Elsevier

Drug Resistance Updates

Volume 26, May 2016, Pages 10-27
Drug Resistance Updates

Repositioning of drugs for intervention in tumor progression and metastasis: Old drugs for new targets

https://doi.org/10.1016/j.drup.2016.03.002Get rights and content

Abstract

The increasing unraveling of the molecular basis of cancer offers manifold novel options for intervention strategies. However, the discovery and development of new drugs for potential clinical applications is a tremendously time-consuming and costly process. Translating a novel lead candidate compound into an approved clinical drug takes often more than a decade, and the success rate is very low due to versatile efforts including defining its pharmacokinetics, pharmacodynamics, side effects as well as lack of sufficient efficacy. Thus, strategies are needed to minimize time and costs, while maximizing success rates. A very attractive strategy for novel cancer therapeutic options is the repositioning of already approved drugs. These medicines, approved for the treatment of non-malignant disorders, have already passed some early costs and time, have been tested in humans and are ready for clinical trials as anti-cancer drugs. Here we discuss the repositioning of nonsteroidal anti-inflammatory drugs (NSAID), statins, anti-psychotic drugs, anti-helminthic drugs and vitamin D as anti-tumor agents. We focus on their novel actions and potential for inhibition of cancer growth and metastasis by interfering with target molecules and pathways, which drive these malignant processes. Furthermore, important pre-clinical and clinical data are reviewed herein, which elucidate their therapeutic mechanisms which enable their repositioning for cancer therapy and disruption of metastasis.

Introduction

For decades a plethora of drugs have been developed for the treatment of different malignant and non-malignant disorders. In this effort, natural or synthetic compounds were isolated or generated and extensively tested to finally use them for the treatment of a particular disease in the clinic. The process of drug discovery usually starts with the identification of a compound, which has the potential to become an active drug. Such a compound then can enter the very time-consuming and costly process to achieve a validation of the preclinical therapeutic efficacy of a given compound and to perform pharmacokinetics, pharmacodynamics, and toxicity studies before clinical application is anticipated (Fig. 1). Due to the immensely high costs of pre-clinical as well as clinical evaluation over many years, introduction of new drugs for a particular disease including cancer has become a challenging endeavor.

Analyses regarding scientific, clinical, and financial efforts and the respective establishment of a novel drug for routine clinical application revealed a significant decrease in the number of drugs introduced to the clinic (Mullard, 2014, Pammolli et al., 2011, Scannell et al., 2012). The number of novel drugs has halved every nine years during the past 40 years of drug development per billion US dollars invested into research and development. The average time for the development of a novel drug has also increased considerably over time. During the 1990s the average time required from drug discovery to its use in the clinic (market launch) was 9.7 years. This has then increased to almost 14 years from 2000 onwards – and still increases (Pammolli et al., 2011). In addition to this dilemma, the Tufts Center for Study of Drug Development stated that costs for market launch of a new drug in 2014 has increased 1.7-fold compared to the costs required in 2003. These facts reflect the inefficiency of the current drug development process in the developed industrial countries as well as the need of repositioning of already approved drugs.

The general hurdles in drug development also apply to the burning need to develop novel anti-cancer drugs. Here the estimated time required for drug discovery until the drug is approved is ∼8 years, in association with a very low success rate of only 6.7% in phase I clinical trials (Kaitin and DiMasi, 2011, Pantziarka et al., 2014). This is in spite of the tremendous progress made for the identification of novel molecular targets for cancer treatment and the molecular deciphering of signaling pathways, which are essential for tumor development, metastasis, immune surveillance of tumors, drug response or drug resistance mechanisms (Fisher et al., 2013, Gonen and Assaraf, 2012, Huang et al., 2014, Livney and Assaraf, 2013, Niewerth et al., 2015, Zhitomirsky and Assaraf, 2016).

The revolution in computational bioanalyses does further contribute to the acceleration in this field due to improved data mining and correlation analyses, such as correlation of drug response and biomarker expression or of mutations of particular genes. One would expect that with such progress in understanding the molecular basis of cancer biology, drug development of novel anti-cancer agents should be easier, faster and will also promote the entry of novel drugs into clinical application. However, this is hindered by the immense time and financial investment required for introduction of novel drugs into the clinic. In fact, the expected and already observed increase in cancer incidence in association with aging populations in developed countries clearly calls for the development of novel, more targeted, effective, and affordable drugs in the coming years (Vineis and Wild, 2014). This will be essential to better fulfill the modern needs for a more individualized cancer therapy.

Taking into account the various hurdles in the development of novel anti-cancer therapeutics, which aims at new druggable targets but also at already known ones, the use of known compounds does represent an attractive alternative. To achieve this goal, drug repositioning or repurposing came into focus. This approach is based on identification of new indications from already existing approved drugs. More importantly these drugs could be used for the treatment of diseases other than the original indication of the drug. This approach is certainly of particular interest in the search for new drugs for cancer treatment. To achieve this goal more efficiently, the Repurposing Drugs in Oncology (ReDO) project was initiated, to identify candidate drugs with the best potential of clinical use and to bring such drugs to better attention of the basic research and clinical community (Pantziarka et al., 2015, Pantziarka et al., 2014). The ReDO project has defined key criteria, by which such drugs can be efficiently selected as high-potential candidates for further re-evaluation:

  • 1.

    The compound should be well known for many years and widely used in the clinic. Generics of the drug are of advantage.

  • 2.

    The drug should have low toxicity (good toxicology profile), even upon long-term administration.

  • 3.

    A putative mechanism or defined target of action should be known for its use in cancer therapy.

  • 4.

    High-level evidence of anti-cancer activity in vitro as well as in vivo should be shown. In this respect, data generated in syngeneic, orthotopic transplantable models or in genetically engineered mice is of highest rating.

  • 5.

    Anti-cancer activity should be demonstrated at standard dosing at low toxicity.

  • 6.

    The drug of interest with anti-cancer activity should currently not be widely pursued as an active compound in cancer therapy.

Such drugs can be identified via high-throughput screens using small molecule libraries, which incorporate approved drugs of different clinical uses; other than cancer therapy. This is recently supported by various open-access libraries of approved and investigational compounds enabling better drug repositioning, as provided by the National Clinical Guidance Center (NCGC), the Pharmaceutical Collection (NPC) for virtual screening, or the Johns Hopkins Drug Library (JHDL) as another assembly of existing drugs (Huang et al., 2011, Shim and Liu, 2014). Furthermore, knowledge of the molecular structure of target proteins could also be indicative (in silico analysis) that particular compounds might interfere with important functional domains of such target proteins to specifically inhibit their activities. This approach is currently under intense evaluation, called “drug-protein interaction-based repurposing (DPIR)”, analyzing drug-protein interactions for in silico prediction of promising drug candidates (Liu et al., 2014).

The first six compounds the ReDO project has selected for further evaluation are mebendazole (anti-helminthic), nitroglycerin (vasodilator), cimetidine (H2-receptor antagonist), clarithromycin (antibiotic), diclofenac (nonsteroidal anti-inflammatory drug, NSAID), and itraconazole (anti-fungal), drugs which were not in use for cancer therapy (Pantziarka et al., 2014).

As discussed in the context of drug repositioning, the key questions remain, at which point clinical testing should start for such drugs, if pre-clinical results clearly demonstrate anti-cancer activity? Should the entry level be at clinical phase II or even phase II/III at reduced size of the respective patient numbers required for the respective patient groups in the trial? To maintain the initial advantage of repositioning of drugs during the phases for clinical testing, the enhanced and widely accepted clinical testing is essential.

In this review not only the search for drug repositioning to treat cancer in general is in focus, but also the immense potential of this approach for identification of drugs, which can effectively prevent or inhibit metastasis. This is of particular interest, since for many malignancies patients die of their cancer metastases rather due to the primary tumor. Therefore, drugs which are able to interfere with tumor progression and also with metastasis-associated signaling pathways, are attractive candidates for intense testing. Such drugs could then be used as inhibitors of tumor progression and metastasis, or even as tumor and metastasis preventing drugs in a long-term treatment of defined patient groups.

For such drugs, similar criteria for their selection will apply as defined by the ReDO project:

  • 1.

    The compound should be well known and widely used in the clinic, with a good toxicology profile, particularly for the long-term medication in metastasis prevention settings.

  • 2.

    A putative mechanism or defined target of tumor and metastasis inhibition/prevention should be known.

  • 3.

    High-level in vitro evidence of anti-tumor and anti-metastatic activity as well as in vivo validation should be demonstrated at standard dosing.

    Furthermore:

  • 4.

    The proposed drug candidate might deliberately have multi-target promiscuous properties (polypharmacology), to hit at multiple sites of tumor progression and metastatic signaling pathways.

In the following chapter, different classes of clinically used drugs are introduced and discussed for their application in anti-tumor and anti-metastatic treatments, including their mechanisms of action.

Section snippets

NSAIDs in cancer therapy

NSAIDs do represent the most widely used group of compounds for many decades. These drugs interfere with the prostanoid synthesis (Vane, 1971). They represent an alternative to the steroidal anti-inflammatory drugs (cortisone, hydrocortisone, dexamethasone), which are related to the glucocorticoids, acting at the level of arachidonic acid release to prevent eicosanoid (prostanoids and leukotrienes) synthesis. In contrast, the NSAIDs target, e.g. the cyclooxygenase (COX) subunit of the

Conclusions

Repositioning of established “old” drugs for new targets bears great promise in cancer therapy. Targeting certain proteins and disrupting molecular cascades driving tumor progression and metastasis by exploiting already approved drugs will certainly enhance cancer treatment options. In the era of personalized medicine, the molecular classification of patient sub-cohorts, the identification of high-risk patients, and patient stratification according to molecular biomarkers, the repositioning of

Acknowledgements

This work was supported by the German Cancer Consortium (DKTK) (to GM and US). CT is a participant in the Charité Clinical Scientist Program funded by the Charité Universitätsmedizin Berlin and the Berlin Institute of Health.

References (225)

  • P.A. Friedman et al.

    Interaction of anthelmintic benzimidazoles with Ascaris suum embryonic tubulin

    Biochim. Biophys. Acta

    (1980)
  • C. Ginestier et al.

    ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome

    Cell Stem Cell

    (2007)
  • F. Goeman et al.

    VDR primary targets by genome-wide transcriptional profiling

    J. Steroid Biochem. Mol. Biol.

    (2014)
  • N. Gonen et al.

    Antifolates in cancer therapy: structure, activity and mechanisms of drug resistance

    Drug Resist. Updat.

    (2012)
  • D.W. Greening et al.

    Sulindac modulates secreted protein expression from LIM1215 colon carcinoma cells prior to apoptosis

    Biochim. Biophys. Acta

    (2013)
  • D.A. Guertin et al.

    Defining the role of mTOR in cancer

    Cancer Cell

    (2007)
  • A. Han et al.

    Sulindac suppresses beta-catenin expression in human cancer cells

    Eur. J. Pharmacol.

    (2008)
  • R. Hanif et al.

    Effects of nonsteroidal anti-inflammatory drugs on proliferation and on induction of apoptosis in colon cancer cells by a prostaglandin-independent pathway

    Biochem. Pharmacol.

    (1996)
  • M. Huang et al.

    Molecularly targeted cancer therapy: some lessons from the past decade

    Trends Pharmacol. Sci.

    (2014)
  • A. Janssen et al.

    Evidence of COX-2 independent induction of apoptosis and cell cycle block in human colon carcinoma cells after S- or R-ibuprofen treatment

    Eur. J. Pharmacol.

    (2006)
  • A. Janssen et al.

    p53 is important for the anti-proliferative effect of ibuprofen in colon carcinoma cells

    Biochem. Biophys. Res. Commun.

    (2008)
  • S. Kapur et al.

    Half a century of antipsychotics and still a central role for dopamine D2 receptors

    Prog. Neuropsychopharmacol. Biol. Psychiatry

    (2003)
  • K. Kashfi et al.

    NOSH-sulindac (AVT-18A) is a novel nitric oxide- and hydrogen sulfide-releasing hybrid that is gastrointestinal safe and has potent anti-inflammatory, analgesic, antipyretic, anti-platelet, and anti-cancer properties

    Redox Biol.

    (2015)
  • K. Kashfi et al.

    Non-COX-2 targets and cancer: expanding the molecular target repertoire of chemoprevention

    Biochem. Pharmacol.

    (2005)
  • D.B. Kastrinsky et al.

    Reengineered tricyclic anti-cancer agents

    Bioorg. Med. Chem.

    (2015)
  • M.S. Katz et al.

    Association of statin use with a pathologic complete response to neoadjuvant chemoradiation for rectal cancer

    Int. J. Radiat. Oncol. Biol. Phys.

    (2005)
  • J.J. Knox et al.

    A Phase I trial of prolonged administration of lovastatin in patients with recurrent or metastatic squamous cell carcinoma of the head and neck or of the cervix

    Eur. J. Cancer

    (2005)
  • I.R.H.M. Konings et al.

    The addition of pravastatin to chemotherapy in advanced gastric carcinoma: a randomised phase II trial

    Eur. J. Cancer

    (2010)
  • J.K. Kundu et al.

    beta-Catenin-mediated signaling: a novel molecular target for chemoprevention with anti-inflammatory substances

    Biochim. Biophys. Acta

    (2006)
  • J.P. Laclette et al.

    Inhibition of tubulin polymerization by mebendazole

    Biochem. Biophys. Res. Commun.

    (1980)
  • H. Li et al.

    Aspirin prevents colorectal cancer by normalizing EGFR expression

    EBioMedicine

    (2015)
  • O. Aguilera et al.

    The Wnt antagonist DICKKOPF-1 gene is induced by 1alpha, 25-dihydroxyvitamin D3 associated to the differentiation of human colon cancer cells

    Carcinogenesis

    (2007)
  • M. Al-Hajj et al.

    Prospective identification of tumorigenic breast cancer cells

    Proc. Natl. Acad. Sci. U. S. A.

    (2003)
  • R.-Y. Bai et al.

    Antiparasitic mebendazole shows survival benefit in 2 preclinical models of glioblastoma multiforme

    Neuro Oncol.

    (2011)
  • R.-Y. Bai et al.

    Effective treatment of diverse medulloblastoma models with mebendazole and its impact on tumor angiogenesis

    Neuro Oncol.

    (2015)
  • A.D. Balgi et al.

    Screen for chemical modulators of autophagy reveals novel therapeutic inhibitors of mTORC1 signaling

    PLoS ONE

    (2009)
  • M. Ben-Shoshan et al.

    1alpha, 25-dihydroxyvitamin D3 (Calcitriol) inhibits hypoxia-inducible factor-1/vascular endothelial growth factor pathway in human cancer cells

    Mol. Cancer Ther.

    (2007)
  • G. Bjelakovic et al.

    Vitamin D supplementation for prevention of cancer in adults

    Cochrane Database Syst. Rev.

    (2014)
  • C.D. Blanke et al.

    Phase II study of calcitriol-enhanced docetaxel in patients with previously untreated metastatic or locally advanced pancreatic cancer

    Invest. New Drugs

    (2009)
  • L. Bongiorno-Borbone et al.

    Anti-tumoral effect of desmethylclomipramine in lung cancer stem cells

    Oncotarget

    (2015)
  • E.M.J. Boon et al.

    Sulindac targets nuclear beta-catenin accumulation and Wnt signalling in adenomas of patients with familial adenomatous polyposis and in human colorectal cancer cell lines

    Br. J. Cancer

    (2004)
  • P. Cafforio et al.

    Statins activate the mitochondrial pathway of apoptosis in human lymphoblasts and myeloma cells

    Carcinogenesis

    (2005)
  • C.R. Cardwell et al.

    Statin use after colorectal cancer diagnosis and survival: a population-based cohort study

    J. Clin. Oncol.

    (2014)
  • L. Ceballos et al.

    Comparative performances of flubendazole and albendazole in cystic echinococcosis: ex vivo activity, plasma/cyst disposition, and efficacy in infected mice

    Antimicrob. Agents Chemother.

    (2011)
  • M. Chen et al.

    The anti-helminthic niclosamide inhibits Wnt/Frizzled1 signaling

    Biochemistry

    (2009)
  • J.R. Emberson et al.

    Lack of effect of lowering LDL cholesterol on cancer: meta-analysis of individual data from 175,000 people in 27 randomised trials of statin therapy

    PLoS ONE

    (2012)
  • H. Clevers

    Colon cancer – understanding how NSAIDs work

    N. Engl. J. Med.

    (2006)
  • N.R. Cook et al.

    Low-dose aspirin in the primary prevention of cancer: the Women's Health Study: a randomized controlled trial

    JAMA

    (2005)
  • C.P. Coyne et al.

    Anti-neoplastic cytotoxicity of gemcitabine-(C4-amide)-[anti-EGFR] in dual-combination with epirubicin-(C3-amide)-[anti-HER2/neu] against chemotherapeutic-resistant mammary adenocarcinoma (SKBr-3) and the complementary effect of mebendazole

    J. Cancer Res. Ther. Oncol.

    (2014)
  • S. Dakshanamurthy et al.

    Predicting new indications for approved drugs using a proteochemometric method

    J. Med. Chem.

    (2012)
  • Cited by (0)

    View full text