulfiram using human tumor cell lines and human tumor cells from patients. Biochem Pharmacol. 2007;73(1):25–33.PubMed Wickström M, Danielsson K, Rickardson L, Gullbo J, Nygren P, Isaksson A, Larsson R, Lövborg H. Pharmacological profiling of disulfiram using human tumor cell lines and human tumor cells from patients. Biochem Pharmacol. 2007;73(1):25–33.PubMed
238.
Zurück zum Zitat Triscott J, Rose Pambid M, Dunn SE. Concise review: bullseye: targeting cancer stem cells to improve the treatment of gliomas by repurposing disulfiram. Stem Cells. 2015;33(4):1042–6.PubMed Triscott J, Rose Pambid M, Dunn SE. Concise review: bullseye: targeting cancer stem cells to improve the treatment of gliomas by repurposing disulfiram. Stem Cells. 2015;33(4):1042–6.PubMed
239.
Zurück zum Zitat Peterson HI. Effects of prostaglandin synthesis inhibitors on tumor growth and vascularization: experimental studies in the rat. Invasion Metastasis. 1983;3(3):151–9.PubMed Peterson HI. Effects of prostaglandin synthesis inhibitors on tumor growth and vascularization: experimental studies in the rat. Invasion Metastasis. 1983;3(3):151–9.PubMed
240.
Zurück zum Zitat Hixson LJ, Alberts DS, Krutzsch M, Einsphar J, Brendel K, Gross PH, Paranka NS, Baier M, Emerson S, Pamukcu R. Antiproliferative effect of nonsteroidal antiinflammatory drugs against human colon cancer cells. Cancer Epidemiol Biomark Prev. 1994;3(5):433–8. Hixson LJ, Alberts DS, Krutzsch M, Einsphar J, Brendel K, Gross PH, Paranka NS, Baier M, Emerson S, Pamukcu R. Antiproliferative effect of nonsteroidal antiinflammatory drugs against human colon cancer cells. Cancer Epidemiol Biomark Prev. 1994;3(5):433–8.
241.
Zurück zum Zitat Dar MO, Mir RH, Mohiuddin R, Masoodi MH, Sofi FA. Metal complexes of xanthine and its derivatives: Synthesis and biological activity. J Inorg Biochem. 2023;10:112290. Dar MO, Mir RH, Mohiuddin R, Masoodi MH, Sofi FA. Metal complexes of xanthine and its derivatives: Synthesis and biological activity. J Inorg Biochem. 2023;10:112290.
242.
Zurück zum Zitat Mayorek N, Naftali-Shani N, Grunewald M. Diclofenac inhibits tumor growth in a murine model of pancreatic cancer by modulation of VEGF levels and arginase activity. PLoS ONE. 2010;5(9):e12715.PubMedPubMedCentral Mayorek N, Naftali-Shani N, Grunewald M. Diclofenac inhibits tumor growth in a murine model of pancreatic cancer by modulation of VEGF levels and arginase activity. PLoS ONE. 2010;5(9):e12715.PubMedPubMedCentral
243.
Zurück zum Zitat Valle BL, D’Souza T, Becker KG, Wood WH III, Zhang Y, Wersto RP, Morin PJ. Non-steroidal anti-inflammatory drugs decrease E2F1 expression and inhibit cell growth in ovarian cancer cells. PLoS ONE. 2013;8(4):e61836.PubMedPubMedCentral Valle BL, D’Souza T, Becker KG, Wood WH III, Zhang Y, Wersto RP, Morin PJ. Non-steroidal anti-inflammatory drugs decrease E2F1 expression and inhibit cell growth in ovarian cancer cells. PLoS ONE. 2013;8(4):e61836.PubMedPubMedCentral
244.
Zurück zum Zitat Cecere F, Iuliano A, Albano F, Zappelli C, Castellano I, Grimaldi P, Masullo M, De Vendittis E, Ruocco MR. Diclofenac-induced apoptosis in the neuroblastoma cell line SH-SY5Y: possible involvement of the mitochondrial superoxide dismutase. Biomed Res Int. 2010;1:2010. Cecere F, Iuliano A, Albano F, Zappelli C, Castellano I, Grimaldi P, Masullo M, De Vendittis E, Ruocco MR. Diclofenac-induced apoptosis in the neuroblastoma cell line SH-SY5Y: possible involvement of the mitochondrial superoxide dismutase. Biomed Res Int. 2010;1:2010.
245.
Zurück zum Zitat Brinkhuizen T, Frencken KJ, Nelemans PJ, Hoff ML, Kelleners-Smeets NW, Zur Hausen A, van der Horst MP, Rennspiess D, Winnepenninckx VJ, van Steensel MA, Mosterd K. The effect of topical diclofenac 3% and calcitriol 3 μg/g on superficial basal cell carcinoma (sBCC) and nodular basal cell carcinoma (nBCC): a phase II, randomized controlled trial. J Am Acad Dermatol. 2016;75(1):126–34.PubMed Brinkhuizen T, Frencken KJ, Nelemans PJ, Hoff ML, Kelleners-Smeets NW, Zur Hausen A, van der Horst MP, Rennspiess D, Winnepenninckx VJ, van Steensel MA, Mosterd K. The effect of topical diclofenac 3% and calcitriol 3 μg/g on superficial basal cell carcinoma (sBCC) and nodular basal cell carcinoma (nBCC): a phase II, randomized controlled trial. J Am Acad Dermatol. 2016;75(1):126–34.PubMed
246.
Zurück zum Zitat Hamy AS, Tury S, Wang X, Gao J, Pierga JY, Giacchetti S, Brain E, Pistilli B, Marty M, Espié M, Benchimol G. Celecoxib with neoadjuvant chemotherapy for breast cancer might worsen outcomes differentially by COX-2 expression and ER status: exploratory analysis of the REMAGUS02 trial. J Clin Oncol. 2019;37(8):624.PubMedPubMedCentral Hamy AS, Tury S, Wang X, Gao J, Pierga JY, Giacchetti S, Brain E, Pistilli B, Marty M, Espié M, Benchimol G. Celecoxib with neoadjuvant chemotherapy for breast cancer might worsen outcomes differentially by COX-2 expression and ER status: exploratory analysis of the REMAGUS02 trial. J Clin Oncol. 2019;37(8):624.PubMedPubMedCentral
247.
Zurück zum Zitat Edelman MJ, Wang X, Hodgson L, Cheney RT, Baggstrom MQ, Thomas SP, Gajra A, Bertino E, Reckamp KL, Molina J, Schiller JH. Phase III randomized, placebo-controlled, double-blind trial of celecoxib in addition to standard chemotherapy for advanced non–small-cell lung cancer with cyclooxygenase-2 overexpression: CALGB 30801 (Alliance). J Clin Oncol. 2017;35(19):2184.PubMedPubMedCentral Edelman MJ, Wang X, Hodgson L, Cheney RT, Baggstrom MQ, Thomas SP, Gajra A, Bertino E, Reckamp KL, Molina J, Schiller JH. Phase III randomized, placebo-controlled, double-blind trial of celecoxib in addition to standard chemotherapy for advanced non–small-cell lung cancer with cyclooxygenase-2 overexpression: CALGB 30801 (Alliance). J Clin Oncol. 2017;35(19):2184.PubMedPubMedCentral
248.
Zurück zum Zitat Kelly JD, Tan WS, Porta N, Mostafid H, Huddart R, Protheroe A, Bogle R, Blazeby J, Palmer A, Cresswell J, Johnson M. BOXIT—a randomised phase III placebo-controlled trial evaluating the addition of celecoxib to standard treatment of transitional cell carcinoma of the bladder (CRUK/07/004). Eur Urol. 2019;75(4):593–601.PubMed Kelly JD, Tan WS, Porta N, Mostafid H, Huddart R, Protheroe A, Bogle R, Blazeby J, Palmer A, Cresswell J, Johnson M. BOXIT—a randomised phase III placebo-controlled trial evaluating the addition of celecoxib to standard treatment of transitional cell carcinoma of the bladder (CRUK/07/004). Eur Urol. 2019;75(4):593–601.PubMed
249.
Zurück zum Zitat Mir RH, Banday N, Sabreen S, Shah AJ, Jan R, Wani TU, Farooq S, Bhat ZA. Resveratrol: a potential drug candidate with multispectrum therapeutic application. Stud Nat Prod Chem. 2022;1(73):99–137. Mir RH, Banday N, Sabreen S, Shah AJ, Jan R, Wani TU, Farooq S, Bhat ZA. Resveratrol: a potential drug candidate with multispectrum therapeutic application. Stud Nat Prod Chem. 2022;1(73):99–137.
250.
Zurück zum Zitat Liu X, Wu Y, Zhou Z, Huang M, Deng W, Wang Y, Zhou X, Chen L, Li Y, Zeng T, Wang G. Celecoxib inhibits the epithelial-to-mesenchymal transition in bladder cancer via the miRNA-145/TGFBR2/Smad3 axis. Int J Mol Med. 2019;44(2):683–93.PubMedPubMedCentral Liu X, Wu Y, Zhou Z, Huang M, Deng W, Wang Y, Zhou X, Chen L, Li Y, Zeng T, Wang G. Celecoxib inhibits the epithelial-to-mesenchymal transition in bladder cancer via the miRNA-145/TGFBR2/Smad3 axis. Int J Mol Med. 2019;44(2):683–93.PubMedPubMedCentral
251.
Zurück zum Zitat Dai H, Zhang S, Ma R, Pan L. Celecoxib inhibits hepatocellular carcinoma cell growth and migration by targeting PNO1. Med Sci Monit. 2019;25:7351.PubMedPubMedCentral Dai H, Zhang S, Ma R, Pan L. Celecoxib inhibits hepatocellular carcinoma cell growth and migration by targeting PNO1. Med Sci Monit. 2019;25:7351.PubMedPubMedCentral
252.
Zurück zum Zitat Qiu Z, Zhang C, Zhou J, Hu J, Sheng L, Li X, Chen L, Li X, Deng X, Zheng G. Celecoxib alleviates AKT/c-Met-triggered rapid hepatocarcinogenesis by suppressing a novel COX-2/AKT/FASN cascade. Mol Carcinog. 2019;58(1):31–41.PubMed Qiu Z, Zhang C, Zhou J, Hu J, Sheng L, Li X, Chen L, Li X, Deng X, Zheng G. Celecoxib alleviates AKT/c-Met-triggered rapid hepatocarcinogenesis by suppressing a novel COX-2/AKT/FASN cascade. Mol Carcinog. 2019;58(1):31–41.PubMed
253.
Zurück zum Zitat Tołoczko-Iwaniuk N, Dziemiańczyk-Pakieła D, Celińska-Janowicz K, Zaręba I, Klupczyńska A, Kokot ZJ, Nowaszewska BK, Reszeć J, Borys J, Miltyk W. Proline-dependent induction of apoptosis in oral squamous cell carcinoma (OSCC)—the effect of celecoxib. Cancers. 2020;12(1):136.PubMedPubMedCentral Tołoczko-Iwaniuk N, Dziemiańczyk-Pakieła D, Celińska-Janowicz K, Zaręba I, Klupczyńska A, Kokot ZJ, Nowaszewska BK, Reszeć J, Borys J, Miltyk W. Proline-dependent induction of apoptosis in oral squamous cell carcinoma (OSCC)—the effect of celecoxib. Cancers. 2020;12(1):136.PubMedPubMedCentral
254.
Zurück zum Zitat Velmurugan BK, Hua CH, Tsai MH, Lee CP, Chung CM, Ko YC. Combination of celecoxib and calyculin-A inhibits epithelial-mesenchymal transition in human oral cancer cells. Biotech Histochem. 2020;95(5):341–8.PubMed Velmurugan BK, Hua CH, Tsai MH, Lee CP, Chung CM, Ko YC. Combination of celecoxib and calyculin-A inhibits epithelial-mesenchymal transition in human oral cancer cells. Biotech Histochem. 2020;95(5):341–8.PubMed
255.
Zurück zum Zitat Aboelella NS, Brandle C, Okoko O, Gazi MY, Ding ZC, Xu H, Gorman G, Bollag R, Davila ML, Bryan LJ, Munn DH. Indomethacin-induced oxidative stress enhances death receptor 5 signaling and sensitizes tumor cells to adoptive T-cell therapy. J Immunother Cancer. 2022;10(7):e004938.PubMedPubMedCentral Aboelella NS, Brandle C, Okoko O, Gazi MY, Ding ZC, Xu H, Gorman G, Bollag R, Davila ML, Bryan LJ, Munn DH. Indomethacin-induced oxidative stress enhances death receptor 5 signaling and sensitizes tumor cells to adoptive T-cell therapy. J Immunother Cancer. 2022;10(7):e004938.PubMedPubMedCentral
256.
Zurück zum Zitat Chennamaneni S, Zhong B, Lama R, Su B. COX inhibitors Indomethacin and Sulindac derivatives as antiproliferative agents: synthesis, biological evaluation, and mechanism investigation. Eur J Med Chem. 2012;1(56):17–29. Chennamaneni S, Zhong B, Lama R, Su B. COX inhibitors Indomethacin and Sulindac derivatives as antiproliferative agents: synthesis, biological evaluation, and mechanism investigation. Eur J Med Chem. 2012;1(56):17–29.
257.
Zurück zum Zitat Touhey S, O’Connor R, Plunkett S, Maguire A, Clynes M. Structure–activity relationship of indomethacin analogues for MRP-1, COX-1 and COX-2 inhibition: identification of novel chemotherapeutic drug resistance modulators. Eur J Cancer. 2002;38(12):1661–70.PubMed Touhey S, O’Connor R, Plunkett S, Maguire A, Clynes M. Structure–activity relationship of indomethacin analogues for MRP-1, COX-1 and COX-2 inhibition: identification of novel chemotherapeutic drug resistance modulators. Eur J Cancer. 2002;38(12):1661–70.PubMed
258.
Zurück zum Zitat Guo YC, Chang CM, Hsu WL, Chiu SJ, Tsai YT, Chou YH, Hou MF, Wang JY, Lee MH, Tsai KL, Chang WC. Indomethacin inhibits cancer cell migration via attenuation of cellular calcium mobilization. Molecules. 2013;18(6):6584–96.PubMedPubMedCentral Guo YC, Chang CM, Hsu WL, Chiu SJ, Tsai YT, Chou YH, Hou MF, Wang JY, Lee MH, Tsai KL, Chang WC. Indomethacin inhibits cancer cell migration via attenuation of cellular calcium mobilization. Molecules. 2013;18(6):6584–96.PubMedPubMedCentral
259.
Zurück zum Zitat Zhang YJ, Bao YJ, Dai Q, Yang WY, Cheng P, Zhu LM, Wang BJ, Jiang FH. mTOR signaling is involved in indomethacin and nimesulide suppression of colorectal cancer cell growth via a COX-2 independent pathway. Ann Surg Oncol. 2011;18:580–8.PubMed Zhang YJ, Bao YJ, Dai Q, Yang WY, Cheng P, Zhu LM, Wang BJ, Jiang FH. mTOR signaling is involved in indomethacin and nimesulide suppression of colorectal cancer cell growth via a COX-2 independent pathway. Ann Surg Oncol. 2011;18:580–8.PubMed
260.
Zurück zum Zitat Brunelli C, Amici C, Angelini M, Fracassi C, Belardo G, Santoro MG. The non-steroidal anti-inflammatory drug indomethacin activates the eIF2α kinase PKR, causing a translational block in human colorectal cancer cells. Biochemical journal. 2012;443(2):379–86.PubMed Brunelli C, Amici C, Angelini M, Fracassi C, Belardo G, Santoro MG. The non-steroidal anti-inflammatory drug indomethacin activates the eIF2α kinase PKR, causing a translational block in human colorectal cancer cells. Biochemical journal. 2012;443(2):379–86.PubMed
261.
Zurück zum Zitat Cheng YL, Zhang GY, Li C, Lin J. Screening for novel protein targets of indomethacin in HCT116 human colon cancer cells using proteomics. Oncol Lett. 2013;6(5):1222–8.PubMedPubMedCentral Cheng YL, Zhang GY, Li C, Lin J. Screening for novel protein targets of indomethacin in HCT116 human colon cancer cells using proteomics. Oncol Lett. 2013;6(5):1222–8.PubMedPubMedCentral
262.
Zurück zum Zitat Lin CC, Suen KM, Stainthorp A, Wieteska L, Biggs GS, Leitão A, Montanari CA, Ladbury JE. Targeting the Shc-EGFR interaction with indomethacin inhibits MAP kinase pathway signalling. Cancer Lett. 2019;10(457):86–97. Lin CC, Suen KM, Stainthorp A, Wieteska L, Biggs GS, Leitão A, Montanari CA, Ladbury JE. Targeting the Shc-EGFR interaction with indomethacin inhibits MAP kinase pathway signalling. Cancer Lett. 2019;10(457):86–97.
263.
Zurück zum Zitat Mazumder S, De R, Debsharma S, Bindu S, Maity P, Sarkar S, Saha SJ, Siddiqui AA, Banerjee C, Nag S, Saha D. Indomethacin impairs mitochondrial dynamics by activating the PKCζ–p38–DRP1 pathway and inducing apoptosis in gastric cancer and normal mucosal cells. J Biol Chem. 2019;294(20):8238–58.PubMedPubMedCentral Mazumder S, De R, Debsharma S, Bindu S, Maity P, Sarkar S, Saha SJ, Siddiqui AA, Banerjee C, Nag S, Saha D. Indomethacin impairs mitochondrial dynamics by activating the PKCζ–p38–DRP1 pathway and inducing apoptosis in gastric cancer and normal mucosal cells. J Biol Chem. 2019;294(20):8238–58.PubMedPubMedCentral
264.
Zurück zum Zitat Cuzick J. Preventive therapy for cancer. Lancet Oncol. 2017;18(8):e472-82.PubMed Cuzick J. Preventive therapy for cancer. Lancet Oncol. 2017;18(8):e472-82.PubMed
265.
Zurück zum Zitat Thun MJ, Jacobs EJ, Patrono C. The role of aspirin in cancer prevention. Nat Rev Clin Oncol. 2012;9(5):259–67.PubMed Thun MJ, Jacobs EJ, Patrono C. The role of aspirin in cancer prevention. Nat Rev Clin Oncol. 2012;9(5):259–67.PubMed
266.
Zurück zum Zitat Rostom A, Dubé C, Lewin G, Tsertsvadze A, Barrowman N, Code C, Sampson M, Moher D. Nonsteroidal anti-inflammatory drugs and cyclooxygenase-2 inhibitors for primary prevention of colorectal cancer: a systematic review prepared for the US Preventive Services Task Force. Ann Intern Med. 2007;146(5):376–89.PubMed Rostom A, Dubé C, Lewin G, Tsertsvadze A, Barrowman N, Code C, Sampson M, Moher D. Nonsteroidal anti-inflammatory drugs and cyclooxygenase-2 inhibitors for primary prevention of colorectal cancer: a systematic review prepared for the US Preventive Services Task Force. Ann Intern Med. 2007;146(5):376–89.PubMed
267.
Zurück zum Zitat Nagaraj AB, Wang QQ, Joseph P, Zheng C, Chen Y, Kovalenko O, Singh S, Armstrong A, Resnick K, Zanotti K, Waggoner S. Using a novel computational drug-repositioning approach (DrugPredict) to rapidly identify potent drug candidates for cancer treatment. Oncogene. 2018;37(3):403–14.PubMed Nagaraj AB, Wang QQ, Joseph P, Zheng C, Chen Y, Kovalenko O, Singh S, Armstrong A, Resnick K, Zanotti K, Waggoner S. Using a novel computational drug-repositioning approach (DrugPredict) to rapidly identify potent drug candidates for cancer treatment. Oncogene. 2018;37(3):403–14.PubMed
268.
Zurück zum Zitat Kundu CN, Das S, Nayak A, Satapathy SR, Das D, Siddharth S. Anti-malarials are anti-cancers and vice versa–one arrow two sparrows. Acta Trop. 2015;1(149):113–27. Kundu CN, Das S, Nayak A, Satapathy SR, Das D, Siddharth S. Anti-malarials are anti-cancers and vice versa–one arrow two sparrows. Acta Trop. 2015;1(149):113–27.
269.
Zurück zum Zitat Cui L, Su XZ. Discovery, mechanisms of action and combination therapy of artemisinin. Expert Rev Anti Infect Ther. 2009;7(8):999–1013.PubMedPubMedCentral Cui L, Su XZ. Discovery, mechanisms of action and combination therapy of artemisinin. Expert Rev Anti Infect Ther. 2009;7(8):999–1013.PubMedPubMedCentral
270.
Zurück zum Zitat Augustin Y, Krishna S, Kumar D, Pantziarka P. The wisdom of crowds and the repurposing of artesunate as an anticancer drug. Ecancermedicalscience. 2015;9:ed50.PubMedPubMedCentral Augustin Y, Krishna S, Kumar D, Pantziarka P. The wisdom of crowds and the repurposing of artesunate as an anticancer drug. Ecancermedicalscience. 2015;9:ed50.PubMedPubMedCentral
271.
Zurück zum Zitat Holien T, Olsen OE, Misund K, Hella H, Waage A, Rø TB, Sundan A. Lymphoma and myeloma cells are highly sensitive to growth arrest and apoptosis induced by artesunate. Eur J Haematol. 2013;91(4):339–46.PubMed Holien T, Olsen OE, Misund K, Hella H, Waage A, Rø TB, Sundan A. Lymphoma and myeloma cells are highly sensitive to growth arrest and apoptosis induced by artesunate. Eur J Haematol. 2013;91(4):339–46.PubMed
272.
Zurück zum Zitat Vandewynckel YP, Laukens D, Geerts A, Vanhove C, Descamps B, Colle I, Devisscher L, Bogaerts E, Paridaens A, Verhelst X, Van Steenkiste C. Therapeutic effects of artesunate in hepatocellular carcinoma: repurposing an ancient antimalarial agent. Eur J Gastroenterol Hepatol. 2014;26(8):861–70.PubMed Vandewynckel YP, Laukens D, Geerts A, Vanhove C, Descamps B, Colle I, Devisscher L, Bogaerts E, Paridaens A, Verhelst X, Van Steenkiste C. Therapeutic effects of artesunate in hepatocellular carcinoma: repurposing an ancient antimalarial agent. Eur J Gastroenterol Hepatol. 2014;26(8):861–70.PubMed
273.
Zurück zum Zitat Hou J, Wang D, Zhang R, Wang H. Experimental therapy of hepatoma with artemisinin and its derivatives: in vitro and in vivo activity, chemosensitization, and mechanisms of action. Clin Cancer Res. 2008;14(17):5519–30.PubMed Hou J, Wang D, Zhang R, Wang H. Experimental therapy of hepatoma with artemisinin and its derivatives: in vitro and in vivo activity, chemosensitization, and mechanisms of action. Clin Cancer Res. 2008;14(17):5519–30.PubMed
274.
Zurück zum Zitat Zhou HJ, Wang WQ, Wu GD, Lee J, Li A. Artesunate inhibits angiogenesis and downregulates vascular endothelial growth factor expression in chronic myeloid leukemia K562 cells. Vascul Pharmacol. 2007;47(2–3):131–8.PubMed Zhou HJ, Wang WQ, Wu GD, Lee J, Li A. Artesunate inhibits angiogenesis and downregulates vascular endothelial growth factor expression in chronic myeloid leukemia K562 cells. Vascul Pharmacol. 2007;47(2–3):131–8.PubMed
275.
Zurück zum Zitat Wang Z, Hu W, Zhang JL, Wu XH, Zhou HJ. Dihydroartemisinin induces autophagy and inhibits the growth of iron-loaded human myeloid leukemia K562 cells via ROS toxicity. FEBS Open Bio. 2012;1(2):103–12. Wang Z, Hu W, Zhang JL, Wu XH, Zhou HJ. Dihydroartemisinin induces autophagy and inhibits the growth of iron-loaded human myeloid leukemia K562 cells via ROS toxicity. FEBS Open Bio. 2012;1(2):103–12.
277.
Zurück zum Zitat Zhou HJ, Wang Z, Li A. Dihydroartemisinin induces apoptosis in human leukemia cells HL60 via downregulation of transferrin receptor expression. Anticancer Drugs. 2008;19(3):247–55.PubMed Zhou HJ, Wang Z, Li A. Dihydroartemisinin induces apoptosis in human leukemia cells HL60 via downregulation of transferrin receptor expression. Anticancer Drugs. 2008;19(3):247–55.PubMed
278.
Zurück zum Zitat Disbrow GL, Baege AC, Kierpiec KA, Yuan H, Centeno JA, Thibodeaux CA, Hartmann D, Schlegel R. Dihydroartemisinin is cytotoxic to papillomavirus-expressing epithelial cells in vitro and in vivo. Can Res. 2005;65(23):10854–61. Disbrow GL, Baege AC, Kierpiec KA, Yuan H, Centeno JA, Thibodeaux CA, Hartmann D, Schlegel R. Dihydroartemisinin is cytotoxic to papillomavirus-expressing epithelial cells in vitro and in vivo. Can Res. 2005;65(23):10854–61.
279.
Zurück zum Zitat Burikhanov R, Hebbar N, Noothi SK, Shukla N, Sledziona J, Araujo N, Kudrimoti M, Wang QJ, Watt DS, Welch DR, Maranchie J. Chloroquine-inducible Par-4 secretion is essential for tumor cell apoptosis and inhibition of metastasis. Cell Rep. 2017;18(2):508–19.PubMedPubMedCentral Burikhanov R, Hebbar N, Noothi SK, Shukla N, Sledziona J, Araujo N, Kudrimoti M, Wang QJ, Watt DS, Welch DR, Maranchie J. Chloroquine-inducible Par-4 secretion is essential for tumor cell apoptosis and inhibition of metastasis. Cell Rep. 2017;18(2):508–19.PubMedPubMedCentral
280.
Zurück zum Zitat Chen D, Xie J, Fiskesund R, Dong W, Liang X, Lv J, Jin X, Liu J, Mo S, Zhang T, Cheng F. Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype. Nat Commun. 2018;9(1):873.PubMedPubMedCentral Chen D, Xie J, Fiskesund R, Dong W, Liang X, Lv J, Jin X, Liu J, Mo S, Zhang T, Cheng F. Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype. Nat Commun. 2018;9(1):873.PubMedPubMedCentral
281.
Zurück zum Zitat Boone BA, Murthy P, Miller-Ocuin J, Doerfler WR, Ellis JT, Liang X, Ross MA, Wallace CT, Sperry JL, Lotze MT, Neal MD. Chloroquine reduces hypercoagulability in pancreatic cancer through inhibition of neutrophil extracellular traps. BMC Cancer. 2018;18:1–2. Boone BA, Murthy P, Miller-Ocuin J, Doerfler WR, Ellis JT, Liang X, Ross MA, Wallace CT, Sperry JL, Lotze MT, Neal MD. Chloroquine reduces hypercoagulability in pancreatic cancer through inhibition of neutrophil extracellular traps. BMC Cancer. 2018;18:1–2.
282.
Zurück zum Zitat Valdés-Abadía B, Morán-Zendejas R, Rangel-Flores JM, Rodríguez-Menchaca AA. Chloroquine inhibits tumor-related Kv10.1 channel and decreases migration of MDA-MB-231 breast cancer cells in vitro. Eur J Pharmacol. 2019;855:262–6.PubMed Valdés-Abadía B, Morán-Zendejas R, Rangel-Flores JM, Rodríguez-Menchaca AA. Chloroquine inhibits tumor-related Kv10.1 channel and decreases migration of MDA-MB-231 breast cancer cells in vitro. Eur J Pharmacol. 2019;855:262–6.PubMed
283.
Zurück zum Zitat Shiratori H, Kawai K, Hata K, Tanaka T, Nishikawa T, Otani K, Sasaki K, Kaneko M, Murono K, Emoto S, Sonoda H. The combination of temsirolimus and chloroquine increases radiosensitivity in colorectal cancer cells. Oncol Rep. 2019;42(1):377–85.PubMed Shiratori H, Kawai K, Hata K, Tanaka T, Nishikawa T, Otani K, Sasaki K, Kaneko M, Murono K, Emoto S, Sonoda H. The combination of temsirolimus and chloroquine increases radiosensitivity in colorectal cancer cells. Oncol Rep. 2019;42(1):377–85.PubMed
284.
Zurück zum Zitat Hounjet J, Habets R, Schaaf MB, Hendrickx TC, Barbeau LM, Yahyanejad S, Rouschop KM, Groot AJ, Vooijs M. The anti-malarial drug chloroquine sensitizes oncogenic NOTCH1 driven human T-ALL to γ-secretase inhibition. Oncogene. 2019;38(27):5457–68.PubMed Hounjet J, Habets R, Schaaf MB, Hendrickx TC, Barbeau LM, Yahyanejad S, Rouschop KM, Groot AJ, Vooijs M. The anti-malarial drug chloroquine sensitizes oncogenic NOTCH1 driven human T-ALL to γ-secretase inhibition. Oncogene. 2019;38(27):5457–68.PubMed
285.
Zurück zum Zitat Choi DS, Blanco E, Kim YS, Rodriguez AA, Zhao H, Huang TH, Chen CL, Jin G, Landis MD, Burey LA, Qian W. Chloroquine eliminates cancer stem cells through deregulation of Jak2 and DNMT1. Stem Cells. 2014;32(9):2309–23.PubMed Choi DS, Blanco E, Kim YS, Rodriguez AA, Zhao H, Huang TH, Chen CL, Jin G, Landis MD, Burey LA, Qian W. Chloroquine eliminates cancer stem cells through deregulation of Jak2 and DNMT1. Stem Cells. 2014;32(9):2309–23.PubMed
286.
Zurück zum Zitat Cook KL, Wärri A, Soto-Pantoja DR, Clarke PA, Cruz MI, Zwart A, Clarke R. Chloroquine inhibits autophagy to potentiate antiestrogen responsiveness in ER+ breast cancer. Clin Cancer Res. 2014;20(12):3222–32.PubMedPubMedCentral Cook KL, Wärri A, Soto-Pantoja DR, Clarke PA, Cruz MI, Zwart A, Clarke R. Chloroquine inhibits autophagy to potentiate antiestrogen responsiveness in ER+ breast cancer. Clin Cancer Res. 2014;20(12):3222–32.PubMedPubMedCentral
287.
Zurück zum Zitat Rosenfeld MR, Ye X, Supko JG, Desideri S, Grossman SA, Brem S, Mikkelson T, Wang D, Chang YC, Hu J, McAfee Q. A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy. 2014;10(8):1359–68.PubMedPubMedCentral Rosenfeld MR, Ye X, Supko JG, Desideri S, Grossman SA, Brem S, Mikkelson T, Wang D, Chang YC, Hu J, McAfee Q. A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy. 2014;10(8):1359–68.PubMedPubMedCentral
288.
Zurück zum Zitat Liu LQ, Wang SB, Shao YF, Shi JN, Wang W, Chen WY, Ye ZQ, Jiang JY, Fang QX, Zhang GB, Xuan ZX. Hydroxychloroquine potentiates the anti-cancer effect of bevacizumab on glioblastoma via the inhibition of autophagy. Biomed Pharmacother. 2019;1(118):109339. Liu LQ, Wang SB, Shao YF, Shi JN, Wang W, Chen WY, Ye ZQ, Jiang JY, Fang QX, Zhang GB, Xuan ZX. Hydroxychloroquine potentiates the anti-cancer effect of bevacizumab on glioblastoma via the inhibition of autophagy. Biomed Pharmacother. 2019;1(118):109339.
289.
Zurück zum Zitat Wang W, Liu L, Zhou Y, Ye Q, Yang X, Jiang J, Ye Z, Gao F, Tan X, Zhang G, Fang Q. Hydroxychloroquine enhances the antitumor effects of BC001 in gastric cancer. Int J Oncol. 2019;55(2):405–14.PubMedPubMedCentral Wang W, Liu L, Zhou Y, Ye Q, Yang X, Jiang J, Ye Z, Gao F, Tan X, Zhang G, Fang Q. Hydroxychloroquine enhances the antitumor effects of BC001 in gastric cancer. Int J Oncol. 2019;55(2):405–14.PubMedPubMedCentral
290.
Zurück zum Zitat Li Y, Cao F, Li M, Li P, Yu Y, Xiang L, Xu T, Lei J, Tai YY, Zhu J, Yang B. Hydroxychloroquine induced lung cancer suppression by enhancing chemo-sensitization and promoting the transition of M2-TAMs to M1-like macrophages. J Exp Clin Cancer Res. 2018;37:1–6. Li Y, Cao F, Li M, Li P, Yu Y, Xiang L, Xu T, Lei J, Tai YY, Zhu J, Yang B. Hydroxychloroquine induced lung cancer suppression by enhancing chemo-sensitization and promoting the transition of M2-TAMs to M1-like macrophages. J Exp Clin Cancer Res. 2018;37:1–6.
291.
Zurück zum Zitat Van Dyke K, Lantz C, Szustkiewicz C. Quinacrine: mechanisms of antimalarial action. Science. 1970;169(3944):492–3.PubMed Van Dyke K, Lantz C, Szustkiewicz C. Quinacrine: mechanisms of antimalarial action. Science. 1970;169(3944):492–3.PubMed
292.
Zurück zum Zitat Requena-Méndez A, Goñi P, Rubio E, Pou D, Fumadó V, Lóbez S, Aldasoro E, Cabezos J, Valls ME, Treviño B, Martínez Montseny AF. The use of quinacrine in nitroimidazole-resistant Giardia duodenalis: an old drug for an emerging problem. J Infect Dis. 2017;215(6):946–53.PubMed Requena-Méndez A, Goñi P, Rubio E, Pou D, Fumadó V, Lóbez S, Aldasoro E, Cabezos J, Valls ME, Treviño B, Martínez Montseny AF. The use of quinacrine in nitroimidazole-resistant Giardia duodenalis: an old drug for an emerging problem. J Infect Dis. 2017;215(6):946–53.PubMed
293.
Zurück zum Zitat Egorin MJ, Trump DL, Wainwright CW. Quinacrine ochronosis and rheumatoid arthritis. JAMA. 1976;236(4):385–6.PubMed Egorin MJ, Trump DL, Wainwright CW. Quinacrine ochronosis and rheumatoid arthritis. JAMA. 1976;236(4):385–6.PubMed
294.
Zurück zum Zitat Larrieu AJ, Tyers GF, Williams EH, O’Neill MJ, Derrick JR. Intrapleural instillation of quinacrine for treatment of recurrent spontaneous pneumothorax. Ann Thorac Surg. 1979;28(2):146–50.PubMed Larrieu AJ, Tyers GF, Williams EH, O’Neill MJ, Derrick JR. Intrapleural instillation of quinacrine for treatment of recurrent spontaneous pneumothorax. Ann Thorac Surg. 1979;28(2):146–50.PubMed
295.
Zurück zum Zitat Geschwind MD, Kuo AL, Wong KS, Haman A, Devereux G, Raudabaugh BJ, Johnson DY, Torres-Chae CC, Finley R, Garcia P, Thai JN. Quinacrine treatment trial for sporadic Creutzfeldt-Jakob disease. Neurology. 2013;81(23):2015–23.PubMedPubMedCentral Geschwind MD, Kuo AL, Wong KS, Haman A, Devereux G, Raudabaugh BJ, Johnson DY, Torres-Chae CC, Finley R, Garcia P, Thai JN. Quinacrine treatment trial for sporadic Creutzfeldt-Jakob disease. Neurology. 2013;81(23):2015–23.PubMedPubMedCentral
296.
Zurück zum Zitat Collinge J, Gorham M, Hudson F, Kennedy A, Keogh G, Pal S, Rossor M, Rudge P, Siddique D, Spyer M, Thomas D. Safety and efficacy of quinacrine in human prion disease (PRION-1 study): a patient-preference trial. Lancet Neurol. 2009;8(4):334–44.PubMedPubMedCentral Collinge J, Gorham M, Hudson F, Kennedy A, Keogh G, Pal S, Rossor M, Rudge P, Siddique D, Spyer M, Thomas D. Safety and efficacy of quinacrine in human prion disease (PRION-1 study): a patient-preference trial. Lancet Neurol. 2009;8(4):334–44.PubMedPubMedCentral
297.
Zurück zum Zitat Oien DB, Pathoulas CL, Ray U, Thirusangu P, Kalogera E, Shridhar V. Repurposing quinacrine for treatment-refractory cancer. Semin Cancer Biol. 2021;68:21–30.PubMed Oien DB, Pathoulas CL, Ray U, Thirusangu P, Kalogera E, Shridhar V. Repurposing quinacrine for treatment-refractory cancer. Semin Cancer Biol. 2021;68:21–30.PubMed
298.
Zurück zum Zitat Gurova KV, Hill JE, Guo C, Prokvolit A, Burdelya LG, Samoylova E, Khodyakova AV, Ganapathi R, Ganapathi M, Tararova ND, Bosykh D. Small molecules that reactivate p53 in renal cell carcinoma reveal a NF-κB-dependent mechanism of p53 suppression in tumors. Proc Natl Acad Sci. 2005;102(48):17448–53.PubMedPubMedCentral Gurova KV, Hill JE, Guo C, Prokvolit A, Burdelya LG, Samoylova E, Khodyakova AV, Ganapathi R, Ganapathi M, Tararova ND, Bosykh D. Small molecules that reactivate p53 in renal cell carcinoma reveal a NF-κB-dependent mechanism of p53 suppression in tumors. Proc Natl Acad Sci. 2005;102(48):17448–53.PubMedPubMedCentral
299.
Zurück zum Zitat Nesher E, Safina A, Aljahdali I, Portwood S, Wang ES, Koman I, Wang J, Gurova KV. Role of chromatin damage and chromatin trapping of FACT in mediating the anticancer cytotoxicity of DNA-binding small-molecule drugs. Can Res. 2018;78(6):1431–43. Nesher E, Safina A, Aljahdali I, Portwood S, Wang ES, Koman I, Wang J, Gurova KV. Role of chromatin damage and chromatin trapping of FACT in mediating the anticancer cytotoxicity of DNA-binding small-molecule drugs. Can Res. 2018;78(6):1431–43.
300.
Zurück zum Zitat Gasparian AV, Burkhart CA, Purmal AA, Brodsky L, Pal M, Saranadasa M, Bosykh DA, Commane M, Guryanova OA, Pal S, Safina A. Curaxins: anticancer compounds that simultaneously suppress NF-κB and activate p53 by targeting FACT. Sci Transl Med. 2011;3(95):95ra74.PubMedPubMedCentral Gasparian AV, Burkhart CA, Purmal AA, Brodsky L, Pal M, Saranadasa M, Bosykh DA, Commane M, Guryanova OA, Pal S, Safina A. Curaxins: anticancer compounds that simultaneously suppress NF-κB and activate p53 by targeting FACT. Sci Transl Med. 2011;3(95):95ra74.PubMedPubMedCentral
301.
Zurück zum Zitat Park S, Oh AY, Cho JH, Yoon MH, Woo TG, Kang SM, Lee HY, Jung YJ, Park BJ. Therapeutic effect of quinacrine, an antiprotozoan drug, by selective suppression of p-CHK1/2 in p53-negative malignant cancers. Mol Cancer Res. 2018;16(6):935–46.PubMed Park S, Oh AY, Cho JH, Yoon MH, Woo TG, Kang SM, Lee HY, Jung YJ, Park BJ. Therapeutic effect of quinacrine, an antiprotozoan drug, by selective suppression of p-CHK1/2 in p53-negative malignant cancers. Mol Cancer Res. 2018;16(6):935–46.PubMed
302.
Zurück zum Zitat Preet R, Siddharth S, Satapathy SR, Das S, Nayak A, Das D, Wyatt MD, Kundu CN. Chk1 inhibitor synergizes quinacrine mediated apoptosis in breast cancer cells by compromising the base excision repair cascade. Biochem Pharmacol. 2016;1(105):23–33. Preet R, Siddharth S, Satapathy SR, Das S, Nayak A, Das D, Wyatt MD, Kundu CN. Chk1 inhibitor synergizes quinacrine mediated apoptosis in breast cancer cells by compromising the base excision repair cascade. Biochem Pharmacol. 2016;1(105):23–33.
303.
Zurück zum Zitat Mohapatra P, Preet R, Das D, Satapathy SR, Choudhuri T, Wyatt MD, Kundu CN. Quinacrine-mediated autophagy and apoptosis in colon cancer cells is through a p53-and p21-dependent mechanism. Oncol Res Featur Preclin Clin Cancer Ther. 2012;20(2–3):81–91. Mohapatra P, Preet R, Das D, Satapathy SR, Choudhuri T, Wyatt MD, Kundu CN. Quinacrine-mediated autophagy and apoptosis in colon cancer cells is through a p53-and p21-dependent mechanism. Oncol Res Featur Preclin Clin Cancer Ther. 2012;20(2–3):81–91.
304.
Zurück zum Zitat Gupta N, Srivastava SK. Atovaquone: an antiprotozoal drug suppresses primary and resistant breast tumor growth by inhibiting HER2/β-catenin signaling. Mol Cancer Ther. 2019;18(10):1708–20.PubMedPubMedCentral Gupta N, Srivastava SK. Atovaquone: an antiprotozoal drug suppresses primary and resistant breast tumor growth by inhibiting HER2/β-catenin signaling. Mol Cancer Ther. 2019;18(10):1708–20.PubMedPubMedCentral
305.
Zurück zum Zitat Gupta N, Gaikwad S, Kaushik I, Wright SE, Markiewski MM, Srivastava SK. Atovaquone suppresses triple-negative breast tumor growth by reducing immune-suppressive cells. Int J Mol Sci. 2021;22(10):5150.PubMedPubMedCentral Gupta N, Gaikwad S, Kaushik I, Wright SE, Markiewski MM, Srivastava SK. Atovaquone suppresses triple-negative breast tumor growth by reducing immune-suppressive cells. Int J Mol Sci. 2021;22(10):5150.PubMedPubMedCentral
306.
Zurück zum Zitat Fiorillo M, Lamb R, Tanowitz HB, Mutti L, Krstic-Demonacos M, Cappello AR, Martinez-Outschoorn UE, Sotgia F, Lisanti MP. Repurposing atovaquone: targeting mitochondrial complex III and OXPHOS to eradicate cancer stem cells. Oncotarget. 2016;7(23):34084.PubMedPubMedCentral Fiorillo M, Lamb R, Tanowitz HB, Mutti L, Krstic-Demonacos M, Cappello AR, Martinez-Outschoorn UE, Sotgia F, Lisanti MP. Repurposing atovaquone: targeting mitochondrial complex III and OXPHOS to eradicate cancer stem cells. Oncotarget. 2016;7(23):34084.PubMedPubMedCentral
307.
Zurück zum Zitat Tüzün F, Ünalan H, Öner N, Özgüzel H, Kirazli Y, İçağasioğlu A, Kuran B, Tüzün Ş, Başar G. Multicenter, randomized, double-blinded, placebo-controlled trial of thiocolchicoside in acute low back pain. Joint Bone Spine. 2003;70(5):356–61.PubMed Tüzün F, Ünalan H, Öner N, Özgüzel H, Kirazli Y, İçağasioğlu A, Kuran B, Tüzün Ş, Başar G. Multicenter, randomized, double-blinded, placebo-controlled trial of thiocolchicoside in acute low back pain. Joint Bone Spine. 2003;70(5):356–61.PubMed
308.
Zurück zum Zitat Reuter S, Prasad S, Phromnoi K, Ravindran J, Sung B, Yadav VR, Kannappan R, Chaturvedi MM, Aggarwal BB. Thiocolchicoside exhibits anticancer effects through downregulation of NF-κB pathway and its regulated gene products linked to inflammation and cancer. Cancer Prev Res. 2010;3(11):1462–72. Reuter S, Prasad S, Phromnoi K, Ravindran J, Sung B, Yadav VR, Kannappan R, Chaturvedi MM, Aggarwal BB. Thiocolchicoside exhibits anticancer effects through downregulation of NF-κB pathway and its regulated gene products linked to inflammation and cancer. Cancer Prev Res. 2010;3(11):1462–72.
309.
Zurück zum Zitat Reuter S, Gupta SC, Phromnoi K, Aggarwal BB. Thiocolchicoside suppresses osteoclastogenesis induced by RANKL and cancer cells through inhibition of inflammatory pathways: a new use for an old drug. Br J Pharmacol. 2012;165(7):2127–39.PubMedPubMedCentral Reuter S, Gupta SC, Phromnoi K, Aggarwal BB. Thiocolchicoside suppresses osteoclastogenesis induced by RANKL and cancer cells through inhibition of inflammatory pathways: a new use for an old drug. Br J Pharmacol. 2012;165(7):2127–39.PubMedPubMedCentral
310.
Zurück zum Zitat Micheau O, Dufour F, Walczak H. Thiocolchicoside a semi-synthetic derivative of the Glory Lily: a new weapon to fight metastatic bone resorption. Br J Pharmacol. 2012;165(7):2124–6.PubMedPubMedCentral Micheau O, Dufour F, Walczak H. Thiocolchicoside a semi-synthetic derivative of the Glory Lily: a new weapon to fight metastatic bone resorption. Br J Pharmacol. 2012;165(7):2124–6.PubMedPubMedCentral
311.
Zurück zum Zitat Sproviero E, Albamonte E, Costantino C, Giossi A, Mancuso M, Rigamonti A, Tornari P, Caggiano G. Efficacy and safety of a fixed combination of intramuscular diclofenac 75 mg+ thiocolchicoside 4 mg in the treatment of acute low back pain: a phase III, randomized, double blind, controlled trial. Eur J Phys Rehabil Med. 2018;54(5):654–62.PubMed Sproviero E, Albamonte E, Costantino C, Giossi A, Mancuso M, Rigamonti A, Tornari P, Caggiano G. Efficacy and safety of a fixed combination of intramuscular diclofenac 75 mg+ thiocolchicoside 4 mg in the treatment of acute low back pain: a phase III, randomized, double blind, controlled trial. Eur J Phys Rehabil Med. 2018;54(5):654–62.PubMed
312.
Zurück zum Zitat Rao R, Panghate A, Chandanwale A, Sardar I, Ghosh M, Roy M, Banerjee B, Goswami A, Kotwal pp. Clinical comparative study: efficacy and tolerability of tolperisone and thiocolchicoside in acute low back pain and spinal muscle spasticity. Asian Spine J. 2012;6(2):115.PubMedPubMedCentral Rao R, Panghate A, Chandanwale A, Sardar I, Ghosh M, Roy M, Banerjee B, Goswami A, Kotwal pp. Clinical comparative study: efficacy and tolerability of tolperisone and thiocolchicoside in acute low back pain and spinal muscle spasticity. Asian Spine J. 2012;6(2):115.PubMedPubMedCentral
313.
Zurück zum Zitat Wang T, Fu X, Jin T, Zhang L, Liu B, Wu Y, Xu F, Wang X, Ye K, Zhang W, Ye L. Aspirin targets P4HA2 through inhibiting NF-κB and LMCD1-AS1/let-7g to inhibit tumour growth and collagen deposition in hepatocellular carcinoma. EBioMedicine. 2019;1(45):168–80. Wang T, Fu X, Jin T, Zhang L, Liu B, Wu Y, Xu F, Wang X, Ye K, Zhang W, Ye L. Aspirin targets P4HA2 through inhibiting NF-κB and LMCD1-AS1/let-7g to inhibit tumour growth and collagen deposition in hepatocellular carcinoma. EBioMedicine. 2019;1(45):168–80.
314.
Zurück zum Zitat Fujiwara N, Singal AG, Hoshida Y. Dose and duration of aspirin use to reduce incidental hepatocellular carcinoma. Hepatology. 2019;70(6):2216.PubMed Fujiwara N, Singal AG, Hoshida Y. Dose and duration of aspirin use to reduce incidental hepatocellular carcinoma. Hepatology. 2019;70(6):2216.PubMed
315.
Zurück zum Zitat Yao Y, Guo Q, Cao Y, Qiu Y, Tan R, Yu Z, Zhou Y, Lu N. Artemisinin derivatives inactivate cancer-associated fibroblasts through suppressing TGF-β signaling in breast cancer. J Exp Clin Cancer Res. 2018;37(1):1–4. Yao Y, Guo Q, Cao Y, Qiu Y, Tan R, Yu Z, Zhou Y, Lu N. Artemisinin derivatives inactivate cancer-associated fibroblasts through suppressing TGF-β signaling in breast cancer. J Exp Clin Cancer Res. 2018;37(1):1–4.
316.
Zurück zum Zitat Li X, Ba Q, Liu Y, Yue Q, Chen P, Li J, Zhang H, Ying H, Ding Q, Song H, Liu H. Dihydroartemisinin selectively inhibits PDGFRα-positive ovarian cancer growth and metastasis through inducing degradation of PDGFRα protein. Cell Discov. 2017;3(1):1–3. Li X, Ba Q, Liu Y, Yue Q, Chen P, Li J, Zhang H, Ying H, Ding Q, Song H, Liu H. Dihydroartemisinin selectively inhibits PDGFRα-positive ovarian cancer growth and metastasis through inducing degradation of PDGFRα protein. Cell Discov. 2017;3(1):1–3.
Metadaten
Titel
Repurposing approved non-oncology drugs for cancer therapy: a comprehensive review of mechanisms, efficacy, and clinical prospects
verfasst von
Roohi Mohi-ud-din
Apporva Chawla
Pooja Sharma
Prince Ahad Mir
Faheem Hyder Potoo
Željko Reiner
Ivan Reiner
Dilek Arslan Ateşşahin
Javad Sharifi-Rad
Reyaz Hassan Mir
Daniela Calina
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
European Journal of Medical Research / Ausgabe 1/2023
Elektronische ISSN: 2047-783X
DOI
https://doi.org/10.1186/s40001-023-01275-4

Weitere Artikel der Ausgabe 1/2023

European Journal of Medical Research 1/2023 Zur Ausgabe