Review
Chloroquine and its analogs: A new promise of an old drug for effective and safe cancer therapies

https://doi.org/10.1016/j.ejphar.2009.06.063Get rights and content

Abstract

Chloroquine (CQ), N′-(7-chloroquinolin-4-yl)-N,N-diethyl-pentane-1,4-diamine, is widely used as an effective and safe anti-malarial and anti-rheumatoid agent. CQ was discovered 1934 as “Resochin” by Andersag and co-workers at the Bayer laboratories. Ironically, CQ was initially ignored for a decade because it was considered too toxic to use in humans. CQ was “re-discovered” during World War II in the United States in the course of anti-malarial drug development. The US government-sponsored clinical trials during this period showed unequivocally that CQ has a significant therapeutic value as an anti-malarial drug. Consequently, CQ was introduced into clinical practice in 1947 for the prophylaxis treatment of malaria (Plasmodium vivax, ovale and malariae). CQ still remains the drug of choice for malaria chemotherapy because it is highly effective and well tolerated by humans. In addition, CQ is widely used as an anti-inflammatory agent for the treatment of rheumatoid arthritis, lupus erythematosus and amoebic hepatitis. More recently, CQ has been studied for its potential as an enhancing agent in cancer therapies. Accumulating lines of evidence now suggest that CQ can effectively sensitize cell-killing effects by ionizing radiation and chemotherapeutic agents in a cancer-specific manner. The lysosomotrophic property of CQ appears to be important for the increase in efficacy and specificity. Although more studies are needed, CQ may be one of the most effective and safe sensitizers for cancer therapies. Taken together, it appears that the efficacy of conventional cancer therapies can be dramatically enhanced if used in combination with CQ and its analogs.

Introduction

If we want to further improve cancer cure rates, we need to understand the detailed control mechanism how anti-cancer therapeutics function in the context of cellular and molecular control pathways in normal and cancer cells. Furthermore, the limitation of drug concentrations used for therapy is a serious problem, mainly due to side effects. Thus, there is an urgent need for developing more effective cancer modalities with minimal side effects. Since cancer is caused by a complex chain of events, combinational modalities may provide a better response to cancer therapy (Cheng et al., 2008, Rosales-Hernandez et al., 2009).

A study of 68 newly approved drugs estimated that developing a single effective cancer drug takes an average of 15 years and US$800 million (DiMasi et al., 2003). One approach to overcome this enormous problem may be developing a new use of existing drugs. The renowned pharmacologist and Nobel laureate James Black said, “the most fruitful basis for the discovery of a new drug is to start with an old drug” (Chong and Sullivan, 2007). Since many existing drugs have been studied for their pharmacokinetics and safety profiles, and often have already been approved by regulatory agencies for human use, any newly identified use of them can be rapidly evaluated in phase II trials (DiMasi et al., 2003). Developing a known drug for another clinical purpose is termed “repurposing”. The efficacy and specificity of a known drug can also be improved by modifications of its chemical side chains and functional groups, which is termed “repositioning”.

Chloroquine is a well-known 4-aminoquinoline class of drug that is widely used for the prophylaxis treatment of malaria (Wiesner et al., 2003). Even after six decades of use, CQ still remains the drug of choice for malaria treatment because it is effective, low toxic to humans, and inexpensive (Breckenridge and Winstanley, 1997). Biochemical data suggest that this class of compounds enter acidic vacuoles of host cells, where they inhibit the growth of parasites by forming a complex with haematin (Dorn et al., 1995, Pandey et al., 2001, Sullivan et al., 1996). The anti-inflammatory property of CQ also makes it a useful agent for the treatment of rheumatoid arthritis (Augustijns et al., 1992, Titus, 1989), lupus erythematosus (Meinao et al., 1996) and amoebic hepatitis (Conan, 1948). Furthermore, CQ inhibits pro-inflammatory cytokine release into the blood stream, suggesting that it may have therapeutic benefits for chronic inflammation diseases as well as for relieving symptoms caused by bacteria-induced inflammation (Karres et al., 1998). Chloroquine is currently in clinical trials as an investigational anti-retroviral agent in humans with HIV-1/AIDS (Savarino et al., 2003, Savarino et al., 2006) and as a potential anti-viral agent against chikungunya fever (Savarino et al., 2007). In addition, CQ has been studied for its potential in the enhancement of radiation therapy (Beierwaltes et al., 1968, Zhao et al., 2005), chemotherapy, and combinational therapy for cancer ( Carew et al., 2006, Degtyarev et al., 2008, Hagihara et al., 2000, Hu et al., 2008). Our current review focuses on CQ and its analogs as enhancement agents for cancer therapies.

Section snippets

Discovery of CQ from quinine

In the 18th century, the first attempt of successful treatment of malaria was made by utilizing the bark of cinchona trees, which had been used for the treatment of fever since the beginning of the 17th century (Meshnick and Dobson, 2001). Subsequently, Pelletier and Caventou isolated the cinchona alkaloid active ingredient quinine (Fig. 1) and cinchonine from the crude extracts of cinchona bark (Pelletier and Caventou, 1820). Quinine was then widely used as an anti-malarial agent, replacing

The property of CQ

Chloroquine is usually prepared as a diphosphate salt of N'-(7-chloroquinolin-4-yl)-N,N-diethyl-pentane-1,4-diamine, which is a diprotic weak base (pka1 = 8.1, pka2 = 10.2) that can exist in both charged (i.e., protonated) and uncharged (i.e., unprotonated) forms. An unprotonated CQ can diffuse freely and rapidly across the membranes of the cell and organelles. Once protonated, CQ may be “trapped” in the organelles such as the lysosomes since it can no longer freely diffuse out. This may be

Combined therapies with CQ and other cancer therapeutics

Combination of one or more therapeutic agents has been used to improve the outcome of cancer therapies (Chau and Cunningham, 2006, Faivre et al., 2006, Hennig et al., 2004, Hosoya et al., 1999). Although some of the combination therapies have been shown to improve overall patient survival, such treatments are often associated with an increase in toxicity and the potential for developing cross-resistance (Ocana et al., 2006, Thomsen and Kolesar, 2008). Thus, there is an urgent need for

Miscellaneous

Accumulating lines of evidence now suggest that the development of weak DNA-intercalating bioreductive compounds is an effective strategy to ensure DNA affinity high enough to produce toxicity yet low enough to permit efficient extravascular diffusion and penetration to hypoxic tumor tissue (Brown, 2000). Based on this strategy, Papadopoulou et al. (2000) synthesized a CQ derivative, 4-[3-(2-nitro-1-imidazolyl)-propylamino]-7-chloroquinoline hydrochloride (NIPCQ, NSC709257, Fig. 6). The

Future directions

The lysosomotropic CQ is accumulated in the lysosomes, raises intra-lysosomal pH, and interferes with autophagosome degradation in the lysosomes. This unique property of CQ may be important for the enhancement of cell killing by cancer therapeutic agents in a variety of different tumors and genetic backgrounds (Amaravadi et al., 2007, Carew et al., 2006, Maclean et al., 2008, Zhao et al., 2005). Together, published data suggest that combined modalities of CQ with other therapeutics are very

Acknowledgements

The authors are grateful to Jane Vanderklift for her help in the editorial preparation of this manuscript. This work was supported by funds from the Canadian Breast Cancer Foundation (CBCF, Ontario Chapter), the Ontario Institute of Cancer Research/Cancer Care Ontario, and the Northern Cancer Research Foundation to H.L. V.R.S. thanks the Ontario Ministry of Research & Innovation for Postdoctoral Fellowship.

References (176)

  • S. Faivre et al.

    New paradigms in anticancer therapy: targeting multiple signaling pathways with kinase inhibitors

    Semin. Oncol.

    (2006)
  • C. Fan et al.

    Chloroquine inhibits cell growth and induces cell death in A549 lung cancer cells

    Bioorg. Med. Chem.

    (2006)
  • H. Glaumann et al.

    Comparison of different autophagic vacuoles with regard to ultrastructure, enzymatic composition, and degradation capacity–formation of crinosomes

    Exp. Mol. Pathol.

    (1987)
  • T.L. Goldsworthy et al.

    Apoptosis and cancer risk assessment

    Mutat. Res.

    (1996)
  • R. Hennig et al.

    Effect of LY293111 in combination with gemcitabine in colonic cancer

    Cancer Lett.

    (2004)
  • Y. Hosoya et al.

    Combination effects of tamoxifen plus 5-fluorouracil on gastric cancer cell lines in vitro

    Cancer Lett.

    (1999)
  • K.Y. Hostetler et al.

    Chloroquine-induced phospholipid fatty liver. Measurement of drug and lipid concentrations in rat liver lysosomes

    J. Biol. Chem.

    (1985)
  • C. Hu et al.

    The efficacy and selectivity of tumor cell killing by Akt inhibitors are substantially increased by chloroquine

    Bioorg. Med. Chem.

    (2008)
  • S.J. Hurwitz et al.

    Vesicular anthracycline accumulation in doxorubicin-selected U-937 cells: participation of lysosomes

    Blood

    (1997)
  • V.G. Johnson et al.

    The role of the diphtheria toxin receptor in cytosol translocation

    J. Biol. Chem.

    (1988)
  • H. Kunze et al.

    Effects of antimalarial drugs on several rat-liver lysosomal enzymes involved in phosphatidylethanolamine catabolism

    Biochim. Biophys. Acta

    (1982)
  • P.Y. Law et al.

    Down-regulation of opiate receptor in neuroblastoma x glioma NG108–15 hybrid cells. Chloroquine promotes accumulation of tritiated enkephalin in the lysosomes

    J. Biol. Chem.

    (1984)
  • S. Leppla et al.

    Inhibition of diphtheria toxin degradation and cytotoxic action by chloroquine

    J. Biol. Chem.

    (1980)
  • R.A. Lockshin et al.

    Apoptosis, autophagy, and more

    Int. J. Biochem. Cell Biol.

    (2004)
  • R.Z. Luo et al.

    Genomic structure and promoter characterization of an imprinted tumor suppressor gene ARHI

    Biochim. Biophys. Acta

    (2001)
  • L.D. Mayer et al.

    Uptake of adriamycin into large unilamellar vesicles in response to a pH gradient

    Biochim. Biophys. Acta

    (1986)
  • C.M. McNeil et al.

    c-Myc overexpression and endocrine resistance in breast cancer

    J. Steroid Biochem. Mol. Biol.

    (2006)
  • R.O. Michael et al.

    Chloroquine inhibition of repair of DNA damage induced in mammalian cells by methyl methanesulfonate

    Mutat. Res.

    (1974)
  • R. Nosal et al.

    Cationic amphiphilic drugs and platelet phospholipase A(2) (cPLA(2))

    Thromb. Res.

    (2002)
  • R.L. O'Brien et al.

    Evidence for intercalation of chloroquine into DNA

    Biochim. Biophys. Acta

    (1966)
  • J.S. Abrams et al.

    New chemotherapeutic agents for breast cancer

    Cancer

    (1994)
  • A. Aderem et al.

    Mechanisms of phagocytosis in macrophages

    Annu. Rev. Immunol.

    (1999)
  • R.K. Amaravadi et al.

    Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma

    J. Clin. Invest.

    (2007)
  • H. Andersag et al.

    Quinoline compound and process of making the same. German Pat. 683 692

    Chem. Abstr.

    (1942)
  • P. Augustijns et al.

    Chloroquine levels in blood during chronic treatment of patients with rheumatoid arthritis

    Eur. J. Clin. Pharmacol.

    (1992)
  • D.I. Beardsley et al.

    N-methyl-N′-nitro-N-nitrosoguanidine activates cell-cycle arrest through distinct mechanisms activated in a dose-dependent manner

    Mol. Pharmacol.

    (2005)
  • W.H. Beierwaltes et al.

    Scintillation scanning of malignant melanomas with radioiodinated quinoline derivatives

    J. Nucl. Med.

    (1968)
  • R.P. Bissonnette et al.

    Apoptotic cell death induced by c-myc is inhibited by bcl-2

    Nature

    (1992)
  • B. Borgonovo et al.

    Regulated exocytosis: a novel, widely expressed system

    Nat. Cell Biol.

    (2002)
  • E. Bork et al.

    Teniposide and etoposide in previously untreated small-cell lung cancer: a randomized study

    J. Clin. Oncol.

    (1991)
  • P. Boya et al.

    Mitochondrial membrane permeabilization is a critical step of lysosome-initiated apoptosis induced by hydroxychloroquine

    Oncogene

    (2003)
  • M.J. Boyer et al.

    Regulation of intracellular pH in tumor cell lines: influence of microenvironmental conditions

    Cancer Res.

    (1992)
  • A.M. Breckenridge et al.

    Clinical pharmacology and malaria

    Ann. Trop. Med. Parasitol.

    (1997)
  • E. Briceno et al.

    Therapy of glioblastoma multiforme improved by the antimutagenic chloroquine

    Neurosurg. Focus

    (2003)
  • D. Butler et al.

    Cellular responses to protein accumulation involve autophagy and lysosomal enzyme activation

    Rejuvenation Res.

    (2005)
  • T. Chatterjee et al.

    Comparative mutagenic and genotoxic effects of three antimalarial drugs, chloroquine, primaquine and amodiaquine

    Mutagenesis

    (1998)
  • G.Z. Cheng et al.

    Advances of AKT pathway in human oncogenesis and as a target for anti-cancer drug discovery

    Curr. Cancer Drug Targets

    (2008)
  • J. Cho et al.

    Specific binding of Hoechst 33258 to site 1 thymidylate synthase mRNA

    Nucleic Acids Res.

    (2000)
  • C.R. Chong et al.

    New uses for old drugs

    Nature

    (2007)
  • G.R. Coatney et al.

    Studies in human malaria; the organization of a program for testing potential antimalarial drugs in prisoner volunteers

    Am. J. Hyg.

    (1948)
  • Cited by (435)

    View all citing articles on Scopus
    View full text