A therapeutic combination of two small molecule toxin inhibitors provides broad preclinical efficacy against viper snakebite

Laura-Oana Albulescu, Chunfang Xie, Stuart Ainsworth, Jaffer Alsolaiss, Edouard Crittenden, Charlotte A Dawson, Rowan Softley, Keirah E Bartlett, Robert A Harrison, Jeroen Kool, Nicholas R Casewell, Laura-Oana Albulescu, Chunfang Xie, Stuart Ainsworth, Jaffer Alsolaiss, Edouard Crittenden, Charlotte A Dawson, Rowan Softley, Keirah E Bartlett, Robert A Harrison, Jeroen Kool, Nicholas R Casewell

Abstract

Snakebite is a medical emergency causing high mortality and morbidity in rural tropical communities that typically experience delayed access to unaffordable therapeutics. Viperid snakes are responsible for the majority of envenomings, but extensive interspecific variation in venom composition dictates that different antivenom treatments are used in different parts of the world, resulting in clinical and financial snakebite management challenges. Here, we show that a number of repurposed Phase 2-approved small molecules are capable of broadly neutralizing distinct viper venom bioactivities in vitro by inhibiting different enzymatic toxin families. Furthermore, using murine in vivo models of envenoming, we demonstrate that a single dose of a rationally-selected dual inhibitor combination consisting of marimastat and varespladib prevents murine lethality caused by venom from the most medically-important vipers of Africa, South Asia and Central America. Our findings support the translation of combinations of repurposed small molecule-based toxin inhibitors as broad-spectrum therapeutics for snakebite.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. The geographical distributions and varying…
Fig. 1. The geographical distributions and varying proteomic venom compositions of the medically important viper species used in this study.
The previously defined venom proteomes of Echis ocellatus (Nigeria), Echis carinatus (India), Bothrops asper (Costa Rica), Bitis arietans (Nigeria) and Daboia russelii (Sri Lanka) are presented in pie charts. Toxin family key: SVMP snake venom metalloproteinase; SVSP snake venom serine protease; PLA2 phospholipase A2; CTL C-type lectin; LAAO l-amino acid oxidase; DIS disintegrin; CRISP cysteine-rich secretory protein; KUN Kunitz-type serine protease inhibitor. Geographical species distributions were drawn using QGIS v3.10 software based on data downloaded from the World Health Organization Venomous Snake Distribution database and the IUCN Red List of Threatened Species database.
Fig. 2. Small molecule toxin inhibitors inhibit…
Fig. 2. Small molecule toxin inhibitors inhibit the in vitro SVMP activities of several geographically distinct viper venoms.
a SVMP activities of the five viper venoms quantified by fluorogenic assay. The data presented represent mean measurements and SEMs of area under the curves of fluorescent arbitrary units taken from three independent experimental runs. EOC E. ocellatus; ECAR E. carinatus; BAS Bothrops asper; BAR Bitis arietans; DRUS Daboia russelii. b The effectiveness of metal chelators and peptidomimetic hydroxamate inhibitors at inhibiting the SVMP activity of the various viper venoms. Drug concentrations from 150 µM to 150 nM (highest to lowest dose tested) are presented. The data is expressed as percentage of the venom-only sample (100%, dashed red line). The negative control is presented as an interval (dashed black lines) and represents the values recorded in the PBS-only samples (expressed as percentage of venom activity), where the highest and the lowest values in each set of independent experiments are depicted. Inhibitors are color-coded (dimercaprol, red; DMPS, turquoise; marimastat, purple). c Comparison of SVMP inhibition by marimastat (purple) and batimastat (orange) at two concentrations (1.5 µM, left; 0.15 µM, right), expressed as the percentage of the venom-only sample (100%, dashed red line). All data represent triplicate independent repeats with SEMs, where each technical repeat represents the mean of n ≥ 2 technical replicates. Source data provided in Supplementary Data S1.
Fig. 3. SVMP-inhibitors neutralize the in vitro…
Fig. 3. SVMP-inhibitors neutralize the in vitro procoagulant activities of several geographically distinct viper venoms.
a The coagulopathic activities of the viper venoms, showing that all, except B. arietans (BAR), exhibit procoagulant effects by increasing the clotting velocity in comparison with the normal plasma control. The data presented represents the maximum clotting velocity, calculated as the maximum of the first derivative of each clotting curve, from triplicate independent repeat experiments with SEMs, where each technical repeat represents the mean of n ≥ 2 technical replicates. EOC E. ocellatus; ECAR E. carinatus; BAS Bothrops asper; BAR, Bitis arietans; DRUS, Daboia russelii. b Neutralization of procoagulant venom activity by four SVMP-inhibitors across four drug concentrations (150 µM to 150 nM). The data is expressed as the maximum clotting velocity at each dose. The negative (PBS) and positive (venom-only) controls are presented as intervals (dashed black and red lines, respectively), with the latter representing the mean maximum clotting velocity in these samples ± SEM. Inhibitors are color-coded (dimercaprol, red; DMPS, turquoise; marimastat, purple; batimastat, blue). The data represent triplicate independent repeats with SEMs, where each technical repeat represents the mean of n ≥ 2 technical replicates. Note the different y-axis scales. Source data provided in Supplementary Data S1.
Fig. 4. Nafamostat inhibits the in vitro…
Fig. 4. Nafamostat inhibits the in vitro serine protease activities of several geographically distinct viper venoms.
a The serine protease (SVSP) activity of five viper venoms expressed as the rate (ΔAbs/time/µg venom) of substrate consumption determined by kinetic chromogenic assay. The data represents triplicate independent repeats with SEMs, where each technical repeat represents the mean of n ≥ 2 technical replicates. EOC E. ocellatus; ECAR E. carinatus; BAS Bothrops asper; BAR Bitis arietans; DRUS Daboia russelii. b Neutralization of SVSP venom activity by the serine protease-inhibitor nafamostat. The data is expressed as rates (ΔAbs/time/µg venom) and represents triplicate independent repeats with SEMs, where each technical repeat represents the mean of n ≥ 2 technical replicates. Venom only activity (venom) is displayed alongside venom incubated with decreasing molarities of nafamostat (150 µM to 150 nM). Note the different y-axis scales. Source data provided in Supplementary Data S1.
Fig. 5. Combinations of small molecule toxin…
Fig. 5. Combinations of small molecule toxin inhibitors broadly protect against venom lethality in an in vivo ‘preincubation’ model of snake envenoming.
Kaplan–Meier survival graphs for experimental animals (n = 5) receiving venom (Ven) preincubated (30 min at 37 °C) with different small molecule inhibitors or inhibitor mixes via the intravenous route and monitored for 6 h. Drug-only controls are presented as black dashed lines at the top of each graph (none of the drugs exhibited any observable toxicity at the given doses). a Survival of mice receiving 45 µg of E. ocellatus venom (2.5 × LD50 dose) with and without 60 µg of marimastat or varespladib or nafamostat. b Survival of mice receiving 45 µg of E. ocellatus venom (2.5 × LD50 dose) with and without a dual combination mixture of marimastat and varespladib (MV, 60 µg each) or a triple combination mixture of marimastat, varespladib and nafamostat (MVN, 60 µg each). c Quantified thrombin-antithrombin (TAT) levels in the envenomed animals from (a) and (b). Where the time of death was the same within experimental groups (e.g., early deaths or complete survival) TAT levels were quantified for n = 3, and where times of death varied, n = 5. The data displayed represents means of the duplicate technical repeats plus SDs. dg Kaplan–Meier survival graphs for experimental animals (n = 5) receiving inhibitor mixes (MV or MVN) preincubated with 2.5 × LD50 dose of E. carinatus (47.5 µg, d), B. asper (47 µg, e), B. arietans (54 µg, f) or D. russelii (20 µg, g) venom. h Quantified TAT levels in the envenomed animals from (d) to (g), with data presented as described for (c). Source data provided in Supplementary Data S1.
Fig. 6. The inhibitor combination of marimastat…
Fig. 6. The inhibitor combination of marimastat and varespladib (MV) provides broad preclinical efficacy against venom lethality in an in vivo ‘challenge then treat’ model of envenoming.
Kaplan–Meier survival graphs for experimental animals (n = 5) receiving venom (Ven), followed by delayed drug treatment (15 min later) with a dual combination of marimastat and varespladib. Both venom and treatment were delivered via the intraperitoneal route, and the end of the experiment was at 24 h. Survival of mice receiving: aE. ocellatus (90 µg, 5× i.v. LD50), bE. carinatus (95 µg, 5× i.v. LD50), cB. asper (303 µg, 3× i.p. LD50), dB. arietans (108 µg, 5× i.v. LD50) and eD. russelii (105 µg, 13× i.v. LD50) venom, with and without the inhibitor mix (120 µg of each drug) 15 min later. The drug-only control is presented as a black dashed line at the top of each graph (no toxicity was observed at the given dose). f Quantified thrombin-antithrombin (TAT) levels in the envenomed animals from (a) to (e). Where the time of death was the same within experimental groups (e.g., early deaths or complete survival) TAT levels were quantified for n = 3, and where times of death varied, n = 5. The data displayed represents means of the duplicate technical repeats plus SDs. Source data provided in Supplementary Data S1.

References

    1. Gutiérrez JM, et al. Snakebite envenoming. Nat. Rev. Dis. Prim. 2017;3:17063.
    1. Harrison RA, Casewell NR, Ainsworth SA, Lalloo DG. The time is now: a call for action to translate recent momentum on tackling tropical snakebite into sustained benefit for victims. Trans. R. Soc. Trop. Med. Hyg. 2019;113:835–838.
    1. Williams DJ, et al. Strategy for a globally coordinated response to a priority neglected tropical disease: Snakebite envenoming. PLoS Negl. Trop. Dis. 2019;13:e0007059.
    1. Casewell NR, et al. Medically important differences in snake venom composition are dictated by distinct postgenomic mechanisms. Proc. Natl Acad. Sci. USA. 2014;111:9205–9210.
    1. Tasoulis T, Isbister GK. A review and database of snake venom proteomes. Toxins. 2017;9:290.
    1. Williams DJ, et al. Ending the drought: New strategies for improving the flow of affordable, effective antivenoms in Asia and Africa. J. Proteom. 2011;74:1735–1767.
    1. Arnold C. Vipers, mambas and taipans: the escalating health crisis over snakebites. Nature. 2016;537:26–28.
    1. Gutiérrez JM. Global availability of antivenoms: The relevance of public manufacturing laboratories. Toxins. 2019;11:5.
    1. Casewell NR, et al. Pre-clinical assays predict pan-African Echis viper efficacy for a species-specific antivenom. PLoS Negl. Trop. Dis. 2010;4:e851.
    1. de Silva HA, et al. Low-dose adrenaline, promethazine, and hydrocortisone in the prevention of acute adverse reactions to antivenom following snakebite: A randomised, double-blind, placebo-controlled trial. PLoS Med. 2011;8:e1000435.
    1. Mohapatra B, et al. Snakebite mortality in India: a nationally representative mortality survey. PLoS Negl. Trop. Dis. 2011;5:e1018.
    1. Bulfone TC, Samuel SP, Bickler PE, Lewin MR. Developing small molecule therapeutics for the initial and adjunctive treatment of snakebite. J. Trop. Med. 2018;2018:1–14.
    1. Knudsen C, Laustsen AH. Recent advances in next generation snakebite antivenoms. Trop. Med. Infect. Dis. 2018;3:42.
    1. Habib AG, Gebi UI, Onyemelukwe GC. Snake bite in Nigeria. Afr. J. Med. &. Med. Sci. 2001;30:171–178.
    1. Otero-Patiño R. Epidemiological, clinical and therapeutic aspects of Bothrops asper bites. Toxicon. 2009;54:998–1011.
    1. Kumar KGS, Narayanan S, Udayabhaskaran V, Thulaseedharan NK. Clinical and epidemiologic profile and predictors of outcome of poisonous snake bites – an analysis of 1,500 cases from a tertiary care center in Malabar, North Kerala, India. Int. J. Gen. Med. 2018;11:209–216.
    1. Slagboom J, Kool J, Harrison RA, Casewell NR. Haemotoxic snake venoms: their functional activity, impact on snakebite victims and pharmaceutical promise. Br. J. Haematol. 2017;177:947–959.
    1. Gutiérrez JM, Rucavado A. Snake venom metalloproteinases: their role in the pathogenesis of local tissue damage. Biochimie. 2000;82:841–850.
    1. Gutiérrez JM, Escalante T, Rucavado A, Herrera C. Hemorrhage caused by snake venom metalloproteinases: a journey of discovery and understanding. Toxins (Basel). 2016;8:93.
    1. Ferraz CR, et al. Multifunctional toxins in snake venoms and therapeutic implications: from pain to hemorrhage and necrosis. Front. Ecol. Evol. 2019;7:1–19.
    1. Howes J-M, Theakston RDG, Laing GD. Neutralization of the haemorrhagic activities of viperine snake venoms and venom metalloproteinases using synthetic peptide inhibitors and chelators. Toxicon. 2007;49:734–739.
    1. Lewin M, Samuel S, Merkel J, Bickler P. Varespladib (LY315920) appears to be a potent, broad-spectrum, inhibitor of snake venom phospholipase A2 and a possible pre-referral treatment for envenomation. Toxins. 2016;8:248.
    1. Arias AS, Rucavado A, Gutiérrez JM. Peptidomimetic hydroxamate metalloproteinase inhibitors abrogate local and systemic toxicity induced by Echis ocellatus (saw-scaled) snake venom. Toxicon. 2017;132:40–49.
    1. Rucavado A, et al. Inhibition of local hemorrhage and dermonecrosis induced by Bothrops asper snake venom: effectiveness of early in situ administration of the peptidomimetic metalloproteinase inhibitor batimastat and the chelating agent CaNa2EDTA. Am. J. Trop. Med. Hyg. 2000;63:313–319.
    1. Ainsworth S, et al. The paraspecific neutralisation of snake venom induced coagulopathy by antivenoms. Commun. Biol. 2018;1:34.
    1. Lewin M, et al. Delayed LY333013 (Oral) and LY315920 (Intravenous) reverse severe neurotoxicity and rescue juvenile pigs from lethal doses of Micrurus fulvius (Eastern coral snake) venom. Toxins. 2018;10:479.
    1. Lewin M, et al. Delayed oral LY333013 rescues mice from highly neurotoxic, lethal doses of Papuan taipan (Oxyuranus scutellatus) venom. Toxins. 2018;10:380.
    1. Albulescu L-O, et al. Preclinical validation of a repurposed metal chelator as an early-intervention therapeutic for hemotoxic snakebite. Sci. Trans. Med. 2020;12:eaay8314.
    1. Wang Y, et al. Exploration of the inhibitory potential of varespladib for snakebite envenomation. Molecules. 2018;23:391.
    1. Layfield HJ, et al. Repurposing cancer drugs batimastat and marimastat to inhibit the activity of a group I metalloprotease from the venom of the Western diamondback rattlesnake, Crotalus atrox. Toxins. 2020;12:309.
    1. Rowsell S, et al. Crystal structure of human MMP9 in complex with a reverse hydroxamate inhibitor. J. Mol. Biol. 2002;319:173–181.
    1. Warrell DA, Arnett C. The importance of bites by the saw scaled or carpet viper (Echis carinatus): Epidemiological studies in Nigeria and a review of the world. Acta Trop. 1976;33:307–341.
    1. Warrell, D. in Handbook of Clinical Toxicology of Animal Venoms and Poisons (eds White, J. & Meier, J.) pp 534–594 (CRC Press, 1995).
    1. Warrell, D. in Handbook of Clinical Toxicology of Animal Venoms and Poisons (eds. White, J. & Meier, J.) pp 455–492 (CRC Press, 1995).
    1. Still K, et al. Multipurpose HTS Coagulation Analysis: Assay Development and Assessment of Coagulopathic Snake Venoms. Toxins. 2017;9:382.
    1. Rogalski A, et al. Differential procoagulant effects of saw-scaled viper (Serpentes: Viperidae: Echis) snake venoms on human plasma and the narrow taxonomic ranges of antivenom efficacies. Toxicol. Lett. 2017;280:159–170.
    1. Slagboom J, et al. High throughput screening and identification of coagulopathic snake venom proteins and peptides using nanofractionation and proteomics approaches. PLoS Negl. Trop. Dis. 2020;14:e0007802.
    1. Winer A, Adams S, Mignatti P. Matrix metalloproteinase inhibitors in cancer therapy: turning past failures into future successes. Mol. Cancer Ther. 2018;17:1147–1155.
    1. Kim EY, et al. Low-dose nafamostat mesilate in hemodialysis patients at high bleeding risk. Kidney Res. Clin. Pract. 2011;30:61–66.
    1. Kim HS, et al. Cardiac arrest caused by nafamostat mesilate. Kidney Res. Clin. Pract. 2016;35:187–189.
    1. Theakston RD, Reid HA. Development of simple standard assay procedures for the characterization of snake venom. Bull. World Health Organ. 1983;61:949–956.
    1. Harrison RA, et al. Preclinical antivenom-efficacy testing reveals potentially disturbing deficiencies of snakebite treatment capability in East Africa. PLoS Negl. Trop. Dis. 2017;11:e0005969.
    1. WHO, WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins (WHO, (2018).
    1. Bolaños R. Toxicity of Costa Rican snake venoms for the white mouse. Am. J. Trop. Med. Hyg. 1972;21:360–363.
    1. Villalta M, et al. Development of a new polyspecific antivenom for snakebite envenoming in Sri Lanka: Analysis of its preclinical efficacy as compared to a currently available antivenom. Toxicon. 2016;122:152–159.
    1. Mora-Obando D, et al. Proteomic and functional profiling of the venom of Bothrops ayerbei from Cauca, Colombia, reveals striking interspecific variation with Bothrops asper venom. J. Proteom. 2014;96:159–172.
    1. Harrison RA, Gutiérrez JM. Priority actions and progress to substantially and sustainably reduce the mortality, morbidity and socioeconomic burden of tropical snakebite. Toxins. 2016;8:351.
    1. de la Rosa G, et al. Horse immunization with short-chain consensus α-neurotoxin generates antibodies against broad spectrum of elapid venomous species. Nat. Commun. 2019;10:3642.
    1. Kini RM, Sidhu SS, Laustsen AH. Biosynthetic oligoclonal antivenom (BOA) for snakebite and next-generation treatments for snakebite victims. Toxins. 2018;10:534.
    1. Laustsen AH, et al. In vivo neutralization of dendrotoxin-mediated neurotoxicity of black mamba venom by oligoclonal human IgG antibodies. Nat. Commun. 2018;9:3928.
    1. Peterson J. The importance of estimating the therapeutic index in the development of matrix metalloproteinase inhibitors. Cardiovasc. Res. 2006;69:677–687.
    1. Millar AW, et al. Results of single and repeat dose studies of the oral matrix metalloproteinase inhibitor marimastat in healthy male volunteers. Br. J. Clin. Pharmacol. 1998;45:21–26.
    1. Rosemurgy A, et al. Marimastat in patients with advanced pancreatic cancer: a dose-finding study. Am. J. Clin. Oncol. 1999;22:247–252.
    1. Nair A, Jacob S. A simple practice guide for dose conversion between animals and human. J. Basic Clin. Pharm. 2016;7:27.
    1. Adis R&D Profile Varespladib. Am. J. Cardiovasc. Drugs. 2011;11:137–143.
    1. Rosenson RS, et al. Effects of varespladib methyl on biomarkers and major cardiovascular events in acute coronary syndrome patients. J. Am. Coll. Cardiol. 2010;56:1079–1088.
    1. Abraham E, et al. Efficacy and safety of LY315920Na/S-5920, a selective inhibitor of 14-kDa group IIA secretory phospholipase A2, in patients with suspected sepsis and organ failure. Crit. Care Med. 2003;31:718–728.
    1. Nicholls SJ, et al. Varespladib and cardiovascular events in patients with an acute coronary syndrome: The VISTA-16 randomized clinical trial. JAMA - J. Am. Med. Assoc. 2014;311:252–262.
    1. Gutiérrez JM, Lewin MR, Williams DJ, Lomonte B. Varespladib (LY315920) and methyl varespladib (LY333013) abrogate or delay lethality induced by presynaptically acting neurotoxic snake venoms. Toxins. 2020;12:131.
    1. Ohtake Y, et al. Nafamostat mesylate as anticoagulant in continuous hemofiltration and continuous hemodiafiltration. Contrib. Nephrol. 1991;93:215–217.
    1. Maiorino RM, Xu ZF, Aposhian HV. Determination and metabolism of dithiol chelating agents. XVII. In humans, sodium 2,3-dimercapto-1-propanesulfonate is bound to plasma albumin via mixed disulfide formation and is found in the urine as cyclic polymeric disulfides. J. Pharmacol. Exp. Ther. 1996;277:375–384.
    1. Kosnett MJ. The role of chelation in the treatment of arsenic and mercury poisoning. J. Med. Toxicol. 2013;9:347–354.
    1. Wagstaff SC, Sanz L, Juárez P, Harrison RA, Calvete JJ. Combined snake venomics and venom gland transcriptomic analysis of the ocellated carpet viper, Echis ocellatus. J. Proteom. 2009;71:609–623.
    1. Tan NH, et al. Functional venomics of the Sri Lankan Russell’s viper (Daboia russelii) and its toxinological correlations. J. Proteom. 2015;128:403–423.
    1. Pla D, et al. Phylovenomics of Daboia russelii across the Indian subcontinent. Bioactivities and comparative in vivo neutralization and in vitro third-generation antivenomics of antivenoms against venoms from India, Bangladesh and Sri Lanka. J. Proteom. 2019;207:103443.
    1. Bradley JD, et al. A randomized, double-blinded, placebo-controlled clinical trial of LY333013, a selective inhibitor of group II secretory phospholipase A2, in the treatment of rheumatoid arthritis. J. Rheumatol. 2005;32:417–423.
    1. Sevenet PO, Depasse F. Clot waveform analysis: Where do we stand in 2017? Int. J. Lab. Hematol. 2017;39:561–568.
    1. Patra A, Kalita B, Chanda A, Mukherjee AK. Proteomics and antivenomics of Echis carinatus carinatus venom: Correlation with pharmacological properties and pathophysiology of envenomation. Sci. Rep. 2017;7:17119.
    1. Alape-Girón A, et al. Studies on the venom proteome of Bothrops asper: perspectives and applications. Toxicon. 2009;54:938–948.
    1. Calvete JJ, Escolano J, Sanz L. Snake venomics of Bitis species reveals large intragenus venom toxin composition variation: application to taxonomy of congeneric taxa. J. Proteome Res. 2007;6:2732–2745.

Source: PubMed

3
Iratkozz fel