Research paper
Comparative analysis of the dual EGFR-DNA targeting and growth inhibitory properties of 6-mono-alkylamino- and 6,6-dialkylaminoquinazoline-based type II combi-molecules

https://doi.org/10.1016/j.ejmech.2020.112185Get rights and content

Highlights

  • Development of the first synthetic strategy to access 6-N, N-disubstituted quinazolines.

  • Comparison between di-substituted and mono-substituted EGFR-DNA combimolecules

  • Hemi-mustard combi-molecules induce less DNA damage than full mustards.

  • In contrast to mono-, di-substitution adversely affects EGFR binding affinity.

  • In silico and X-Ray studies confirmed that steric bulk hinder EGFR binding.

Abstract

Over the past decade, we described a novel tumour targeted approach that sought to design “combi-molecules” to hit two distinct targets in tumour cells. Here, to generate small combi-molecules with strong DNA damaging potential while retaining EGFR inhibitory potency, we developed the first synthetic strategy to access the 6-N, N-disubstituted quinazoline scaffold and designed JS61 to possess a nitrogen mustard function directly attached to the 6-position of the quinazoline ring. We compared its biological activity with that of structures containing either a hemi mustard or a non-alkylating substituent. Surprisingly, the results showed that JS61, while capable of inducing strong DNA damage, exhibited moderate EGFR inhibitory potency. In contrast, “combi-molecules” with no bulky substituent at the N-6 position (e.g. ZR2002 and JS84) showed stronger EGFR and growth inhibitory potency than JS61 in a panel of lung cancer cells. To rationalize these results, X-ray crystallography and molecular modeling studies were undertaken, and the data obtained indicated that bulkiness of the 6-N,N-disubstituted moieties hinder its binding to the ATP site and affects binding reversibility.

Introduction

The epidermal growth factor receptor (EGFR) is known to be overexpressed in a wide variety of human cancers including non-small cell lung carcinoma (NSCLC) for which this overexpression has been associated with poor prognosis [[1], [2], [3], [4]]. Significant advances in the biological understanding of EGFR expression and its mutational status have led to the design of several inhibitors. A large variety of first-class EGFR inhibitors have been developed such as gefitinib (1), which has been used in advanced NSCLC patients (Fig. 1) [[5], [6], [7]]. However, acquired resistance to these inhibitors is frequently developed, leading to the design of second and third generation of EGFR inhibitors. These include afatinib (2) (2nd generation) and osimertinib (3) (3rd generation) [[8], [9], [10]]. However, acquired resistance still arises rapidly, generally over a period of 9–13 months [[11], [12], [13]].

Another approach to enhance the potency of EGFR inhibitor-based treatment consists of its combination with cytotoxic agents. Indeed, several studies showed that the combination of an inhibitor of EGFR and a DNA damaging agent induced either additive or synergistic growth inhibitory effects [14]. However, one of the major challenges of combination therapy is the risk of additive toxicity associated with the deleterious effects of each individual drug. To circumvent these problems, we developed a novel drug design approach termed “combi-targeting” that seeks to reduce the pharmacotoxicity of multiple antitumour mechanisms to that of a single drug. The proof-of-concept of this approach has already been confirmed in our laboratory [[15], [16], [17], [18], [19], [20], [21], [22], [23], [24]]. We classified these “combi-molecules” in different types according to their mechanism of action. As depicted in Fig. 2, type I combi-molecule (EGFR-DNA) is designed as a single agent that, intact, can act exclusively on EGFR. Hydrolysis under physiological conditions allows the activation of the second targeting arm and the subsequent release of both bioactive species, whereupon they can act independently on both their targets EGFR and DNA. Type II combi-molecules are systems that cannot undergo hydrolysis and the single molecule can act only on one of its respective targets. Finally, a type III combi-molecule; AL776, has been designed which acts as a hybrid between type I and type II, but is not described here [25]. Regardless of the combi-molecule type, these combi-molecules have shown superior potency and/or reduced toxicity when compared with traditional combinations that compose these molecules, indicating the potential therapeutic benefit of the whole being greater than the sum of its parts. However, the synthesis of multifunctional molecular systems capable of reacting with or binding to the target, and ultimately overcoming drug resistance is a real challenge. Indeed, these systems must maintain strong binding affinity for their desired targets, have sufficient solubility for biological applications and be of appropriate size for diffusion through the cell membrane.

ZR2002 (5) was the first type II combi-molecule designed to inhibit EGFR while inducing DNA damage through its 2-chloroethylamino moiety (Fig. 1, compound 5) [23]. Previous work on this molecule has shown that it can target either EGFR with its inhibitory scaffold or DNA with its 2-chloroethyl function, however a single molecule cannot effect both at the same time (Fig. 2). We have recently achieved a key step in its pre-clinical development by demonstrating that ZR2002 was able to increase overall survival of mice intracranially injected with glioblastoma mesenchymal stem cells harboring temozolomide resistance [26].

In order to enhance the lipophilicity of the drug as well as DNA alkylating potential, we designed and synthesized JS61 (Chart 1), a type II combi-molecule carrying an additional chloroethyl group on the nitrogen, leading to a highly reactive aromatic nitrogen mustard moiety. We and others have already studied N-mustard-quinazoline molecules (Fig. 1, compound 6) [21,[27], [28], [29]]. However, these molecules were only achieved through coupling with a linker between the nitrogen mustard and the quinazoline moieties. Furthermore, the addition of a lipophilic chloroethyl group was expected to enhance its lipophilicity without significantly increasing its molecular weight (<500 MW), a property that may be attractive for penetration through the blood brain barrier and potential indication for the therapy of advanced brain tumours.

Herein, for the first time, the synthesis and the biological activity of the first prototype of 6,6-dialkylaminoquinazoline JS61, a combi-molecule with a nitrogen mustard directly grafted on the quinazoline scaffold, is described. Further, we challenged its activity by replacing the chloroethyl group(s) with various non alkylating moieties such as methyl (JS84), hydroxyethyl and acetoxyethyl (Chart 1) and compared their EGFR binding affinity and growth inhibitory potencies with those of ZR2002. To rationalize the results, we performed molecular modeling and X-ray diffraction analyses of the N-disubstituted combi-molecules JS61, JS84 and the N-monosubstituted ZR2002.

Section snippets

Chemistry

As previously reported, synthetic strategies involving direct alkylation of the aminoquinazoline or formation of the unstable chloroethyl triazeno-quinazoline to form ZR2002 were tedious [22,30]. Herein, we describe for the first time a new synthetic pathway that allows the facile synthesis of 6-N,N-disubstituted aminoquinazoline in good yield. Although more recent generations of EGFR inhibitors (e.g. osimertinib, Fig. 1, molecule 3) showed unique potency against resistant EGFR mutants, we

Conclusion

The purpose of this study was primarily to determine the effect of adding a second 2-chloroethyl substituent moiety on the potency of the type II combi-molecule of the aminoquinazoline class. The study conclusively demonstrated that the formation of a mustard directly grafted onto the quinazoline moiety, while increasing DNA damaging potential, affected the EGFR targeting potency in a rather complex way. Indeed, JS61 was shown to be more potent in the Comet assay, whereas its potency was not

General procedures

All chemicals were of the best commercially available grade and used without further purification. All chemicals were purchased from Sigma Aldrich excepted for the starting material 5-fluoro-2-nitrobenzonitrile purchased from ArkPharm. Anhydrous solvents were purchased from Aldrich Chemicals. The conversion of starting materials was monitored by thin-layer chromatography (TLC) using silica gel plates (silica gel 60 F254 and visualization when required was achieved using UV light (254 and

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

This study was supported by CIHR (MOP-130363). The present study was partially supported by Canada-Brazil bilateral project DFATD-CAPES-88887.137283/2017–00. We also thank DFATD-CAPES for ING and MNR fellowships. JS thanks the Research Institute of the McGill University Health Centre (RI MUHC) for financial support. The China Scholarship Council (CSC) is acknowledged for SH scholarship. We wish to thank the Center for Translational Biology (CTB) of the RI MUHC Drug Discovery Platform for NMR

References (41)

  • G.V. Scagliotti et al.

    The biology of epidermal growth factor receptor in lung cancer

    Clin. Canc. Res.

    (2004)
  • D. Veale et al.

    The relationship of quantitative epidermal growth factor receptor expression in non-small cell lung cancer to long term survival

    Br. J. Canc.

    (1993)
  • M. Volm et al.

    Prognostic value of ERBB-1, VEGF, cyclin A, FOS, JUN and MYC in patients with squamous cell lung carcinomas

    Br. J. Canc.

    (1998)
  • G. Bethune et al.

    Epidermal growth factor receptor (EGFR) in lung cancer: an overview and update

    J. Thorac. Dis.

    (2010)
  • V.D. Cataldo et al.

    Treatment of non–small-cell lung cancer with erlotinib or gefitinib

    N. Engl. J. Med.

    (2011)
  • W. Zhong et al.

    Phase II study of biomarker-guided neoadjuvant treatment strategy for IIIA-N2 non-small cell lung cancer based on epidermal growth factor receptor mutation status

    J. Hematol. Oncol.

    (2015)
  • A. Chi et al.

    EGFR inhibition in non-small cell lung cancer: current evidence and future directions

    Biomarker Research

    (2013)
  • G.M. O’Kane et al.

    Resistance to epidermal growth factor receptor tyrosine kinase inhibitors, T790M, and clinical trials

    Curr. Oncol.

    (2018)
  • K. Politi et al.

    The next wave of EGFR tyrosine kinase inhibitors enter the clinic

    Canc. Cell

    (2015)
  • S. Wang et al.

    Third-generation inhibitors targeting EGFR T790M mutation in advanced non-small cell lung cancer

    J. Hematol. Oncol.

    (2016)
  • B.P. Brown et al.

    On-target resistance to the mutant-selective EGFR inhibitor osimertinib can develop in an allele specific manner dependent on the original EGFR activating mutation

    Clin. Canc. Res.

    (2019)
  • C. Tomasello et al.

    Resistance to EGFR inhibitors in non-small cell lung cancer: clinical management and future perspectives

    Crit. Rev. Oncol.-Hematol.

    (2018)
  • Y.-C. Zhang et al.

    Clinical management of third-generation EGFR inhibitor-resistant patients with advanced non-small cell lung cancer: current status and future perspectives

    Canc. Lett.

    (2019)
  • L.A. Knight et al.

    The in vitro effect of gefitinib (’Iressa’) alone and in combination with cytotoxic chemotherapy on human solid tumours

    BMC Canc.

    (2004)
  • R. Banerjee et al.

    The combi-targeting concept: mechanism of action of the pleiotropic combi-molecule RB24 and discovery of a novel cell signaling-based combination principle

    Cell. Signal.

    (2011)
  • M.I. Todorova et al.

    Subcellular distribution of a fluorescence-labeled combi-molecule designed to block epidermal growth factor receptor tyrosine kinase and damage DNA with a green fluorescent species

    Mol. Canc. Therapeut.

    (2010)
  • Y. Wang et al.

    Synthesis and antitumor activity evaluation of a novel combi-nitrosourea prodrug: BGCNU

    ACS Med. Chem. Lett.

    (2017)
  • G. Sun et al.

    The potential of combi-molecules with DNA-damaging function as anticancer agents

    Future Med. Chem.

    (2017)
  • R. Banerjee et al.

    The combi-targeting concept: selective targeting of the epidermal growth factor receptor- and Her2-expressing cancer cells by the complex combi-molecule RB24

    J. Pharmacol. Exp. Therapeut.

    (2010)
  • M. Heravi et al.

    ZRBA1, a mixed EGFR/DNA targeting molecule, potentiates radiation response through delayed DNA damage repair process in a triple negative breast cancer model

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

    (2015)
  • Cited by (0)

    View full text