Melanocyte-Directed enzyme prodrug therapy (MDEPT): Development of second generation prodrugs for targeted treatment of malignant melanoma

https://doi.org/10.1016/S0968-0896(01)00039-6Get rights and content

Abstract

Evaluation of second generation prodrugs for MDEPT, by oximetry, has highlighted structural properties that are advantageous and disadvantageous for efficient oxidation using mushroom tyrosinase. In particular, a sterically undemanding prodrug bis-(2-chloroethyl)amino-4-hydroxyphenylaminomethanone 28 was synthesised and found to be oxidised by mushroom tyrosinase at a superior rate to tyrosine methyl ester, the carboxylic acid of which is the natural substrate for tyrosinase. The more sterically demanding phenyl mustard prodrugs 9 and 10 were oxidised by mushroom tyrosinase at a similar rate to tyrosine methyl ester. In contrast, tyramine chain elongation via heteroatom insertion was detrimental and the rate of mushroom tyrosinase oxidation of phenyl mustard prodrugs 21 and 22 decreased by 10 nanomol/min.

Evaluation of second generation prodrugs for MDEPT, by oximetry, has highlighted structural properties that are advantageous and disadvantageous for efficient oxidation using mushroom tyrosinase. In particular, a sterically undemanding prodrug bis-(2-chloroethyl)amino-4-hydroxyphenylaminomethanone was synthesised and found to be oxidised by mushroom tyrosinase at a superior rate to tyrosine methyl ester, the carboxylic acid of which is the natural substrate for tyrosinase.

  1. Download : Download full-size image

Introduction

Malignant melanoma continues to be a serious clinical problem on account of its increasing incidence, the relatively young population that it affects and the poor prognosis of the disseminated disease.1 The high mortality rate is a reflection of the failure of melanoma cells to respond to current cytotoxic treatment in the form of radiation and chemotherapy. Thus, there is an urgent need for improved treatment.

A selective strategy towards the treatment of malignant melanoma in the form of a melanocyte-directed enzyme prodrug therapy (MDEPT) has previously been reported by our group.2 Based on the unique occurrence of tyrosinase expression in melanocytes,3 MDEPT offers a highly selective drug delivery system. Therefore, with increased selectivity in the delivery of cytotoxic agents, intolerable side effects and the need for administration of large amounts of drugs should be minimised. Initial studies into MDEPT were carried out using prodrug 1 (Scheme 1). The proposed drug release mechanism, mediated by tyrosinase, is outlined in scheme 1.

Prodrug 1 can be viewed as a three component system, with a dopamine moiety acting as the tyrosinase substrate, a drug such as phenyl mustard, as the cytotoxic agent and a carbamate linkage coupling the two components together. In order to further develop this strategy, we have now prepared a more extensive range of compounds and examined their ability to be oxidised by mushroom tyrosinase. Since oxidation by tyrosinase is the first step in our proposed drug release mechanism, it is essential that good substrates for tyrosinase are identified at an early stage of our programme.

Three different cytotoxic agents were incorporated within the prodrugs, namely, phenyl mustard,4 a bis-ethyl amine mustard5 and daunomycin.6 All three of these cytotoxic agents have been previously used as anti-cancer drugs in the clinic.

Herein, we describe the synthesis and analysis of the prodrugs, with particular emphasis on their ability to act as substrates for tyrosinase, as determined by oximetry.

Section snippets

Synthesis of phenyl mustard prodrugs

The general protocol for the synthesis of the various prodrugs was based on the premise of forming a reactive carbonate that was prone to nucleophilic attack by a primary or secondary amine. Prodrugs with a carbamate linkage were synthesised as previously reported, and required access to the reactive carbonate intermediate 6. Facile preparation of p-hydroxyphenyl mustard hydrochloride Scheme 2, Scheme 3 was achieved by treating benzyloxyaniline 2 with ethylene oxide to give diol 3.7 Conversion

Conclusions

A second generation of prodrugs has been synthesised and the abilities of the individual prodrugs to act as substrates for tyrosinase have been examined. Prodrugs that closely resemble natural tyrosinase substrates were readily oxidised making them suitable MDEPT candidates. Functional group transformation, that is, carbamate to thiocarbamate resulted in a decrease in tyrosinase oxidation over 25 min incubation; Graph 1, PD(12) versus PD(25). Removal of steric bulk from the substrate's oxidative

Experimental

All NMR spectra were recorded on a Bruker WM250, Bruker AC250, Bruker Avance DPX 250, Bruker AMX400 or Jeol AX400 spectrometer, using CHCl3 as an internal standard unless stated otherwise (7.26 ppm for 1H NMR, 77.0 ppm for 13C NMR). 13C spectra were recorded using Distortionless Enhancement by Polarisation Transfer. Mass spectra were recorded on a Fisons VG Autospec. Infra-red spectra were recorded on a Perkin–Elmer Paragon 1000 FT-IR spectrometer. Optical activities were determined using a

Acknowledgements

We gratefully acknowledge Phairson Medical Ltd (Post-doctoral fellowship to H.M.), the BBSRC (Post-doctoral fellowship to A.M.J., grant no. 45/SBDO7534) and the Royal Society for their financial support of this work. We also gratefully acknowledge the Drug Synthesis & Chemistry Branch, Development Therapeutics Program, Division of Cancer Treatment, National Cancer Institute for the donation of daunomycin.

References (15)

  • A.M. Jordan et al.

    Bio. Org. Med. Chem.

    (1999)
  • T. Nakagawa et al.

    Tetrahedron Lett.

    (1994)
  • K.A. Jorgensen et al.

    Tetrahedron

    (1982)
  • M. Artico et al.

    J. Biochem. Pharmacol.

    (1968)
  • Grin-Jorgensen, C.; Rigel, D. S.; Friedman, R. J. Cutaneous Melanoma, 2nd ed.; J. B. Lippincott, 1992; pp 27–39....
  • D. Valmori et al.

    Cancer Res.

    (1999)
  • R. Lougerstay-Madec et al.

    Anti-Cancer Drug Des.

    (1998)
There are more references available in the full text version of this article.

Cited by (49)

  • Nanomedicine: A pragmatic approach for tackling melanoma skin cancer

    2023, Journal of Drug Delivery Science and Technology
  • smProdrugs: A repository of small molecule prodrugs

    2023, European Journal of Medicinal Chemistry
  • Why Great Mitotic Inhibitors Make Poor Cancer Drugs

    2020, Trends in Cancer
    Citation Excerpt :

    For reasons discussed earlier (inappropriate pharmacophore), these approaches had shortcomings that preluded further clinical development. Despite the drawbacks in rationale, these applications show that rational design of a chemical moiety can be subject to TYR activation [134–136]. Changing the pharmacophore to a mitotic kinase inhibitor inherently narrows the scope of possible targetable cells to those that are actively dividing.

  • The selective cytotoxicity of new triazene compounds to human melanoma cells

    2017, Bioorganic and Medicinal Chemistry
    Citation Excerpt :

    Because aniline is always derived from the precursor MMT and in its mass balance sometimes reaches 100%, it is our understanding that the 5% value obtained for MMT is not necessarily a low value. In fact, the MMT release from derivatives 3c–f is much faster in comparison with other drug release pathways described in the literature for tyrosinase activation.29 In the assays in which we did not observe a complete mass balance for the aniline, the observation could be explained by the fact that aromatic amines (e.g.: anilines) are also tyrosinase substrates.

  • Synthesis and analysis of activity of a potential anti-melanoma prodrug with a hydrazine linker

    2014, European Journal of Medicinal Chemistry
    Citation Excerpt :

    The concept of selectively releasing cytotoxic agents in melanocytes from tyrosinase-activated prodrugs was developed more than a decade ago and named Melanocyte-Directed Enzyme Prodrug Therapy (MDEPT) [9]. Initially, the cytotoxic agents were attached to the primary amino group of tyrosinase substrates (tyrosine, dopamine and related compounds) by carbamate or urea linkers [9–11]. However, later studies on such tyramine and dopamine derivatives showed that conversion of the amino group to amide, carbamate or urea derivatives made the nitrogen atom insufficiently nucleophilic for cyclization of the corresponding o-quinones to dihydroxydihydroindoles and therefore the release of the active group was unlikely [12].

View all citing articles on Scopus

Present address: Ribotargets Ltd, Granta Park, Abington, Cambridge CB1 6GB, UK.

View full text