Elsevier

Drug Discovery Today

Volume 26, Issue 2, February 2021, Pages 577-584
Drug Discovery Today

Review
Post screen
How can the potential of the duocarmycins be unlocked for cancer therapy?

https://doi.org/10.1016/j.drudis.2020.11.020Get rights and content

Highlights

  • Duocarmycins are powerful cell-killing agents with potential to treat tumours.

  • The exceptional potency of the duocarmycins might also be their achilles’ heel.

  • No major resistance mechanism has been linked to duocarmycin treatment.

  • Prodrug/ADC strategies could unlock the potential of duocarmycins for clinical use.

The duocarmycins belong to a class of agent that has fascinated scientists for over four decades. Their exquisite potency, unique mechanism of action, and efficacy in multidrug-resistant tumour models makes them attractive to medicinal chemists and drug hunters. However, despite great advances in fine-tuning biological activity through structure–activity relationship studies (SARS), no duocarmycin-based therapeutic has reached clinical approval. In this review, we provide an overview of the most promising strategies currently used and include both tumour-targeted prodrug approaches and antibody-directed technologies.

Introduction

Duocarmycins are a family of DNA minor groove-binding compounds with exquisite cytotoxicity originally identified in Streptomyces [1]. Cyclopropapyrroloindole-based duocarmycins, such as (+)-duocarmycin SA (DSA, 1) and CC-1065 (2, Fig. 1a), comprise a DNA recognition motif (DNA-RM) and a pharmacophore responsible for alkylating DNA (Fig. 1a). In the AT-rich regions of the minor groove, the N3 position of adenine performs a nucleophilic attack on the least substituted carbon in the duocarmycin cyclopropane in a stereoelectronically dependant manner, forming DNA adducts [2]. Synthetic manipulation of the DNA alkylating subunit (variation in ‘A’, Figs 1c and 2) dramatically impacts the cellular potency in the pM–nM range, with most compounds forcing cells to undergo apoptosis [3] via direct S-phase inhibition and subsequent cell cycle arrest [4].

Seco-duocarmycins derived from common scaffolds, such as CI (3), DSA (4), CPI (5), and CBI (6), are precursor molecules (Fig. 1b) that contain a phenolic hydroxyl group that is responsible for initiating rearrangement of the pharmacophore to produce a cyclopropane-containing cytotoxin, via a process known as spirocyclisation (Fig. 1c). The natural duocarmycin products are produced in the enantiomerically S form at the chloromethane chiral centre. The chiral configuration of the duocarmycins has been demonstrated to influence both the sequence selectivity of DNA alkylation and the potency [5]. In regard to the latter, the S enantiomer is 100–1000-fold more potent than the R enantiomer in generating DNA damage, and synthetic approaches are typically aimed at producing the pure S form for this reason [6].

Despite their considerable potential as therapeutics, market approval for the clinical use of duocarmycins has not yet been granted. Clinical administration of adozelesin [7], bizelesin [8] and the two carbamate prodrugs carzelesin [9] and KW-2189 [10] (carbamate protection shown in Figure 11 and 12, respectively) has been associated with severe adverse effects and no therapeutic index, leading to all trials being discontinued. Despite this clinical failure, interest in these molecules has remained high and has led to a large body of work that has demonstrated fascinating insights in both biological and chemical explorations [6]. The inherent capacity of duocarmycins to evade traditional resistance mechanisms and retain exquisite potency in multidrug-resistant cells 11, 12, 13 still attracts considerable interest from both academia and industry to develop new approaches for this class of molecule, with a focus on expanding the therapeutic index for patient benefit. Accordingly, in this review, we outline and discuss the most promising drug discovery strategies over the past decade focussed on maximising the potential of these powerful compounds derived and inspired by nature.

Section snippets

Hypoxia-activated duocarmycin prodrugs

Solid tumours typically contain regions of high cell density and poor vascularisation leading to low oxygenation and hypoxia. These regions are often therapy resistant and house some of the most aggressive cancerous cells [14]. Although hypoxic fractions remain an obstacle to effective treatment, they also offer opportunities for the development of hypoxia-activated prodrugs (HAPs). Generally, HAPs are designed to exploit the presence of one or two-electron oxidoreductases, which can catalyse

Antibody–drug conjugates

ADCs have gained prominence over the past decade, with nine approved drugs and >100 in clinical trials. ADCs combine the excellent targeting properties of antibodies with the cell-killing power of conjugated payloads [45]. Currently, none of the clinically approved ADCs have a duocarmycin as payload. Investigations to this end are discussed further herein.

Small-molecule drug conjugate and peptide–drug conjugate

Small-molecule drug conjugates (SMDC) and peptide–drug conjugates (PDC) are attractive because they are smaller than ADCs and, therefore, are likely to confer superior tumour penetration. Both SMDCs and PDCs are designed to exploit the specificities of protein-targeting small molecules or peptides and the cytotoxicity of nonspecific chemotoxins by combining them with a cleavable linker [67].

Initial work published in 2014 demonstrated that duocarmycins could be viable payloads for SMDCs by

Future perspectives

Nature has undoubtedly produced intricate compound architectures that have fascinated and inspired drug hunters for medicinal use. Despite the promise of the powerful cell-killing duocarmycins, they have yet to be approved for clinical use. As reviewed here, several promising tumour-selective prodrug and ADC approaches are underway, with the latter strategy especially gaining momentum. For both prodrug and ADC strategies, low enzyme or antigen expression as well as tumour penetration are

Acknowledgements

The authors would like to thank Institute of Cancer Therapeutics Doctoral Training Centre for a PhD studentship to Z.J. and Yorkshire Cancer Research (Programme grant B381PA) for supporting our work focused on exploring CYPs as targets for duocarmycins bioprecursor development.

References (73)

  • O.C. Cartwright

    A peptide–duocarmycin conjugate targeting the Thomsen-Friedenreich antigen has potent and selective antitumor activity

    Bioconjug. Chem.

    (2020)
  • L.J. Hanka

    CC-1065 (NSC-298223), a new antitumor antibiotic. production, in vitro biological activity, microbiological assays and taxonomy of the producing microorganism

    J. Antibiot.

    (1978)
  • D.L. Boger et al.

    CC-1065 and the duocarmycins: understanding their biological function through mechanistic studies

    Angew. Chem. Int. Ed. English

    (1996)
  • B.K. Bhuyan

    Cell cycle effects of CC-1065

    Cancer Res.

    (1983)
  • J.P. Lajiness et al.

    Asymmetric synthesis of 1,2,9,9a-tetrahydrocyclopropa c benzo e indol-4-one (CBI)

    J. Org. Chem.

    (2011)
  • N. Ghosh

    Chemical and biological explorations of the family of CC-1065 and the duocarmycin natural products

    Curr. Top. Med. Chem.

    (2009)
  • M. Cristofanilli

    Phase II study of adozelesin in untreated metastatic breast cancer

    Anticancer Drugs

    (1998)
  • N. Pavlidis

    Carzelesin phase II study in advanced breast, ovarian, colorectal, gastric, head and neck cancer, non-Hodgkin’s lymphoma and malignant melanoma: a study of the EORTC early clinical studies group (ECSG)

    Cancer Chemother. Pharmacol.

    (2000)
  • S.N. Markovic

    Phase II trial of KW2189 in patients with advanced malignant melanoma

    Am. J. Clin. Oncol.

    (2002)
  • K. Gomi

    Anticellular and antitumor activity of duocarmycins, novel antitumor antibiotics

    Jpn. J. Cancer Res.

    (1992)
  • H. Ogasawara

    Intracellular carboxyl esterase activity is a determinant of cellular sensitivity to the antineoplastic agent KW‐2189 in cell lines resistant to cisplatin and CPT‐11

    Jpn. J. Cancer Res.

    (1995)
  • M. Nadal-Serrano

    The second generation antibody-drug conjugate SYD985 overcomes resistances to T-DM1

    Cancers

    (2020)
  • J.M. Brown et al.

    Exploiting tumour hypoxia in cancer treatment

    Nat. Rev. Cancer

    (2004)
  • M. Tercel

    Preparation and antitumour properties of the enantiomers of a hypoxia-selective nitro analogue of the duocarmycins

    ChemMedChem

    (2011)
  • M. Tercel

    Hypoxia-activated prodrugs: substituent effects on the properties of nitro seco-1,2,9,9a-tetrahydrocyclopropacbenzeindol-4-one (nitroCBI) prodrugs of DNA minor groove alkylating agents

    J. Med. Chem.

    (2009)
  • M. Tercel

    Selective treatment of hypoxic tumor cells in vivo: phosphate pre-prodrugs of nitro analogues of the duocarmycins

    Angew. Chem. Int. Ed.

    (2011)
  • M. Tercel

    Influence of a basic side chain on the properties of hypoxia-selective nitro analogues of the duocarmycins: demonstration of substantial anticancer activity in combination with irradiation or chemotherapy

    J. Med. Chem.

    (2017)
  • Nitro compounds. https://go.drugbank.com/categories/DBCAT000765 [Accessed 16 November...
  • P. Kovacic et al.

    Nitroaromatic compounds: environmental toxicity, carcinogenicity, mutagenicity, therapy and mechanism

    J. Appl. Toxicol.

    (2014)
  • W. Jin

    A unique class of duocarmycin and CC-1065 analogues subject to reductive activation

    J. Am. Chem. Soc.

    (2007)
  • J.P. Lajiness

    Design, synthesis, and evaluation of duocarmycin O-amino phenol prodrugs subject to tunable reductive activation

    J. Med. Chem.

    (2010)
  • A.L. Wolfe

    Efficacious cyclic N-acyl O-amino phenol duocarmycin prodrugs

    J. Med. Chem.

    (2013)
  • F.W. Hunter

    Hypoxia-activated prodrugs: paths forward in the era of personalised medicine

    Br. J. Cancer

    (2016)
  • C. Rodriguez-Antona et al.

    Cytochrome P450 pharmacogenetics and cancer

    Oncogene

    (2006)
  • H.M. Sheldrake

    Re-engineering of the duocarmycin structural architecture enables bioprecursor development targeting CYP1A1 and CYP2W1 for biological activity

    J. Med. Chem.

    (2013)
  • K. Pors

    Modification of the duocarmycin pharmacophore enables CYP1A1 targeting for biological activity

    Chem. Commun.

    (2011)
  • Cited by (16)

    • Site-specific drug delivery utilizing monoclonal antibodies

      2023, Advanced and Modern approaches for Drug Delivery
    • Duocarmycin-based antibody–drug conjugates as an emerging biotherapeutic entity for targeted cancer therapy: Pharmaceutical strategy and clinical progress

      2021, Drug Discovery Today
      Citation Excerpt :

      Second, the DCM mechanism of action is defined. These compounds bind to the DNA minor groove at A-T-rich regions and alkylate adenine residues at the N3 position, which results in a DNA strand break [34,35]. Third, cancer cells showing hypoxic, chemoresistant, and stemness phenotypes are sensitive to DCMs [34,35].

    View all citing articles on Scopus
    View full text