Elsevier

Journal of Controlled Release

Volume 247, 10 February 2017, Pages 28-54
Journal of Controlled Release

Review article
Safe approaches for camptothecin delivery: Structural analogues and nanomedicines

https://doi.org/10.1016/j.jconrel.2016.12.023Get rights and content

Abstract

Twenty-(S)-camptothecin is a strongly cytotoxic molecule with excellent antitumor activity over a wide spectrum of human cancers. However, the direct formulation is limited by its poor water solubility, low plasmatic stability and severe toxicity, which currently limits its clinical use. As a consequence, two strategies have been developed in order to achieve safe and efficient delivery of camptothecin to target cells: structural analogues and nanomedicines. In this review, we summarize recent advances in the design, synthesis and development of camptothecin molecular derivatives and supramolecular vehicles, following a systematic classification according to structure-activity relationships (structural analogues) or chemical nature (nanomedicines). A series of organic, inorganic and hybrid materials are presented as nanoplatforms to overcome camptothecin restrictions in administration, biodistribution, pharmacokinetics and toxicity. Nanocarriers which respond to a variety of stimuli endogenously (e.g., pH, redox potential, enzyme activity) or exogenously (e.g., magnetic field, light, temperature, ultrasound) seem the best positioned therapeutic materials for optimal spatial and temporal control over drug release. The main goal of this review is to be used as a source of relevant literature for others interested in the field of camptothecin-based therapeutics. To this end, final remarks on the most important formulations currently under clinical trial are provided.

Introduction

Camptothecin (CPT) is a water insoluble, natural pentacyclic alkaloid isolated from the oriental tree Camptotheca accuminata by Wall et al. in 1966 [1]. CPT is well-known for its antitumor activity against a wide spectrum of human cancers [2], [3], [4], [5], [6], [7], [8]. In addition, a variety of biological activities, such as pesticidal [9], [10], antipsoriasis [11], [12], antiparasitic [13], antifungal [14], antimicrobial [15] and antiviral [16], [17] has been described for this compound. Certainly, several researchers have reported that CPT inhibits a cellular enzyme DNA topoisomerase I and induces apoptosis in various cancer cells [18], [19].

CPT is a planar pentacyclic quinoline that includes 3 rings of pyrrolo-(3,4-β)-quinoline part (A, B, and C rings), an unsaturated pyridone moiety in ring D and a α-hydroxy lactone ring (E ring), containing one chiral center with (S)-configuration (Fig. 1) [20], [21]. It is worth noting that the isomer 20(R) hydroxyl has little activity while the isomer 20(S) is between 10 and 100 fold more active [22]. Also, it is known that E-ring exists in equilibrium between the lactone form (closed ring, not water insoluble) and the carboxylate form (open ring, water soluble). In acidic pH, the lactone predominates which, according to different clinical trials and structure-activity studies, is the only therapeutic molecule.

However, at physiological pH, the lactone ring is converted into the carboxylate, much less active, which predominates at neutral and alkaline pH [23], [24].

Unfortunately, CPT presents some major limitations with regards to therapeutic application, like poor water solubility and the rapid lactone ring hydrolysis at physiological pH, which gives rise to the inactive carboxylate form [25]. In addition, CPT is extremely insoluble in organic compounds except for dimethyl sulfoxide, in which it shows moderate solubility [26]. Due to CPT insolubility in biocompatible solvents, it is very difficult to apply conventional drug administration routes, including oral, intravenous or intramuscular injection, to distribute this compound throughout the body [27]. Furthermore, there are other negatives aspects that limit the use of CPT in clinical trials: pronounced loss of activity due to lactone-ring hydrolysis, reversibility of drug-target interaction and severe toxicity, including hemorrhagic cystitis and myelotoxicity [28]. All these drawbacks have precluded its clinical application, making necessary to develop alternative compounds and structures for CPT use in humans.

In an effort to improve CPT solubility and pharmacokinetics structural derivatives as topotecan and irinotecan have been developed [29], [30] (see Table 1). Unfortunately, as it will be reported later on, CPT quinoline structure modification usually brings out a dramatic drop of cytotoxic activity, which results one order or even lower than pristine drug, strongly limiting the therapeutic use.

To overcome these issues two strategies have been pointed out for safety and efficient CPT delivery to target cells: i) structural analogues, in which the CPT molecule is chemically modified for increased solubility and stability in biological fluids; and ii) nanomedicines, wherein CPT is incorporated by physico-chemical methods to nanoparticles which act as stable carriers for drug delivery. In this review, we summarize the most advanced and recent achievements in the design, synthesis and development of CPT analogues and nanomedicines, including the most relevant formulations currently under clinical use.

Section snippets

Camptothecin analogues

Since the discovery of CPT and the associated therapeutic limitations, huge scientific efforts are being made to improve the pharmacokinetics, drug resistance, clinical efficacy, and toxicity profiles of the original molecule, by introducing organic ligands [31], [32], [33], [34], [35], [36].

Nanoparticles as drug delivery systems for camptothecin

Many types of nanovehicles have been developed for CPT delivery, and these drug delivery systems (DDSs) may be classified in several ways, according to their chemical nature, structure, composition, morphology, etc. Herein, we present a systematic classification of CPT nanocarriers according to their chemical nature in organic, inorganic and hybrid materials, presenting different properties and behaviors [72], [73], [74], [75]. These novel nanoplatforms have been designed to solve most of the

Stimuli responsive systems

These are currently the most representative DDSs in Nanomedicine. The main characteristic is their ability to deliver a therapeutic agent into the target cells with no previous release. This is due to the accurate control of the release process through a specific intracellular stimulus, which minimizes undesired side effects. Stimuli-responsive nanocarriers can be classified in endogenous or exogenous, depending on the stimulus which activates the drug release mechanism. Examples of endogenous

Clinical testing

One of the most disputable arguments again nanomedicines is that, despite the strong publication statistics over the last decade, very few drugs have reached the commercial market yet [79]. Although it is accepted that nanocarriers improve solubility, efficacy and biodistribution, while decreasing adverse side effects of very cytotoxic drugs as CPT, strict regulations concerning their use in human beings have hampered, so far, a quicker translation of these therapeutic platforms into the

Conclusion and future directions

CPT is one of the most active molecules for cancer treatment, but its particular limitations as poor solubility, lack of stability of the lactone ring and strong toxicity, have encouraged the development of different delivery forms in order to achieve a safe approach. Here, it is a fact that current cancer treatment cannot be tackled as the administration of a single therapeutic molecule but, instead, the new target is personalized medicine, which selects the appropriate drug combination and

Acknowledgements

Financial support of the Spanish Ministry of Economy and Competitiveness (projects MAT2012-39290-C02-02 and SEV-2012-0267) is gratefully acknowledged. Dr. E.M. Rivero thanks the Cursol Foundation for a post-doctoral scholarship. Prof. Eduardo Fernández and Dr. Ibane Abasolo are acknowledged for the useful discussion.

References (219)

  • D. Subrahmanyam et al.

    Novel C ring analogues of 20(S)-camptothecin. Part-2: synthesis and in vitro cytotoxicity of 5-C-substituted 20(S)-camptothecin analogues

    Bioorg. Med. Chem. Lett.

    (1999)
  • C.Y. Wang et al.

    Synthesis and antitumor activity of 20-O-linked nitrogen-based camptothecin ester derivatives

    Bioorg. Med. Chem.

    (2004)
  • Q. Huang et al.

    Evolution in medicinal chemistry of E-ring-modified camptothecin analogs as anticancer agents

    Eur. J. Med. Chem.

    (2013)
  • H. Bleiberg

    CPT-11 in gastrointestinal cancer

    Eur. J. Cancer

    (1999)
  • V. Bala et al.

    Prodrug and nanomedicine approaches for the delivery of the camptothecin analogue SN38

    J. Control. Release

    (2013)
  • S. Lim et al.

    Phase II study of camtobell inj. (belotecan) in combination with cisplatin in patients with previously untreated, extensive stage small cell lung cancer

    Lung Cancer

    (2013)
  • A. Gabizon et al.

    Cancer nanomedicines: closing the translational gap

    Lancet

    (2014)
  • V.J. Venditto et al.

    Cancer nanomedicines: so many papers and so few drugs!

    Adv. Drug Deliv. Rev.

    (2013)
  • V.P. Torchilin

    Multifunctional nanocarriers

    Adv. Drug Deliv. Rev.

    (2006)
  • V. López-Dávila et al.

    Organic nanocarriers for cancer drug delivery

    Curr. Opin. Pharmacol.

    (2012)
  • Y. Matsumura

    Preclinical and clinical studies of NK012, an SN-38-incorporating polymeric micelles, which is designed based on EPR effect

    Adv. Drug Deliv. Rev.

    (2011)
  • R. Jeetah et al.

    Polymeric nanomicelles for sustained delivery of anti-cancer drugs

    Mutat. Res.

    (2014)
  • M. Watanabe et al.

    Preparation of camptothecin-loaded polymeric micelles and evaluation of their incorporation and circulation stability

    Int. J. Pharm.

    (2006)
  • N. Fan et al.

    Fabrication of nanomicelle with enhanced solubility and stability of camptothecin based on α,β-poly[(N-carboxybutyl)-L-aspartamide]-camptothecin conjugate

    Colloids Surf., B

    (2010)
  • W. Zhang et al.

    Nanomicelle with long-term circulation and enhanced stability of camptothecin based on mPEGylated α,β-poly(L-aspartic acid)-camptothecin conjugate

    Colloids Surf., B

    (2010)
  • D.L. Emerson

    Liposomal delivery of camptothecins

    Pharm. Sci. Technol. Today

    (2000)
  • M. Watanabe et al.

    In vivo antitumor activity of camptothecin incorporated in liposomes formulated with an artificial lipid and human serum albumin

    J. Control. Release

    (2008)
  • Y. Cheng et al.

    Potential of poly(amidoamine) dendrimers as drug carriers of camptothecin based on encapsulation studies

    Eur. J. Med. Chem.

    (2008)
  • J. Lim et al.

    Triazine dendrimers as drug delivery systems: from synthesis to therapy

    Adv. Drug Deliv. Rev.

    (2012)
  • M.E. Wall et al.

    Plant antitumor agents. I. The isolation and structure of camptothecin, a novel alkaloidal leukemia and tumor inhibitor from Camptotheca acuminata

    J. Am. Chem. Soc.

    (1966)
  • D.R. Beidler et al.

    Camptothecin resistance involving steps subsequent to the formation of protein-linked DNA breaks in human camptothecin-resistant KB cell lines

    Cancer Res.

    (1996)
  • B.C. Giovanella et al.

    Complete growth inhibition of human cancer xenografts in nude mice by treatment with 20-(S)-camptothecin

    Cancer Res.

    (1991)
  • G. Del Bino et al.

    Diverse effects of camptothecin, an inhibitor of topoisomerase I, on the cell cycle of lymphocytic (L1210, MOLT-4) and myelogenous (HL-60, KG1) leukemic cells

    Cancer Res.

    (1990)
  • L.H. Li et al.

    Action of camptothecin on mammalian cells in culture

    Cancer Res.

    (1972)
  • A.Y. Chen et al.

    DNA topoisomerases: essential enzymes and lethal targets

    Annu. Rev. Pharmacol. Toxicol.

    (1994)
  • N.H. Oberlies et al.

    Camptothecin and taxol: historic achievements in natural products research

    J. Nat. Prod.

    (2004)
  • M.K. Clements et al.

    Camptothecin exhibits selective cytotoxicity towards human breast carcinoma as compared to normal bovine endothelial cells in vitro

    Anti-Cancer Drugs

    (1996)
  • Y.Q. Liu et al.

    Synthesis and insecticidal activities of novel spin-labeled derivatives of camptothecin

    Heteroat. Chem.

    (2011)
  • L. Zhang et al.

    Characterization of DNA topoisomerase-1 in Spodoptera exigua for toxicity evaluation of camptothecin and hydroxycamptothecin

    PLoS One

    (2013)
  • X.R. Lin et al.

    Topical camptothecine in treatment of psoriasis

    Int. J. Dermatol.

    (1988)
  • C.Y. Chiao et al.

    Effect of topical use of camptothecine-dimethyl sulfoxide solution in psoriasis

    Chin. Med. J.

    (1975)
  • A.L. Bodley et al.

    Molecular and cytotoxic effects of camptothecin, a topoisomerase I inhibitor, on trypanosomes and Leishmania

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

    (1995)
  • M. del Poeta et al.

    Comparison of in vitro activities of camptothecin and nitidine derivatives against fungal and cancer cells

    Antimicrob. Agents Chemother.

    (1999)
  • A.N.M.A. Alaghaz et al.

    Synthesis, spectroscopic, and antimicrobial activity studies of novel 10-substituted camptothecin phosphorothioate analogs

    Phosphorus Sulfur

    (2012)
  • S.B. Horwitz et al.

    Antiviral action of camptothecin

    Antimicrob. Agents Chemother.

    (1972)
  • E. Boven et al.

    New analogues of camptothecins. Activity and resistance

    Ann. N. Y. Acad. Sci.

    (2000)
  • P.J. Tobin et al.

    Camptothecins and key lessons in drug design

    Drug Des. Rev. Online

    (2004)
  • W. Muqeet et al.

    Camptothecin and its analogs antitumor activity by poisoning topoisomerase I, their structure activity relationship and clinical development perspective of analogs

    J. Appl. Pharmacol.

    (2014)
  • R. Garcia-Carbonero et al.

    Current perspectives on the clinical experience, pharmacology, and continued development of the camptothecins

    Clin. Cancer Res.

    (2002)
  • S.W. Carrigan et al.

    Comparative molecular field analysis and molecular modeling studies of 20-(S)-camptothecin analogs as inhibitors of DNA topoisomerase I and anticancer/antitumor agents

    J. Comput. Aided Mol. Des.

    (1997)
  • Cited by (0)

    View full text