You are on page 1of 8

Biochemical Pharmacology xxx (xxxx) xxxx

Contents lists available at ScienceDirect

Biochemical Pharmacology
journal homepage: www.elsevier.com/locate/biochempharm

Review

Snake antivenom: Challenges and alternate approaches


Aswathy Alangode, Karthika Rajan, Bipin G. Nair

School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana P.O, Kollam, Kerala 690 525, India

ARTICLE INFO ABSTRACT

Keywords: Snake envenomation is still a serious threat to many countries in the world. The only mainstay treatment de-
Antivenom pends on the administration of animal derived immunoglobulin based antivenom. Significant limitations to these
Snake venom antivenoms are a challenge in the treatment of snake envenomation. Many alternate approaches have been
Peptide inhibitors explored to overcome the limitations of antivenom. Exploring alternate approaches like use of bioactive com-
Phage display
ponents from plant sources, use of peptide and small molecule inhibitors are some aspects taken towards im-
proving the current limitations of antivenom therapy. However, all these alternate approaches also have many
drawbacks which should be improved by more in vitro and in vivo experiments. Here, we review some of the
limitations of current antivenom therapy and developments as well as drawbacks of these alternate treatment
strategies.

1. Introduction to the action of different enzymatic and non-enzymatic toxins present in


the venom. Snake venom is composed of both enzymatic toxins like
Snake envenomation is a serious threat to many developed and snake venom metalloproteases (SVMPs), PLA2, L-amino acid oxidases
developing countries. Injection of venomous toxins from snakes to the (LAAO), serine proteases and non-enzymatic components including
victim evokes a chain of reactions which unless treated will pose a disintegrins, three finger toxins, protease inhibitors and many more
threat to the life of the victim. Worldwide, there are approximately [5,6]. These toxins affect different physiological systems of the victims
81,000 to 138,000 deaths (Fig. 1) per year due to snake envenomation thereby leading to death (Fig. 2). In addition, morbidity due to en-
[1–3]. Major areas affected by snakebite include South Asia, with India venomation can cause many physical defects like amputation, necrosis
having a statistics of 50,000 deaths and 85,000 snakebites per year [4]. and severe local damage [7].
Significant morbidity and mortality due to envenomation is attributed


Corresponding author.
E-mail address: bipin@am.amrita.edu (B.G. Nair).

https://doi.org/10.1016/j.bcp.2020.114135
Received 30 April 2020; Received in revised form 25 June 2020; Accepted 1 July 2020
0006-2952/ © 2020 Elsevier Inc. All rights reserved.

Please cite this article as: Aswathy Alangode, Karthika Rajan and Bipin G. Nair, Biochemical Pharmacology,
https://doi.org/10.1016/j.bcp.2020.114135
A. Alangode, et al. Biochemical Pharmacology xxx (xxxx) xxxx

polyclonal (raised against the venom of different species of snakes) or


Nomenclature
monoclonal (raised against venom of one snake species) and the venom
batches vary significantly between younger and older snakes, geo-
SVMP Snake venom metalloproteainases
graphical area, sex and age of snakes [10]. This variability in venom
PLA2 phospholipase A2
toxicity and activity is of relevance because it leads to batch variability
LAAO L-amino acid oxidases LPS – lipopolysaccharide
in the antivenom. Furthermore, adverse reactions triggered by animal
VICC venom induced consumption coagulopathy
proteins, such as polyvalent horse antibodies, is a major problem of
LMW low molecular weight
antivenom therapy (Table 1) [11,12] which requires time consuming
allergy testing prior to administration of antivenom therapy, in a si-
tuation where time is of concern [13]. Despite these limitations, the
Current treatment strategy for snakebite is the administration of
current treatment available in the market owes for conventional anti-
antivenom, which are antibodies obtained after immunising animals
venoms that can neutralize the toxins [8,14].
like horses with sub lethal doses of venom [8,9]. Antivenom can be

Fig. 1. Estimated number of worldwide deaths and snake envenoming. Total global estimated incidence rate is 1.8–2.7 million bites with 81,000–138,000 deaths
annually. Major incidence of snakebite estimated is in south and south-east Asia followed by Africa and South America whereas envenomation is lower in Europe and
North America. Adapted from J.M. Gutierrez et al., 2017. Ref. [3].

2
A. Alangode, et al. Biochemical Pharmacology xxx (xxxx) xxxx

Fig. 2. Diagram showing the physiological effects of different snake venom toxins. Snake venoms exert wide range of pharmacological activities on the physiological
system and cause various effects depending on the toxins present in the venom like snake venom metalloproteases (SVMPs), snake venom serine proteases (SVSPs),
phospholipase A2 (PLA2), three finger toxins (3FTX), disintegrins and many more. Viperid and elapids affect different proteins of the coagulation cascade leading to
consumption coagulopathy and can also induce neuromuscular paralysis. Systemic haemorrhage leading to cardiovascular shock is the major effect caused by
viperids. Other effects of envenomations include myotoxicity, local tissue damage and acute kidney injury. Adapted from J.M. Gutierrez et al., 2017. Ref. [3].

Table 1
Adverse reactions of antivenom.
Adverse reactions of antivenom

1. Early IgE mediated anaphylactic reactions 1. Type 1 hypersensitivity reaction [20].


2. IgE is produced against antivenom components due to previous exposure to animal immunoglobulins [16].
3. Mechanism involves mast cell degranulation and release of histamine, prostaglandin and leukotrienes [20].
4. Results in increased vascular permeability, vasodilation, inflammation, smooth muscle contraction, mucuos secretion, local
inflammation, anaphylactic shock and edema [16,20].
Non-IgE mediated anaphylactic 1. Occur in patients who have not been previously exposed to antivenom components [16].
reactions 2. Presence of immunoglobulin aggregates against blood cells in antivenom [20].
3. Complement activation by aggregates results in mast cell degranulation [16].
4. Results in increased vascular permeability, vasodilation, inflammation, rash, urticaria and pain [20].
Pyrogenic 1. Induced by bacterial endotoxins present in antivenom [16].
2. Mechanism involves activation of macrophages by endotoxin and results in production of cytokines such as TNF-alpha, IL-
1β,IL-6 [20].
3. Results in fever, myalgia, headache, rigors, nausea, sweating, chills, increase in heart rate and vasodilation [11]
2. Late Antigen-Antibody mediated 1. Type III hypersensitivity reaction [17].
2. Also called as serum sickness [17].
3. Mechanism involves complement system activation by immune-complexes and leukocyte infiltration [20].
4. Results in mast cell degranulation and release of histamine, prostaglandin, serotonin and leukotrienes [20].
5. Results in fever, myalgia, arthralgia, utricaria, lymphadenopathy and gastrointestinal disorders [16].
6. Rarely develops rash, glomerulonephritis, laryngeal edema, and vasculitis [17]

3
A. Alangode, et al. Biochemical Pharmacology xxx (xxxx) xxxx

Adverse reactions due to antivenom are classified by WHO as - early antivenom [23]. Hence, adrenaline is the treatment of choice for ad-
(anaphylactic reactions and pyrogenic reactions) which occur within 24 verse reactions of antivenom.
hrs of antivenom administration and late reactions (serum sickness) In addition to these adverse reactions, antivenoms do not always be
which develop between 5 and 24 days of treatment [15]. Anaphylactic effective against various complications related to snakebite. For in-
reactions occur within the first hour of antivenom administration to the stance, envenomation due to viper and elapid bite leads to venom in-
patient and are due to foreign nature of the immunoglobulins in anti- duced consumption coagulopathy (VICC) which is the major outcome of
venom. Pyrogens are substances that induce fever. These substances can envenomation by these species [24,25] and administration of anti-
most likely be due to the presence of bacterial components in the an- venom may not be effective in treating VICC since it is irreversible
tivenom preparation. Due to the presence of bacterial components like [26–28]. This could be due to the rapid absorption of venom resulting
LPS, pyrogenic reactions occur within an hour after antivenom ad- in faster onset of VICC [29]. Whereas studies have shown that some
ministration. Pyrogenic reactions are usually characterized by fever, antivenoms are effective against VICC induced by Echis envenomations
chills, headache, nausea, sweating and increase in heart rate [16]. In [30] however other studies show it is less effective against coagulo-
the case of late adverse reactions, a delayed immune response is in- pathy and displays less cross reactivity towards by Echis envenomations
duced where there is an increase in IgG mediated antibody response. [31]. For an effective evaluation of these effects, randomized placebo
This corresponds to type III hypersensitivity reaction due to the pre- controlled trials are required which is challenging and considered as
sence of circulating antigen-antibody or immune complexes. Moreover, unethical [32,33]. In addition, many studies have shown that anti-
this leads to complement activation and triggers inflammatory reaction venoms show cross reactivity against many species but some variations
due to the presence of these complexes in various tissues, joints and in specificity and efficacy can be observed [34–36]. Such variations in
lymph nodes. Clinical manifestations include rash, fever, malaise, ar- efficacy could be due to minor changes in the structure or surface
thralgia and arthritis [17]. Despite these adverse reactions antivenom is chemistry of the toxins.
still the current mainstay treatment for snakebite. Furthermore, there are situations where all the venom components
Adverse reactions to antivenom are commonly treated by adminis- are not targeted by the antivenom. For instance, many studies including
tration of adrenaline [18], antihistamine and hydrocortisone before the data from our lab [37] have shown that there are antivenoms which do
administration of antivenom [19]. The drugs that reduce these adverse not recognize the low molecular weight (LMW) protein components of
reactions are reviewed in detail elsewhere [20], but its efficacy were the venom compared to the higher molecular weight toxins which play
questioned [21]. In snake envenomation involving coagulopathy, the significant role in envenomation [38–40]. Also, differential binding of
administration of adrenaline may increase the risk of intracerebral antivenom to toxins present in different species of snakes are observed.
hemorrhgae [22]. In contrast, a report by Tibballs mentions that pa- Studies have shown that in some species, fibrinogen cleavage was due
tients had very low intra cerebral hemorrhage after snakebite irre- to the action of venom serine proteases and in other species by me-
spective of whether adrenaline was administered or not [18]. But this talloproteases. Hence, antivenom generated against species with serine
study involved a limited number of patients and hence not conclusive. protease enzymes may not effectively neutralize the metalloprotease
Whereas another study by De silva et al. demonstrated that low dose of species [41].
subcutaneous adrenaline were safe and limit the risk of adverse reac- To overcome these limitations of antivenom therapy (Table 2), there
tions by 43% in 1hr and 38% upto 48 hr after antivenom infusion, is an urgent need to develop new approaches which will serve the
however promethazine and hydrocortisone did not have any effect at 1 function of antivenom and will provide minimum adverse reactions. A
or 48 h [19]. Moreover, a meta-analysis conducted based on three more detailed understanding about current antivenom therapies and
randomized and four non-randomized control studies with pre-medi- their safety as well as effectiveness have been covered elsewhere
cation and control groups found that adrenaline pre-medication might [42–44].
have a beneficial effect against early adverse reactions (EAR) to

Table 2
Limitations of antivenom.
Type of antivenom Drawbacks

Polyvalent antivenom animal derived 1. Batch variability [44].


2. Adverse reactions like severe hypersensitivity reactions occur due to immune response generated against proteins in the
serum of immunized animals [11,16,20].
3. Less effective against venom-induced local effects [44].
4. Containing large proportion of irrelevant antibodies
compared to toxin neutralising antibodies [40,44].
5. Complex manufacturing processes that depend on two biological systems (snake and the immunized animal) [44].
6. Antibody molecule has poor distribution to peripheral sites, but a longer elimination half-life [44,99].
7. Not effective in neutralising irreversible symptoms like VICC, pre-synaptic neurotoxicity, cardiovascular collapse and local
necrosis [44].
8. Geographical variation owing to venom collected from different geographical regions, diet and age of snakes does show
variation in venom composition thereby leads to variation in antivenom [10].
Monoclonal 1. Have all limitations of polyclonal antivenom.
2. Targets single toxins (monoclonal), and are not able to neutralize the large number of different toxins in snake venom
[44].
Recombinant antibody fragments (including Fab 1. They have a shorter serum half-life in vivo [44].
and Fab')2

4
A. Alangode, et al. Biochemical Pharmacology xxx (xxxx) xxxx

isolated from plant sources are not validated and time spent in devel-
oping and purifying these components is a drawback [48].

2.2. Peptide inhibitors

The concept of using peptides specifically to bind and neutralize


major toxins present in snake venom is a significant thought towards
achieving a peptide based antivenom therapy. Use of peptide inhibitors
seems to be advantageous as this could be specific to toxin components
present in venom [51]. Studies published shows two classes of in-
hibitors, where one class belongs to endogenous inhibitors which are
inhibitors found in the blood circulation of venomous snakes [43,52].
These inhibitors provides resistance to their own venoms [53]. Most of
the Viperidae family of snakes has metalloproteinases and phospholi-
pase A2 enzymes in their venom in abundance. SVMPs are proteolytic
enzymes that cause local tissue damage and haemorrhage at bite site
[54,55]. PLA2 are responsible for inflammation and severe pain during
envenomation [53,56]. Peptide Inhibitors that can bind with high af-
finity to these toxins and effectively neutralize the activity of these
enzymes can be of much advantage.
Searching for mechanisms involved in natural resistance shown by
snake venoms is a promising lead. Several endogenous phospholipase
and metalloprotease inhibitors are secreted for self-protection of the
snake from its own venom [57–61]. Characterization of these natural
inhibitor mechanisms through proteomics and transcriptomic studies
can provide a deeper understanding of their interactions with the
venom components [62].
Exploring the immunity to snakebite shown by certain animals like
Fig. 3. Schematic representation of different steps involved in peptide phage the opossum forms another class of inhibitors based on the natural
display technology. A synthetic random peptide phage display libraries are predators of snakes. Work published in this regard by Rocha et al.,
screened against toxin components of snake venom for binding. These bound found that certain proteins isolated from the serum of opossum species
peptides are selected and enriched through multiple panning steps. Enriched could neutralize venom from certain snakes [63]. A short peptide called
peptides are expressed in bacterial host cells. The recombinant peptide obtained LTNF peptide (Lethal Toxin Neutralizing Factor) has been purified from
is optimized and used in assays to confirm peptide-venom binding and neu- the sera of Didelphis virginiana which inhibited lethality of venom from
tralization. Adapted from E.C. Roncolato et al., 2015. Ref. [96]. Crotalidae, Elapidae, Viperidae and Hydrophiidae [64]. Further studies
lead by Komives et al., has expressed these peptides in E. coli as re-
Although this review does not exhaustively discuss all the chal- combinant peptides and were able to completely neutralize C. atrox and
lenges related to antivenom and alternate treatment modalities for Russell's viper venom [65]. Such recombinant expression in lower
snake envenomation, we hope to offer few important aspects related to prokaryotes yields much cheaper and good yield of peptides that can be
the advantages and drawbacks of current treatments and the ad- further used in several studies to inhibit snake venom toxins. Never-
vantages of developing peptide inhibitors as an alternate to antivenom theless, optimization of expression parameters for maximum yield is
therapy and small molecule inhibitors for complications related to one of the limitations regarding such recombinant proteins.
snake envenomation.
2.3. Small molecule inhibitors

2. Need for a better antivenom and alternate therapies 2.3.1. PLA2 inhibitor
As discussed above, low immunogenicity of certain small toxins in
2.1. Natural products the venom like svPLA2 could be overcome by using small molecule
inhibitors. In this regard, use of varespladib is highly recommended by
Many studies have been done to evaluate the effects of natural many researchers. Varespladib is a potent inhibitor of secretory PLA2
products for snake envenomation. Traditional medicines derived from and was used to treat acute coronary syndrome [66]. Since svPLA2 is a
plant sources could provide a promising lead towards venom neu- major toxin in the venoms of many snake species, studies showing the
tralization [45]. As India is rich in biodiversity with varied plant species use of varespladib to inhibit svPLA2 toxicity have been shown. For in-
of which medicinally important plants can serve to be good candidate stance, M. fulvius due to the presence of PLA2, manifest neuromuscular
for effective snakebite treatment [46]. Literature surveys have shown paralysis, intravascular hemolysis and myonecrosis in animals [67–69].
that several active natural products obtained from plant species like Lewin et al. demonstated that varespladib and orally bioavailable me-
Aristolochia indica [47,48], Withania somnifera [37,49], Azadirachta in- thyl varespladib rescued pigs that were experimentally injected with
dica extract [50] have been successful in reducing inflammation, local the M. fulvius venom [70]. Another study shows that the IC50 of var-
tissue damage and severe pain at the bite site. The active components espladib and methyl varespladib for Russell’s viper venom is 0.0006
present in these plant species acts on particular enzymes present in and 0.02uM respectively. Both showed high inhibitory activity (low
venom and neutralize their effects. One such enzyme is snake venom IC50) towards svPLA2 from 28 different snakes from 6 continents [71].
phospholipase A2. Similarly, varespladib was able to inhibit the PLA2 induced antic-
Several databases are published which contains bioactive molecules oagulant activity of African spitting cobras [72]. Haemorrhagic toxicity
derived from natural products that show activity against snake venom of D. acutus and G. Halys were significantly inhibited by varespladib
components [48]. Even though a lot of research is successful in isolating [73]. Gutierrez showed that varespladib inhibits myotoxicity and cy-
new bioactive compounds, the toxicity profile of the active components totoxicity exhibited by the PLA2, MjTX II, present in venom of Bothrops

5
A. Alangode, et al. Biochemical Pharmacology xxx (xxxx) xxxx

moojeni venom using in vivo and in vitro experiments in mice, respec- synthesized chemically and used in neutralization assays to check for
tively. Both structural and functional studies of the complex formed by venom enzyme neutralization. This involves selection of a particular
the venom and the inhibitor have been studied [74]. Mice injected with venom component or target against which we need to develop peptide
O. scutellatus, B. multicinctus, and C. d. terrificus venom were able to inhibitors, then fractionation of the crude venom to obtain our com-
inhibit the neurotic effects by these venom in the presence of var- ponent of interest, and a method to detect the target and finally purify it
espladib and its orally available drug methyl varespladib [75]. In ad- to use it as bait for phage display (Fig. 3).
dition, a study by Zdenek et al. found that coagulotoxic effects of 9 As compared against recently available phage antibody library,
pseudechis species from Australia, due to PLA2 toxins (except P. por- which invokes several disadvantages like the size of the antibody or its
phyriacus) inhibitory activity towards FVa and or prothrombinase fragments, low immunogenicity of fragments, cost of development,
complex were not neutralised by both BSAV and TSAV, but varespladib stability issues [51,88] and less bioavailability, short peptides with
exhibited potent inhibitory activity towards these venoms [76]. A more modified structural properties as lead molecules can be of great ad-
comprehensive review has been published on drug development against vantage. A major concern is the cost of synthesizing the lead peptides
svPLA2 for snake envenomation [77]. that needs to be addressed. One way it can be surpassed is by producing
Eventhough varespladib has shown inhibitory effect on PLA2 of it in larger yields in E. coli systems. But more studies are required for
many snake species, it is still not approved by FDA [70]. More studies recombinant peptide expression in E. coli cells.
with timing of administration and different doses of venom in animal
models are required. Varespladib alone or in combination with other
inhibitors should be tested in animal models for effectiveness against 4. Combinatorial therapy
important snake venoms.
Eventhough, antivenom is the main treatment for systemic effects of
2.3.2. SVMP inhibitors envenomation it is ineffective in treating local damage due to en-
Venom metalloproteases are abundant in many viperid venoms and venomation [98,99]. A combination of antivenom along with inhibitors
induce various effects such as hemmorhage, and local damages in- to different toxins causing local tissue damage may be effective against
cluding necrosis and edema [78]. Bothrops asper is a pit viper which is completely inhibiting the effects of envenomation. In this regard, work
prevalent in Central America and southern Mexico and induce hem- done by Howes et al. shows that matrix metalloprotease inhibitor
morhage, myonecrosis and edema [79–82]. Often these local damages AG3340 inhibits the hemorrhagic activity of whole E. ocelatus venom
cannot be prevented by the administration of antivenom. In this regard, [86]. But more studies are required for evaluating the efficacy of drugs
effect of the MMPIs batimastat and marimastat have been studied. combined with antivenom, its dosages and route of administration.
Batimastat has been shown to inhibit local haemorrhage, dermonecrosis
and hemorrhagic effect in lungs due to Bothrops asper envenomation 5. Conclusions
[83–85]. Similarly, the effect of matrix metalloproteases inhibitors
(MMPI) marimastat on SVMPs has also been studied [86]. When in- Current antivenom therapy poses many disadvantages like lack of
cubated with venom, both inhibited the lethal and hemorrhagic effects specificity, hypersensitivity or adverse reactions to animal proteins in-
induced by E. ocellatus venom [87]. volved in antivenom production, owing to which alternative therapy for
These studies show that small molecule inhibitors are effective snake envenomation is of great importance. Exploring varied variety of
against local damages caused by the action of different toxins during available natural products is one among them, wherein bioactive mo-
envenomation. These small molecule inhibitors could thus improve the lecules from plant extracts are used for treating snakebites or to neu-
treatment strategy by their administration even before the adminis- tralize venom components. Lack of knowledge about active components
tration of antivenom to minimise the effects of local tissue damages of the extracts and effective analysis procedures for active components
which are not usually inhibited by antivenom. are its drawbacks. Another area focuses on developing antibody library
by phage display technique and modifying existing limitations of an-
3. Phage display library of peptides and antibodies tibody derived phage library by introducing monoclonal antibodies that
can cross react with many snake venom proteins. Major concerns about
Research studies report the use of phage display technique in de- antibody library including the high cost of production, low half -life
veloping short peptides that are known to bind snake venom toxins. stability, low immunogenicity along with developmental issues stand as
Snake venom contains a variety of peptide components that can be a hindrance. In this regard, short peptides that can be generated against
isolated and purified [3]. Newer in vitro selection of antibody for use as specific venom toxins can be advantageous. Several known peptide
antivenom was developed using antibody phage display technology by inhibitors are identified against snake venom components but to pro-
applying the filamentous fusion phage [88,89]. duce them in a way that is less expensive and less time consuming is
An antibody phage display library development involves fusing the important. Use of peptide phage display library will be a significant
single chain variable fragments (ScFv's) of antibody domain with the thought in that direction. Furthermore, the peptides that show affinity
phage coat protein gene III and thus the phage coat protein display towards venom proteins can be amplified and enriched in E. coli cells
these domains on the surface of phage. When an appropriate antigen thereby achieving a good yield for further studies. Optimization of
binding site is screened against this antibody library, high affinity ones expression conditions and host bacteria environments can further lead
can be selected and enriched [51,90]. Besides these conventional an- to optimum production in a smaller time frame and cheap production
tibody libraries, antibody phage display with multivalent and multi- cost. Similarly, the use of small molecule inhibitors have shown to be
specific constructs has also been developed [91]. Further studies have effective against local damages due to envenomation. Developing drugs
developed monoclonal antibody fragments specific to more than one using small molecule inhibitors could be of great advantage in the
toxin in snake venom, thereby providing one monoclonal antibody treatment of snake envenomation. In this regard, multidisciplinary
targeted against two or more venom toxins [92–94]. A common pro- approaches are required to improve current antivenom therapy.
blem related to antibodies are it's half-life and stability issues which
leads to dosage irregularities, as the required dose may not reach the
target and thus gets eliminated in the renal system [95,96]. In this re- CRediT authorship contribution statement
gard, a phage display library which contains random synthetic peptides
incorporated into the phage coat proteins, is used to screen for binding Aswathy Alangode: Writing - original draft. Karthika Rajan:
partners against snake venom toxins [97]. These peptides are then Writing - original draft. Bipin G. Nair: Writing - review & editing.

6
A. Alangode, et al. Biochemical Pharmacology xxx (xxxx) xxxx

Acknowledgements [28] P.P. Tanos, G.K. Isbister, D.G. Lalloo, C.M.J. Kirkpatrick, S.B. Duffull, A model for
venom-induced consumptive coagulopathy in snake bite, Toxicon 52 (2008)
769–780.
We acknowledge Mata Amritanandamayi Devi, Chancellor, Amrita [29] A. Gulati, G.K. Isbister, S.B. Duffull, Effect of Australian elapid venoms on blood
Vishwa Vidyapeetham for being the inspiration behind the work. We coagulation: Australian Snakebite Project (ASP-17), Toxicon 61 (2013) 94–104.
would like to thank Dr. Jyotsna Nambiar for her help with the manu- [30] G. Mion, S. Larréché, A. Benois, F. Petitjeans, M. Puidupin, Hemostasis dynamics
during coagulopathy resulting from Echis envenomation, Toxicon 76 (2013)
script preparation. Ms. Aswathy Alangode was supported by 103–109.
Department of Biotechnology, Govt of India (DBT) for JRF/SRF fel- [31] A. Rogalski, C. Soerensen, B. op den Brouw, C. Lister, D. Dashvesky, K. Arbuckle, A.
lowship DBT-JRF/2010-11/119 and Ms. Karthika Rajan is supported by Gloria, C.N. Zdenek, N.R. Casewell, J.M. Gutiérrez, W. Wüster, S.A. Ali, P. Masci, P.
Rowley, N. Frank, B.G. Fry, Differential procoagulant effects of saw-scaled viper
JRF/SRF fellowship 19/06/2016(i)EU-V from University Grants (Serpentes: Viperidae: Echis) snake venoms on human plasma and the narrow
Commission. The authors are also thankful to the Department of taxonomic ranges of antivenom efficacies, Toxicol. Lett. 280 (2017) 159–170.
Science and Technology (DST), India for their research grant SR/SO/ [32] K. Maduwage, N.A. Buckley, H.J. de Silva, D.G. Lalloo, G. Isbister, Snake antivenom
for snake venom induced consumption coagulopathy, Cochrane Database Syst. Rev.
BB-29/2010. The work was also supported by Institutional funding
2014 (2014).
(Amrita University Research) grant (BGN). [33] C.J. Gerardo, E.J. Lavonas, R.E. McKinney, Ethical considerations in design of a
study to evaluate a US Food and Drug Administration-approved indication: anti-
References venom versus placebo for copperhead envenomation, Clin. Trials 11 (2014)
560–564.
[34] C. Lister, K. Arbuckle, T.N.W. Jackson, J. Debono, C.N. Zdenek, D. Dashevsky,
[1] J.P. Chippaux, Snake-bites: appraisal of the global situation, Bull. World Health N. Dunstan, L. Allen, C. Hay, B. Bush, A. Gillett, B.G. Fry, Catch a tiger snake by its
Organ. 76 (1998) 515–524. tail: differential toxicity, co-factor dependence and antivenom efficacy in a pro-
[2] A. Kasturiratne, A.R. Wickremasinghe, N. De Silva, N.K. Gunawardena, coagulant clade of Australian venomous snakes, Comp. Biochem. Physiol. Part - C
A. Pathmeswaran, R. Premaratna, L. Savioli, D.G. Lalloo, H.J. De Silva, The global Toxicol. Pharmacol. 202 (2017) 39–54.
burden of snakebite: a literature analysis and modelling based on regional estimates [35] C.N. Zdenek, B. op den Brouw, D. Dashevsky, A. Gloria, N.J. Youngman, E. Watson,
of envenoming and deaths, PLoS Med. 5 (2008) 1591–1604. P. Green, C. Hay, N. Dunstan, L. Allen, B.G. Fry, Clinical implications of convergent
[3] J.M. Gutiérrez, J.J. Calvete, A.G. Habib, R.A. Harrison, D.J. Williams, D.A. Warrell, procoagulant toxicity and differential antivenom efficacy in Australian elapid snake
Snakebite envenoming, Nat. Rev. Dis. Prim. (2017). venoms, Toxicol. Lett. 316 (2019) 171–182.
[4] J.M. Gutiérrez, Understanding and confronting snakebite envenoming: the harvest [36] C.N. Zdenek, C. Hay, K. Arbuckle, T.N.W. Jackson, M.H.A. Bos, B. op den Brouw, J.
of cooperation, Toxicon 109 (2016) 51–62. Debono, L. Allen, N. Dunstan, T. Morley, M. Herrera, J.M. Gutiérrez, D.J. Williams,
[5] T.S. Kang, D. Georgieva, N. Genov, M.T. Murakami, M. Sinha, R.P. Kumar, P. Kaur, B.G. Fry, Coagulotoxic effects by brown snake (Pseudonaja) and taipan (Oxyuranus)
S. Kumar, S. Dey, S. Sharma, A. Vrielink, C. Betzel, S. Takeda, R.K. Arni, T.P. Singh, venoms, and the efficacy of a new antivenom, Toxicol. Vitr. 58 (2019) 97–109.
R.M. Kini, Enzymatic toxins from snake venom: structural characterization and [37] M. Vanuopadath, S.K. Shaji, D. Raveendran, B.G. Nair, S.S. Nair, Delineating the
mechanism of catalysis, FEBS J. 278 (2011) 4544–4576. venom toxin arsenal of Malabar pit viper (Trimeresurus malabaricus) from the
[6] R.J.R. McCleary, R.M. Kini, Non-enzymatic proteins from snake venoms: a gold Western Ghats of India and evaluating its immunological cross-reactivity and in
mine of pharmacological tools and drug leads, Toxicon 62 (2013) 56–74. vitro cytotoxicity, Int. J. Biol. Macromol. 148 (2020) 1029–1045.
[7] S. Waiddyanatha, A. Silva, S. Siribaddana, G.K. Isbister, Long-term effects of snake [38] R.R. Senji Laxme, S. Khochare, H.F. de Souza, B. Ahuja, V. Suranse, G. Martin, R.
envenoming, Toxins (Basel) 11 (2019) 1–13. Whitaker, K. Sunagar, Beyond the ‘Big four’: Venom profiling of the medically
[8] E. Bermúdez-Méndez, A. Fuglsang-Madsen, S. Føns, B. Lomonte, J.M. Gutiérrez, important yet neglected Indian snakes reveals disturbing antivenom deficiencies,
A.H. Laustsen, Innovative immunization strategies for antivenom development, 2019.
Toxins (Basel) 10 (2018) 1–37. [39] J. Antúnez, J. Fernández, B. Lomonte, Y. Angulo, L. Sanz, A. Pérez, J.J. Calvete,
[9] WHO, Guidelines for the Production, Control and Regulation of Snake Antivenom J.M. Gutiérrez, Antivenomics of Atropoides mexicanus and Atropoides picadoi
Immunoglobulins, World Heal. Organ. (2016) 1–138. snake venoms: relationship to the neutralization of toxic and enzymatic activities, J.
[10] J.P. Chippaux, V. Williams, J. White, Snake venom variability: methods of study Venom Res. 1 (2010) 8–17.
results and interpretation, Toxicon 29 (1991) 1279–1303. [40] J.M. Gutiérrez, L. Sanz, M. Flores-Díaz, L. Figueroa, M. Madrigal, M. Herrera,
[11] H.A. De Silva, N.M. Ryan, H.J. De Silva, Adverse reactions to snake antivenom, and M. Villalta, G. León, R. Estrada, A. Borges, A. Alape-Girón, J.J. Calvete, Impact of
their prevention and treatment, Br. J. Clin. Pharmacol. 81 (2016) 446–452. regional variation in Bothrops asper snake venom on the design of antivenoms:
[12] H.R. Hoogenboom, Selecting and screening recombinant antibody libraries, Nat. integrating antivenomics and neutralization approaches, J. Proteome Res. 9 (2010)
Biotechnol. 23 (2005) 1105–1116. 564–577.
[13] K. Heard, G.F. O’Malley, R.C. Dart, Antivenom therapy in the Americas, Drugs 58 [41] J. Debono, M.H.A. Bos, N. Frank, B. Fry, Clinical implications of differential anti-
(1999) 5–15. venom efficacy in neutralising coagulotoxicity produced by venoms from species
[14] J.P. Chippaux, The development and use of immunotherapy in Africa, Toxicon 36 within the arboreal viperid snake genus Trimeresurus, Toxicol. Lett. 316 (2019)
(1998) 1503–1506. 35–48.
[15] World Health Organization, Progress in the characterization of venoms and stan- [42] D.J. Williams, A.G. Habib, D.A. Warrell, Clinical studies of the effectiveness and
dardization of antivenoms, 1981. safety of antivenoms, Toxicon. 150 (2018) 1–10.
[16] G. León, M. Herrera, Á. Segura, M. Villalta, M. Vargas, J.M. Gutiérrez, Pathogenic [43] C. Knudsen, A.H. Laustsen, Tropical medicine and infectious disease recent ad-
mechanisms underlying adverse reactions induced by intravenous administration of vances in next generation snakebite antivenoms, Trop. Med. Infect. Dis. 3 (2018)
snake antivenoms, Toxicon 76 (2013) 63–76. 1–11.
[17] C.Y. Huang, D.Z. Hung, W.K. Chen, Antivenin-related serum sickness, J. Chin. Med. [44] A. Silva, G.K. Isbister, Current research into snake antivenoms, their mechanisms of
Assoc. 73 (2010) 540–542. action and applications, Biochem. Soc. Trans. (2020) 1–10.
[18] J. Tibballs, Premedication for snake antivenom, Med. J. Aust. 160 (1994) 4–7. [45] S.V. Upasani, V.G. Beldar, A.U. Tatiya, M.S. Upasani, S.J. Surana, D.S. Patil,
[19] H.A. de Silva, A. Pathmeswaran, C.D. Ranasinha, S. Jayamanne, S.B. Samarakoon, Ethnomedicinal plants used for snakebite in India: a brief overview, Integr. Med.
A. Hittharage, R. Kalupahana, G.A. Ratnatilaka, W. Uluwatthage, J.K. Aronson, Res. 6 (2017) 114–130.
J.M. Armitage, D.G. Lalloo, H.J. de Silva, Low-dose adrenaline, promethazine, and [46] J. Félix-Silva, A.A. Silva-Junior, S.M. Zucolotto, M.D.F. Fernandes-Pedrosa,
hydrocortisone in the prevention of acute adverse reactions to antivenom following Medicinal plants for the treatment of local tissue damage induced by snake venoms:
snakebite: a randomised, double-blind, placebo-controlled trial, PLoS Med. 8 an overview from traditional use to pharmacological evidence, evidence-based
(2011). complement, Altern. Med. 2017 (2017).
[20] V. Morais, Antivenom therapy: efficacy of premedication for the prevention of [47] B.S. Vishwanath, A.G. Appu Rao, T.V. Gowda, Interaction of phospholipase A2 from
adverse reactions, J. Venom. Anim. Toxins Incl. Trop. Dis. 24 (2018) 1–7. Vipera russelli venom with aristolochic acid: a circular dichroism study, Toxicon 25
[21] A.S. Hertzberg SR, A life-threatening anaphylactoid reaction to polyvalent snake (1987) 939–946.
antivenom despite pretreatment, n.d. [48] B. Janardhan, V.M. Shrikanth, B.L. Dhananjaya, S.S. More, Antisnake venom
[22] I. Nuchprayoon, P. Garner, Interventions for preventing reactions to snake anti- properties of medicinal plants, Int. J. Pharm. Pharm. Sci. 7 (2015) 21–26.
venom, Cochrane Database Syst. Rev. (1999). [49] M. Madhusudan, F. Zameer, A. Naidu, M.N. Nagendra Prasad, B.L. Dhananjaya,
[23] A.G. Habib, Effect of pre-medication on early adverse reactions following anti- R. Hegdekatte, Evaluating the inhibitory potential of Withania somnifera on platelet
venom use in snakebite: a systematic review and meta-analysis, Drug Saf. 34 (2011) aggregation and inflammation enzymes: an in vitro and in silico study, Pharm. Biol.
869–880. 54 (2016) 1936–1941.
[24] G.K. Isbister, Snakebite doesn’t cause disseminated intravascular coagulation: [50] A.K. Mukherjee, R. Doley, D. Saikia, Isolation of a snake venom phospholipase A2
Coagulopathy and thrombotic microangiopathy in snake envenoming, Semin. (PLA2) inhibitor (AIPLAI) from leaves of Azadirachta indica (Neem): Mechanism of
Thromb. Hemost. 36 (2010) 444–451. PLA2 inhibition by AIPLAI in vitro condition, Toxicon 51 (2008) 1548–1553.
[25] K. Maduwage, G.K. Isbister, Current treatment for venom-induced consumption [51] J.C. Almagro, M. Pedraza-Escalona, H.I. Arrieta, S.M. Pérez-Tapia, Phage display
coagulopathy resulting from snakebite, PLoS Negl. Trop. Dis. 8 (2014). libraries for antibody therapeutic discovery and development, Antibodies 8
[26] G.K. Isbister, S.B. Duffull, S.G.A. Brown, Failure of antivenom to improve recovery (2019) 44.
in Australian snakebite coagulopathy, Qjm 102 (2009) 563–568. [52] N.A.-S., C.M. Modahl, Snakebite Therapeutics Based on Endogenous Inhibitors from
[27] G.K. Isbister, Procoagulant snake toxins: laboratory studies, diagnosis, and under- Vipers., Intech. (2016) 13.
standing snakebite coagulopathy, Semin. Thromb. Hemost. 35 (2009) 93–103. [53] K. Kinkawa, R. Shirai, S. Watanabe, M. Toriba, K. Hayashi, K. Ikeda, S. Inoue, Up-

7
A. Alangode, et al. Biochemical Pharmacology xxx (xxxx) xxxx

regulation of the expressions of phospholipase A2 inhibitors in the liver of a ve- [75] J.M. Gutiérrez, M.R. Lewin, D.J. Williams, B. Lomonte, Varespladib (LY315920)
nomous snake by its own venom phospholipase A2, Biochem. Biophys. Res. and methyl varespladib (LY333013) abrogate or delay lethality induced by pre-
Commun. 395 (2010) 377–381. synaptically acting neurotoxic snake venoms, Toxins (Basel) 12 (2020).
[54] J.M. Gutiérrez, A. Rucavado, F. Chaves, C. Díaz, T. Escalante, Experimental pa- [76] C.N. Zdenek, N.J. Youngman, C. Hay, J. Dobson, N. Dunstan, L. Allen, L. Milanovic,
thology of local tissue damage induced by Bothrops asper snake venom, Toxicon 54 B.G. Fry, Anticoagulant activity of black snake (Elapidae: Pseudechis) venoms:
(2009) 958–975. mechanisms, potency, and antivenom efficacy, Toxicol. Lett. 330 (2020) 176–184.
[55] C. Herrera, T. Escalante, M.B. Voisin, A. Rucavado, D. Morazán, J.K.A. Macêdo, [77] H. Xiao, H. Pan, K. Liao, M. Yang, C. Huang, Snake venom PLA2, a promising target
J.J. Calvete, L. Sanz, S. Nourshargh, J.M. Gutiérrez, J.W. Fox, Tissue localization for broad-spectrum antivenom drug development, Biomed Res. Int. 2017 (2017).
and extracellular matrix degradation by PI, PII and PIII snake venom metallopro- [78] J.M. Gutiérrez, A. Rucavado, Snake venom metalloproteinases: their role in the
teinases: clues on the mechanisms of venom-induced hemorrhage, PLoS Negl. Trop. pathogenesis of local tissue damage, Biochimie 82 (2000) 841–850.
Dis. 9 (2015) 1–20. [79] J. Gutiérrez, C.L. Ownby, G.V. Odell, Pathogenesis of myonecrosis induced by crude
[56] R.M. Kini, Excitement ahead: structure, function and mechanism of snake venom venom and a myotoxin of Bothrops asper, Exp. Mol. Pathol. 40 (1984) 367–379.
phospholipase A2 enzymes, Toxicon. 42 (2003) 827–840. [80] L. Moreira, J. Gutiérrez, G. Borkow, M. Ovadia, Ultrastructural alterations in mouse
[57] M.M. Thwin, R.P. Samy, S.D. Satyanarayanajois, P. Gopalakrishnakone, Venom capillary blood vessels after experimental injection of venom from the snake
neutralization by purified bioactive molecules: Synthetic peptide derivatives of the Bothrops asper (Terciopelo), Exp. Mol. Pathol. 57 (1992) 124–133.
endogenous PLA2 inhibitory protein PIP (a mini-review), Toxicon. 56 (2010) [81] B. Lomonte, J. Lundgren, B. Johansson, U. Bagge, The dynamics of local tissue
1275–1283. damage induced by Bothrops asper snake venom and myotoxin II on the mouse
[58] K.F. Huang, C.C. Hung, S.H. Wu, S.H. Chiou, Characterization of three endogenous cremaster muscle: an intravital and electron microscopic study, Toxicon 32 (1994)
peptide inhibitors for multiple metalloproteinases with fibrinogenolytic activity 41–55.
from the venom of Taiwan habu (Trimeresurus mucrosquamatus), Biochem. [82] F. Chaves, M. Barboza, J. Gutiérrez, Pharmacological study of edema induced by
Biophys. Res. Commun. 248 (1998) 562–568. venom of the snake Bothrops asper (terciopelo) in mice, Toxicon 33 (1995) 31–39.
[59] K.T. Yee, M. Pitts, P. Tongyoo, P. Rojnuckarin, M.C. Wilkinson, Snake venom me- [83] A. Rucavado, T. Escalante, J.M. Gutiérrez, Effect of the metalloproteinase inhibitor
talloproteinases and their peptide inhibitors from Myanmar Russell’s viper venom, batimastat in the systemic toxicity induced by Bothrops asper snake venom: un-
Toxins (Basel) 9 (2017). derstanding the role of metalloproteinases in envenomation, Toxicon 43 (2004)
[60] S.C. Wagstaff, P. Favreau, O. Cheneval, G.D. Laing, M.C. Wilkinson, R.L. Miller, 417–424.
R. Stöcklin, R.A. Harrison, Molecular characterisation of endogenous snake venom [84] A. Rucavado, T. Escalante, A. Franceschi, F. Chaves, G. León, Y. Cury, M. Ovadia,
metalloproteinase inhibitors, Biochem. Biophys. Res. Commun. 365 (2008) J.M. Gutiérrez, Inhibition of local hemorrhage and dermonecrosis induced by
650–656. Bothrops asper snake venom: effectiveness of early in situ administration of the
[61] R.H. Valente, B. Dragulev, J. Perales, J.W. Fox, G.B. Domont, BJ46a, a snake venom peptidomimetic metalloproteinase inhibitor batimastat and the chelating agent
metalloproteinase inhibitor isolation, characterization, cloning and insights into its CaNa2EDTA, Am. J. Trop. Med. Hyg. 63 (2000) 313–319.
mechanism of action, Eur. J. Biochem. 268 (2001) 3042–3052. [85] T. Escalante, A. Franceschi, A. Rucavado, J.M. Gutiérrez, Effectiveness of batima-
[62] D.J. Williams, J.M. Gutiérrez, J.J. Calvete, W. Wüster, K. Ratanabanangkoon, stat, a synthetic inhibitor of matrix metalloproteinases, in neutralizing local tissue
O. Paiva, N.I. Brown, N.R. Casewell, R.A. Harrison, P.D. Rowley, M. O’Shea, damage induced by BaP1, a hemorrhagic metalloproteinase from the venom of the
S.D. Jensen, K.D. Winkel, D.A. Warrell, Ending the drought: New strategies for snake Bothrops asper, Biochem. Pharmacol. 60 (2000) 269–274.
improving the flow of affordable, effective antivenoms in Asia and Africa, J. [86] J.M. Howes, R.D.G. Theakston, G.D. Laing, Neutralization of the haemorrhagic
Proteomics 74 (2011) 1735–1767. activities of viperine snake venoms and venom metalloproteinases using synthetic
[63] S.L.G. Rocha, A.G.C. Neves-Ferreira, M.R.O. Trugilho, Y. Angulo, B. Lomonte, peptide inhibitors and chelators, Toxicon 49 (2007) 734–739.
R.H. Valente, G.B. Domont, J. Perales, Screening for target toxins of the antiophidic [87] A.S. Arias, A. Rucavado, J.M. Gutiérrez, Peptidomimetic hydroxamate metallo-
protein DM64 through a gel-based interactomics approach, J. Proteomics 151 proteinase inhibitors abrogate local and systemic toxicity induced by Echis ocellatus
(2017) 204–213. (saw-scaled) snake venom, Toxicon 132 (2017) 40–49.
[64] B.V. Lipps, Anti-lethal factor from opossum serum is a potent antidote for animal, [88] I. Gómez-Betancur, V. Gogineni, A. Salazar-Ospina, F. León, Perspective on the
plant and bacterial toxins, J. Venom. Anim. Toxins. 5 (1999) 56–66. therapeutics of anti-snake venom, Molecules 24 (2019) 1–29.
[65] V.J. Claire, F. Komives, Elda E. Sanchez, Anurag S. Rathore, Brandon White, [89] S. GP, Filamentous fusion phage: novel expression vectors that display cloned an-
Montamas Suntravat, Michael Balderrama, Angela Cifelli, Opossum peptide that tigens on the virion surface, Science (80-.), 228 (1985) 1315–1317.
can neutralize rattlesnake venom is expressed in Escherichia coli, Physiol. Behav. [90] J. McCafferty, A.D. Griffiths, G. Winter, D.J. Chiswell, Phage antibodies: fila-
176 (2017) 139–148. mentous phage displaying antibody variable domains, Nature 348 (1990) 552–554.
[66] C. Drugs, A. Data, I. Bv, Varespladib, Am. J. Cardiovasc. Drugs 11 (2011) 137–143. [91] Á.M. Cuesta, N. Sainz-Pastor, J. Bonet, B. Oliva, L. Álvarez-Vallina, Multivalent
[67] I. Vergara, M. Pedraza-Escalona, D. Paniagua, R. Restano-Cassulini, F. Zamudio, antibodies: when design surpasses evolution, Trends Biotechnol. 28 (2010)
C.V.F. Batista, L.D. Possani, A. Alagón, Eastern coral snake Micrurus fulvius venom 355–362.
toxicity in mice is mainly determined by neurotoxic phospholipases A2, J. [92] J.C. Funayama, M.B. Pucca, E.C. Roncolato, T.B. Bertolini, L.B. Campos,
Proteomics 105 (2014) 295–306. J.E. Barbosa, Production of human antibody fragments binding to melittin and
[68] R. Arce-Bejarano, B. Lomonte, J.M. Gutiérrez, Intravascular hemolysis induced by phospholipase A2 in Africanised bee venom: minimising venom toxicity, Basic Clin.
the venom of the Eastern coral snake, Micrurus fulvius, in a mouse model: identi- Pharmacol. Toxicol. 110 (2012) 290–297.
fication of directly hemolytic phospholipases A2, Toxicon 90 (2014) 26–35. [93] L. Ledsgaard, T.P. Jenkins, K. Davidsen, K.E. Krause, A. Martos-Esteban,
[69] A.R. De Roodt, N.R. Lago, R.P. Stock, Myotoxicity and nephrotoxicity by Micrurus M. Engmark, M.R. Andersen, O. Lund, A.H. Laustsen, Antibody cross-reactivity in
venoms in experimental envenomation, Toxicon 59 (2012) 356–364. antivenom research, Toxins (Basel) 10 (2018) 1–19.
[70] M.R. Lewin, L.L. Gilliam, J. Gilliam, S.P. Samuel, T.C. Bulfone, P.E. Bickler, [94] M. Jianxin, T.R. John, I.I. Kaiser, Specificity and binding affinity of an anti-crotoxin
J.M. Gutiérrez, Delayed LY333013 (oral) and LY315920 (intravenous) reverse se- combinatorial antibody selected from a phage-displayed library, Biochem.
vere neurotoxicity and rescue juvenile pigs from lethal doses of micrurus fulvius Pharmacol. 50 (1995) 1969–1977.
(eastern coral snake) venom, Toxins (Basel) 10 (2018). [95] J. Gutierrez, B. Lomonte, G. Leon, A. Rucavado, F. Chaves, Y. Angulo, Trends in
[71] M. Lewin, S. Samuel, J. Merkel, P. Bickler, Varespladib (LY315920) appears to be a snakebite envenomation therapy: scientific, technological and public health con-
potent, broad-spectrum, inhibitor of snake venom phospholipase A2 and a possible siderations, Curr. Pharm. Des. 13 (2007) 2935–2950.
pre-referral treatment for envenomation, Toxins (Basel) 8 (2016). [96] E.C. Roncolato, L.B. Campos, G. Pessenda, L. Costa E Silva, G.P. Furtado, J.E.
[72] M.A. Bittenbinder, C.N. Zdenek, B. Op Den Brouw, N.J. Youngman, J.S. Dobson, Barbosa, Phage display as a novel promising antivenom therapy: a review, Toxicon
A. Naude, F.J. Vonk, B.G. Fry, Coagulotoxic cobras: clinical implications of strong 93 (2015) 79–84.
anticoagulant actions of african spitting naja venoms that are not neutralised by [97] J.K. Titus, M.K. Kay, J.J. Glaser, Y.Y. Hwang, Application of phage display for the
antivenom but are by LY315920 (varespladib), Toxins (Basel) 10 (2018). development of a novel inhibitor of PLA2 activity in western cottonmouth venom,
[73] Y. Wang, J. Zhang, D. Zhang, H. Xiao, S. Xiong, C. Huang, Exploration of the in- Toxicon 158 (2019) S35.
hibitory potential of varespladib for snakebite envenomation, Molecules 23 (2018). [98] J.M. Gutiérrez, G. León, G. Rojas, B. Lomonte, A. Rucavado, F. Chaves,
[74] G.H.M. Salvador, A.A.S. Gomes, W. Bryan-Quirós, J. Fernández, M.R. Lewin, Neutralization of local tissue damage induced by Bothrops asper (terciopelo) snake
J.M. Gutiérrez, B. Lomonte, M.R.M. Fontes, Structural basis for phospholipase A2- venom, Toxicon 36 (1998) 1529–1538.
like toxin inhibition by the synthetic compound Varespladib (LY315920), Sci. Rep. [99] D.G. Lalloo, R.D.G. Theakston, Snake antivenoms, J. Toxicol. - Clin. Toxicol. 41
9 (2019) 1–13. (2003) 277–290.

You might also like