Register      Login
Australian Journal of Chemistry Australian Journal of Chemistry Society
An international journal for chemical science
RESEARCH ARTICLE (Open Access)

Discovery and optimisation of conotoxin Vc1.1 and analogues with analgesic properties

Majbrit Frøsig-Jørgensen https://orcid.org/0000-0002-9349-1691 A , Jing Ji https://orcid.org/0000-0001-9861-0533 A , Declan M. Gorman https://orcid.org/0000-0003-4933-3271 A , Meng-Wei Kan https://orcid.org/0000-0001-8189-834X A and David J. Craik https://orcid.org/0000-0003-0007-6796 A *
+ Author Affiliations
- Author Affiliations

A Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld 4072, Australia.

* Correspondence to: d.craik@imb.uq.edu.au

Handling Editor: Curt Wentrup

Australian Journal of Chemistry 76(10) 655-670 https://doi.org/10.1071/CH23155
Submitted: 16 August 2023  Accepted: 15 September 2023   Published: 6 October 2023

© 2023 The Author(s) (or their employer(s)). Published by CSIRO Publishing. This is an open access article distributed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (CC BY-NC-ND)

Abstract

A specimen of the marine cone snail Conus victoriae collected from a beach in Broome, Western Australia, by a group from The University of Melbourne led to the discovery of the α-conotoxin Vc1.1, which was found to have analgesic activity in rodents. The discovery of this venom-derived peptide led to a series of structural, mechanistic and pharmacological studies directed towards the development of a new analgesic for neuropathic pain by groups in Australia and internationally. Solid-phase peptide synthesis played an important role in developing structure–activity relationships. Studies in a rat model of neuropathic pain showed that a cyclic analogue of the peptide, cVc1.1, had comparable analgesic activity with that of gabapentin, one of the foremost clinically used drugs for neuropathic pain, with cVc1.1 delivered orally at a 120-fold lower dose than gabapentin. Originally, Vc1.1 was believed to act primarily through nicotinic acetylcholine receptors, but evidence for a mechanism mediated through γ-aminobutyric acid B (GABAB) receptors later emerged. Efforts to optimise the binding and pharmacological properties of analogues of Vc1.1 revealed that the affinity towards either receptor can be modulated by sequence mutations, disulfide bond modifications and backbone cyclisation. This Account describes the discovery, structure, chemistry and pharmacology of Vc1.1, with a focus on studies carried out in Australian laboratories.

Keywords: conotoxin, drug design, drug development, drug discovery, GABABR, gamma-aminobutyric acid receptor, nAChR, nicotinic acetylcholine receptors, pain, peptide, pharmacology, venom.

References

Sandall DW, Satkunanathan N, Keays DA, Polidano MA, Liping X, Pham V, Down JG, Khalil Z, Livett BG, Gayler KR. A novel α-conotoxin identified by gene sequencing is active in suppressing the vascular response to selective stimulation of sensory nerves in vivo. Biochemistry 2003; 42: 6904-6911.
| Crossref | Google Scholar | PubMed |

Jakubowski JA, Keays DA, Kelley WP, Sandall DW, Bingham J-P, Livett BG, Gayler KR, Sweedler JV. Determining sequences and post-translational modifications of novel conotoxins in Conus victoriae using cDNA sequencing and mass spectrometry. J Mass Spectrom 2004; 39: 548-557.
| Crossref | Google Scholar | PubMed |

Clark RJ, Fischer H, Nevin ST, Adams DJ, Craik DJ. The synthesis, structural characterization, and receptor specificity of the α-conotoxin Vc1.1. J Biol Chem 2006; 281: 23254-23263.
| Crossref | Google Scholar | PubMed |

Nevin ST, Clark RJ, Klimis H, Christie MJ, Craik DJ, Adams DJ. Are α9α10 nicotinic acetylcholine receptors a pain target for α-conotoxins? Mol Pharmacol 2007; 72: 1406-1410.
| Crossref | Google Scholar | PubMed |

Callaghan B, Haythornthwaite A, Berecki G, Clark RJ, Craik DJ, Adams DJ. Analgesic α-conotoxins Vc1.1 and Rg1A inhibit N-type calcium channels in rat sensory neurons via GABAB receptor activation. J Neurosci 2008; 28: 10943-10951.
| Crossref | Google Scholar | PubMed |

Metabolic Pharmaceuticals Limited. Metabolic’s neuropathic pain drug, ACV1 – Clinical trials update. Melbourne, Vic., Australia: ASX; 2006. Available at https://www.asx.com.au/asxpdf/20061129/pdf/3zv2c96tyh1nx.pdf[cited 11 October 2018].

Metabolic Pharmaceuticals Limited. Metabolic’s neuropathic pain drug ACV1 – additional preclinical studies reveal greater potential. Melbourne, Vic., Australia: ASX; 2006. Available at https://www.asx.com.au/asxpdf/20061123/pdf/3zqm91n1jhpff.pdf [cited 11 October 2018].

Metabolic Pharmaceuticals Limited. Metabolic discontinues clinical trial programme for neuropathic pain drug, ACV1. Melbourne, Vic., Australia: ASX; 2007. Available at https://www.asx.com.au/asxpdf/20070814/pdf/313yjgpf7jl4lg.pdf[cited 11 October 2018].

Kennedy AC, Belgi A, Husselbee BW, Spanswick D, Norton RS, Robinson AJ. α-conotoxin peptidomimetics: probing the minimal binding motif for effective analgesia. Toxins 2020; 12: 505.
| Crossref | Google Scholar | PubMed |

10  Li X, Tae H-S, Chu Y, Jiang T, Adams DJ, Yu R. Medicinal chemistry, pharmacology, and therapeutic potential of α-conotoxins antagonizing the α9α10 nicotinic acetylcholine receptor. Pharmacol Ther 2021; 222: 107792.
| Crossref | Google Scholar | PubMed |

11  Jin A-H, Muttenthaler M, Dutertre S, Himaya SWA, Kaas Q, Craik DJ, Lewis RJ, Alewood PF. Conotoxins: chemistry and biology. Chem Rev 2019; 119: 11510-11549.
| Crossref | Google Scholar | PubMed |

12  Olivera BM, Hillyard DR, Marsh M, Yoshikami D. Combinatorial peptide libraries in drug design: lessons from venomous cone snails. Trends Biotechnol 1995; 13: 422-426.
| Crossref | Google Scholar | PubMed |

13  Livett BG, Gayler KR, Khalil Z. Drugs from the sea: conopeptides as potential therapeutics. Curr Med Chem 2004; 11: 1715-1723.
| Crossref | Google Scholar | PubMed |

14  Norton RS, Olivera BM. Conotoxins down under. Toxicon 2006; 48: 780-798.
| Crossref | Google Scholar | PubMed |

15  Halai R, Craik DJ. Conotoxins: natural product drug leads. Nat Prod Rep 2009; 26: 526-536.
| Crossref | Google Scholar | PubMed |

16  Muttenthaler M, Akondi KB, Alewood PF. Structure–activity studies on alpha-conotoxins. Curr Pharm Des 2011; 17: 4226-4241.
| Crossref | Google Scholar | PubMed |

17  Lewis RJ, Dutertre S, Vetter I, Christie MJ. Conus venom peptide pharmacology. Pharmacol Rev 2012; 64: 259-298.
| Crossref | Google Scholar | PubMed |

18  Akondi KB, Muttenthaler M, Dutertre S, Kaas Q, Craik DJ, Lewis RJ, Alewood PF. Discovery, synthesis, and structure–activity relationships of conotoxins. Chem Rev 2014; 114: 5815-5847.
| Crossref | Google Scholar | PubMed |

19  Robinson SD, Norton RS. Conotoxin gene superfamilies. Mar Drugs 2014; 12: 6058-6101.
| Crossref | Google Scholar | PubMed |

20  Gao B, Peng C, Yang J, Yi Y, Zhang J, Shi Q. Cone anails: a big store of conotoxins for novel drug discovery. Toxins 2017; 9: 397.
| Crossref | Google Scholar | PubMed |

21  Zhang R, Snyder GH. Factors governing selective formation of specific disulfides in synthetic variants of alpha-conotoxin. Biochemistry 1991; 30: 11343-11348.
| Crossref | Google Scholar | PubMed |

22  Kaas Q, Westermann J-C, Craik DJ. Conopeptide characterization and classifications: an analysis using ConoServer. Toxicon 2010; 55: 1491-1509.
| Crossref | Google Scholar | PubMed |

23  Yu R, Kompella SN, Adams DJ, Craik DJ, Kaas Q. Determination of the α-conotoxin Vc1.1 binding site on the α9α10 nicotinic acetylcholine receptor. J Med Chem 2013; 56: 3557-3567.
| Crossref | Google Scholar | PubMed |

24  van Lierop BJ, Robinson SD, Kompella SN, Belgi A, McArthur JR, Hung A, MacRaild CA, Adams DJ, Norton RS, Robinson AJ. Dicarba α-conotoxin Vc1.1 analogues with differential selectivity for nicotinic acetylcholine and GABAB receptors. ACS Chem Biol 2013; 8: 1815-1821.
| Crossref | Google Scholar | PubMed |

25  Yu R, Seymour VAL, Berecki G, Jia X, Akcan M, Adams DJ, Kaas Q, Craik DJ. Less is more: design of a highly stable disulfide-deleted mutant of analgesic cyclic α-conotoxin Vc1.1. Sci Rep 2015; 5: 13264.
| Crossref | Google Scholar | PubMed |

26  McIntosh JM, Absalom N, Chebib M, Elgoyhen AB, Vincler M. Alpha9 nicotinic acetylcholine receptors and the treatment of pain. Biochem Pharmacol 2009; 78: 693-702.
| Crossref | Google Scholar | PubMed |

27  Gwak YS, Hulsebosch CE. GABA and central neuropathic pain following spinal cord injury. Neuropharmacology 2011; 60: 799-808.
| Crossref | Google Scholar | PubMed |

28  PainAustralia. Painful Facts. Deakin, ACT, Australia: PainAustralia Limited; 2018. Available at http://www.painaustralia.org.au/about-pain/painful-facts [cited 11 October 2018].

29  Colloca L, Ludman T, Bouhassira D, Baron R, Dickenson AH, Yarnitsky D, Freeman R, Truini A, Attal N, Finnerup NB, Eccleston C, Kalso E, Bennett DL, Dworkin RH, Raja SN. Neuropathic pain. Nat Rev Dis Primers 2017; 3: 17002.
| Crossref | Google Scholar | PubMed |

30  Fornasari D. Pharmacotherapy for neuropathic pain: a review. Pain Ther 2017; 6: 25-33.
| Crossref | Google Scholar | PubMed |

31  Miljanich GP. Ziconotide: neuronal calcium channel blocker for treating severe chronic pain. Curr Med Chem 2004; 11: 3029-3040.
| Crossref | Google Scholar | PubMed |

32  Westermann J-C, Clark RJ, Craik DJ. Binding mode of α-conotoxins to an acetylcholine binding protein determined by saturation transfer difference NMR. Protein Peptide Lett 2008; 15: 910-914.
| Crossref | Google Scholar | PubMed |

33  Halai R, Clark RJ, Nevin ST, Jensen JE, Adams DJ, Craik DJ. Scanning mutagenesis of α-conotoxin Vc1.1 reveals residues crucial for activity at the α9α10 nicotinic acetylcholine receptor. J Biol Chem 2009; 284: 20275-20284.
| Crossref | Google Scholar | PubMed |

34  Yu R, Tae H-S, Tabassum N, Shi J, Jiang T, Adams DJ. Molecular determinants conferring the stoichiometric-dependent activity of α-conotoxins at the human α9α10 nicotinic acetylcholine receptor subtype. J Med Chem 2018; 61: 4628-4634.
| Crossref | Google Scholar | PubMed |

35  Sadeghi M, Carstens BB, Callaghan BP, Daniel JT, Tae H-S, O’Donnell T, Castro J, Brierley SM, Adams DJ, Craik DJ, Clark RJ. Structure–activity studies reveal the molecular basis for GABAB-receptor mediated inhibition of high voltage-activated calcium channels by α-conotoxin Vc1.1. ACS Chem Biol 2018; 13: 1577-1587.
| Crossref | Google Scholar | PubMed |

36  Clark RJ, Jensen J, Nevin ST, Callaghan BP, Adams DJ, Craik DJ. The engineering of an orally active conotoxin for the treatment of neuropathic pain. Angew Chem Int Ed 2010; 49: 6545-6548.
| Crossref | Google Scholar | PubMed |

37  Vincler M, Wittenauer S, Parker R, Ellison M, Olivera BM, McIntosh JM. Molecular mechanism for analgesia involving specific antagonism of α9α10 nicotinic acetylcholine receptors. Proc Natl Acad Sci 2006; 103: 17880-17884.
| Crossref | Google Scholar | PubMed |

38  Indurthi DC, Pera E, Kim H-L, Chu C, McLeod MD, Michael McIntosh J, Absalom NL, Chebib M. Presence of multiple binding sites on α9α10 nAChR receptors alludes to stoichiometric-dependent action of the α-conotoxin, Vc1.1. Biochem Pharmacol 2014; 89: 131-140.
| Crossref | Google Scholar | PubMed |

39  Cuny H, de Faoite A, Huynh TG, Yasuda T, Berecki G, Adams DJ. γ-Aminobutyric acid type B (GABAB) receptor expression is needed for inhibition of N-type (Cav2.2) calcium channels by analgesic α-conotoxins. J Biol Chem 2012; 287: 23948-23957.
| Crossref | Google Scholar | PubMed |

40  Berecki G, McArthur JR, Cuny H, Clark RJ, Adams DJ. Differential Cav2.1 and Cav2.3 channel inhibition by baclofen and α-conotoxin Vc1.1 via GABAB receptor activation. J Gen Physiol 2014; 143: 465-479.
| Crossref | Google Scholar | PubMed |

41  Klimis H, Adams DJ, Callaghan B, Nevin S, Alewood PF, Vaughan CW, Mozar CA, Christie MJ. A novel mechanism of inhibition of high-voltage activated calcium channels by α-conotoxins contributes to relief of nerve injury-induced neuropathic pain. Pain 2011; 152: 259-266.
| Crossref | Google Scholar | PubMed |

42  Napier IA, Klimis H, Rycroft BK, Jin AH, Alewood PF, Motin L, Adams DJ, Christie MJ. Intrathecal α-conotoxins Vc1.1, AuIB and MII acting on distinct nicotinic receptor subtypes reverse signs of neuropathic pain. Neuropharmacology 2012; 62: 2202-2207.
| Crossref | Google Scholar | PubMed |

43  Huynh TG, Cuny H, Slesinger PA, Adams DJ. Novel mechanism of voltage-gated N-type (Cav) calcium channel inhibition revealed through α-conotoxin Vc1.1 activation of the GABAB receptor. Mol Pharmacol 2015; 87: 240-250.
| Crossref | Google Scholar | PubMed |

44  Castro J, Harrington AM, Garcia-Caraballo S, Maddern J, Grundy L, Zhang J, Page G, Miller PE, Craik DJ, Adams DJ, Brierley SM. α-Conotoxin Vc1.1 inhibits human dorsal root ganglion neuroexcitability and mouse colonic nociception via GABAB receptors. Gut 2017; 66: 1083-1094.
| Crossref | Google Scholar | PubMed |

45  Castro J, Grundy L, Deiteren A, Harrington AM, O’Donnell T, Maddern J, Moore J, Garcia-Caraballo S, Rychkov GY, Yu R, Kaas Q, Craik DJ, Adams DJ, Brierley SM. Cyclic analogues of α-conotoxin Vc1.1 inhibit colonic nociceptors and provide analgesia in a mouse model of chronic abdominal pain. Br J Pharmacol 2018; 175: 2384-2398.
| Crossref | Google Scholar | PubMed |

46  Carstens BB, Swedberg J, Berecki G, Adams DJ, Craik DJ, Clark RJ. Effects of linker sequence modifications on the structure, stability, and biological activity of a cyclic α-conotoxin. Biopolymers 2016; 106: 864-875.
| Crossref | Google Scholar | PubMed |

47  Carstens BB, Berecki G, Daniel JT, Lee HS, Jackson KAV, Tae H-S, Sadeghi M, Castro J, O’Donnell T, Deiteren A, Brierley SM, Craik DJ, Adams DJ, Clark RJ. Structure–activity studies of cysteine-rich α-conotoxins that inhibit high-voltage-activated calcium channels via GABAB receptor activation reveal a minimal functional motif. Angew Chem Int Ed 2016; 55: 4692-4696.
| Crossref | Google Scholar | PubMed |

48  Chu X, Tae H-S, Xu Q, Jiang T, Adams DJ, Yu R. α‑Conotoxin Vc1.1 structure–activity relationship at the human α9α10 nicotinic acetylcholine receptor investigated by minimal side chain replacement. ACS Chem Neurosci 2019; 10: 4328-4336.
| Crossref | Google Scholar | PubMed |

49  Cai F, Xu N, Liu Z, Ding R, Yu S, Dong M, Wang S, Shen J, Tae H-S, Adams DJ, Zhang X, Dai Q. Targeting of N-type calcium channels via GABAB-receptor activation by α-conotoxin Vc1.1 variants displaying improved analgesic activity. J Med Chem 2018; 61: 10198-10205.
| Crossref | Google Scholar | PubMed |

50  Belgi A, Burnley JV, MacRaild CA, Chhabra S, Elnahriry KA, Robinson SD, Gooding SG, Tae H-S, Bartels P, Sadeghi M, Zhao F-Y, Wei H, Spanswick D, Adams DJ, Norton RS, Robinson AJ. Alkyne-bridged α‑conotoxin Vc1.1 potently reverses mechanical allodynia in neuropathic pain models. J Med Chem 2021; 64: 3222-3233.
| Crossref | Google Scholar | PubMed |

51  Tabassum N, Tae H-S, Jia X, Kaas Q, Jiang T, Adams DJ, Yu R. Role of CysI–CysIII disulfide bond on the structure and activity of α-conotoxins at human neuronal nicotinic acetylcholine receptors. ACS Omega 2017; 2: 4621-4631.
| Crossref | Google Scholar | PubMed |

52  Knuhtsen A, Whitmore C, McWhinnie FS, McDougall L, Whiting R, Smith BO, Timperley CM, Green AC, Kinnear KI, Jamieson AG. α-Conotoxin GI triazole-peptidomimetics: potent and stable blockers of a human acetylcholine receptor. Chem Sci 2019; 10: 1671-1676.
| Crossref | Google Scholar |

53  Lang PM, Burgstahler R, Haberberger RV, Sippel W, Grafe P. A conus peptide blocks nicotinic receptors of unmyelinated axons in human nerves. Neuroreport 2005; 16: 479-483.
| Crossref | Google Scholar | PubMed |

54  Satkunanathan N, Livett B, Gayler K, Sandall D, Down J, Khalil Z. Alpha-conotoxin Vc1.1 alleviates neuropathic pain and accelerates functional recovery of injured neurones. Brain Res 2005; 1059: 149-158.
| Crossref | Google Scholar | PubMed |

55  Wright AB, Norimatsu Y, McIntosh JM, Elmslie KS. Limited efficacy of α-conopeptides, Vc1.1 and RgIA, to inhibit sensory neuron CaV current. eNeuro 2015; 2(1): e0057-0014.2015.
| Crossref | Google Scholar | PubMed |

56  Plazas PV, Katz E, Gomez-Casati ME, Bouzat C, Elgoyhen AB. Stoichiometry of the α9α10 nicotinic cholinergic receptor. J Neurosci 2005; 25: 10905-10912.
| Crossref | Google Scholar | PubMed |

57  Callaghan B, Adams DJ. Analgesic α-conotoxins Vc1.1 and Rg1A inhibit N-type calcium channels in sensory neurons of α9 nicotinic receptor knockout mice. Channels 2010; 4: 51-54.
| Crossref | Google Scholar | PubMed |

58  Christensen SB, Hone AJ, Roux I, Kniazeff J, Pin JP, Upert G, Servent D, Glowatzki E, McIntosh JM. RgIA4 potently blocks mouse α9α10 nAChRs and provides long-lasting protection against oxaliplatin-induced cold allodynia. Front Cell Neurosci 2017; 11: 219.
| Crossref | Google Scholar | PubMed |

59  Galvez T, Parmentier M-L, Joly C, Malitschek B, Kaupmann K, Kuhn R, Bittiger H, Froestl W, Bettler B, Pin J-P. Mutagenesis and modeling of the GABAB receptor extracellular domain support a venus flytrap mechanism for ligand binding. J Biol Chem 1999; 274: 13362-13369.
| Crossref | Google Scholar | PubMed |

60  Bony AR, McArthur JR, Komori A, Wong AR, Hung A, Adams DJ. Analgesic α-conotoxin binding site on the human GABAB receptor. Mol Pharmacol 2022; 102: 196-208.
| Crossref | Google Scholar | PubMed |

61  Finnerup NB, Sindrup SH, Jensen TS. The evidence for pharmacological treatment of neuropathic pain. Pain 2010; 150: 573-581.
| Crossref | Google Scholar | PubMed |

62  Woolf CJ, Mannion RJ. Neuropathic pain: aetiology, symptoms, mechanisms, and management. Lancet 1999; 353(9168): 1959-1964.
| Crossref | Google Scholar | PubMed |