Open access

Snake Venom Peptides: Promising Molecules with Anti-Tumor Effects

Written By

Sameh Sarray, Jose Luis, Mohamed El Ayeb and Naziha Marrakchi

Submitted: 31 May 2012 Published: 30 January 2013

DOI: 10.5772/51263

From the Edited Volume

Bioactive Food Peptides in Health and Disease

Edited by Blanca Hernandez-Ledesma and Chia-Chien Hsieh

Chapter metrics overview

3,713 Chapter Downloads

View Full Metrics

1. Introduction

Tumorigenesis and metastasis are two processes with inter-related mechanisms. These include tumor growth and angiogenesis, detachment of tumor cells from the primary tumor, followed by migration through the local connective tissue and penetration into the circulation (intravasation). Once in the blood stream, tumor cells interact with circulating blood cells, arrest in the microvasculature of target organs, then extravasate and secondary proliferate. During each of these steps, integrin-mediated adhesion, migration, proliferation and survival of tumor cells and angiogenic endothelial cells play crucial roles [1,2].

Integrins are a family of heterodimeric transmembrane receptors that mediate cell-cell and cell-extracellular matrix (ECM) interactions. These cell adhesion molecules are composed by non covalent association of α and β subunits. Although 18 α and 8 β subunits have been described, only 24 different combinations have been identified to date [3]. Specific integrin heterodimers preferentially bind distinct ECM proteins. The repertory of integrins present on a given cell dictates the extent to which that cell will adhere to and migrate on different matrices. Several integrins, among others αv and α5β1, recognize the RGD sequence on their respective ligands. Other adhesive sequences in ECM proteins have also been observed, including the EILDV and REDV sequences that are recognized by integrin α4β1 in an alternatively spliced form of fibronectin [3]. On ligation to the ECM, integrins cluster in the plane of the membrane and recruit various signalling and adaptor proteins to form structures known as focal adhesions [4].

Integrin expression can also vary considerably between normal and tumor tissue. Most notably, integrins αvβ3, α5β1 and αvβ6 are usually expressed at low or undetectable levels in most adult epithelia but can be highly up-regulated in some tumors. Expression levels of some integrins, such as α2β1, decrease in tumor cells; potentially increasing tumor cell dissemination [5]. The integrin αvβ3 is particularly important for tumor growth and invasiveness [6]. The receptor plays a major role in neo-vessels formation, its expression being strongly up-regulated in endothelial cells and specifically required during angiogenesis stimulated by basic fibroblast growth factor (bFGF) and tumor necrosis factor-α [7,8]. αvβ3 is functionally involved in the malignant spread of various tumor cell types such as breast carcinoma, prostate carcinoma and melanoma, and supports tumor cell adhesion and migration through endothelium [9] and matrix proteins [10,1]. Blocking αvβ3 is therefore expected to have a broad impact in cancer therapy and diagnosis. In the last decade, several clinical trials evaluating the efficacy of αvβ3 blockers have led to encouraging results. Thus, MEDI-522 (Vitaxin), a humanized antibody derived from the mouse LM609 monoclonal antibody, was recently reported to give positive results in a phase II trial enrolling patients with stage IV metastatic melanoma [11]. Cilengitide is an inhibitor of both αvβ3 and αvβ5 integrins; it is currently being tested in phase II trials in patients with lung and prostate cancers [12] and in phase II and Phase III trials studying their role against glioblastoma are currently underway.

In addition to their role in tumor cells, integrins are also important for the host cellular response against cancer. Endothelial cells, fibroblasts, pericytes, bone marrow-derived cells, inflammatory cells and platelets all use integrins for various functions, including angiogenesis, desmoplasia and immune response.

Nature has been a source of medicinal products for thousands of years among which snake venoms form a rich source of bioactive molecules such as peptides, proteins and enzymes with important pharmacological activities. International research and development in this area, based on multidisciplinary approaches including molecular screening, proteomics, genomics and pharmacological in vitro, ex vivo and in vivo assays, allow the identification and characterization of highly specific molecules from snake venom that can potently inhibit integrin functions. These anti-adhesive snake venom proteins belong to different families (phospholipases, disintegrins, C-type lectins and metalloproteinases). By targeting integrins, they exhibit various pharmacological activities such as anti-tumor, anti-angiogenic and/or pro-apoptotic effects.

Advertisement

2. Snake venom protein families

2.1. The Snake Venom Metalloproteinases (SVMP)

Metalloproteinases are among the most abundant toxins in many Viperidae venoms. SVMPs are monozinc endopeptidases varying in size from 20 to 100 kDa. They are phylogenetically most closely related to the mammalian disintegrin and metalloproteinase (ADAM) family of proteins. SVMPs are grouped into several subclasses according to their domain organization [13, 14, 15]. P-I SVMPs are the simplest class of enzymes that contain only a metalloproteinase (M) domain. P-II SVMPs contain a M domain followed by a disintegrin (D) domain. P-III SVMPs contain M, disintegrin-like (D) and cysteine-rich (C) domains. Formally called P-IV, the heterotrimeric class of SVMPs that contain an additional snake C-type lectin-like (snaclec) domain [16] is now included in the P-III group as a subclass (P-IIId).

Most of the functional activities of SVMPs are associated with hemorrhage or the disruption of the hemostatic system, which are primarily mediated by the proteolytic activity of the M domain. SVMPs cause hemorrhage by disturbing the interactions between endothelial cells and the basement membrane through the degradation of endothelial cell membrane proteins (e.g., integrin, cadherin) and basement membrane components (e.g., fibronectin, laminin, nidogen, type IV collagen) [17]. Blood coagulation proteins (e.g., fibrinogen, factor X, prothrombin) are also targets of their proteolytic activities.

Echis carinatus venom contains the specific prothrombin activators, ecarin [18,19] and carinactivase [20]. Adamalysin II, a non-hemorrhagic P-I SVMP isolated from Crotalus adamantus venom, cleaves and inactivates serum proteinase inhibitors including antithrombin III [21]. Kaouthiagin, isolated from the venom of Naja kaouthia specifically binds and cleaves von Willebrand factor (vWF), resulting in loss of both the ristocetin-induced platelet aggregation and collagen-binding activity of vWF [22]. Additionally, a large number of the P-III SVMPs can inhibit platelet aggregation, thus enhancing the hemorrhagic state [23]. The hemorrhagic P-III SVMP jararhagin from the venom of Bothrops jararaca has been shown to degrade platelet collagen receptor α2β1 integrin in addition to fibrinogen and vWF, resulting in the inhibition of platelet aggregation [24]. Other platelet receptors are also degraded by SVMPs. GPIbα is cleaved by kistomin; mocarhagin and crotalin [25-27], and GPVI is degraded by alborhagin, crotarhagin and kistomin [28,29].

In the other side, it was reported that several SVMPs inhibited integrin-mediated adhesion of cancer cells on ECM proteins (table 1). BaG, a dimeric PIII class of SVMP from Bothrops alternatus with inactivated enzymatic domain but intact D/C domain, has been reported to inhibit fibronectin-mediated K562 cell adhesion via α5β1 integrin [30].

ProteinsSnakeIntegrinsEffectsReferences
VAP1, VAP2Crotalus atroxα3,α6,β1Induce apoptosis of HUVEC[31,36]
HV1Trimeruserus flavoviridis-Inhibits adhesion of HUVEC and induces apoptosis[32]
HalysaseGloydius halysα1β1;α5β1Inhibits proliferation and Induces apoptosis of HUVEC[33]
VLAIPsVipera lebetina-Inhibits proliferation and Induces apoptosis of HUVEC[34]
GraminelysinTrimeresurus gramineusα1β1;α5β1Inhibits proliferation and Induces apoptosis of HUVEC[35]
BaGBothrops alternatusα5β1Inhibits adhesion of K562 cells[30]
TSV-DMTrimeresurus stejnegeri-Inhibits cell proliferation and induces transient cell morphologic changes of endothelial cells.[113]

Table 1.

SVMP affecting tumor cells

Several apoptosis-inducing proteins have been purified from hemorrhagic snake venom, such as VAP1 and VAP2 (Crotalus atrox), HV1 (Trimeresurus flavoviridis), halysase (Gloydius halys), and VLAIPs (Vipera lebetina) [31-34], graminelysin [35]. They are members of the SVMP and ADAM family and induce apoptosis of human umbilical vein endothelial cells (HUVECs) [31,36]. The detachment of endothelial cells and resulting apoptosis could be an additional mechanism for the disruption of normal hemostasis by SVMPs. TSV-DM a basic metalloproteinase from Trimeresurus stejnegeri venom inhibits cell proliferation and induces cell morphologic changes transiently of ECV304 cells. However, DNA fragmentation and DNA content analysis demonstrated that this metalloproteinase could not induce ECV304 cells apoptosis.

2.2. The disintegrins

Disintegrins are a family of non-enzymatic and low molecular weight proteins derived from viper venom [37-39]. They are able to inhibit platelet aggregation and interact with adhesion molecules in particular integrins in a dose-dependent manner. They have a K / RTS sequence which is known as the RGD adhesive loop [37-39]. Their primary structure shows a strong conservation in the arrangement of cysteines [38]. Most disintegrins represent the C-terminal domain of metalloproteinases PIIa, d and e classes and are released into the venom by proteolytic cleavage [40,37,38]. A minority of these proteins exist as D / C domains from the class of SVMPs PIIIb.

Disintegrins can be conveniently divided into five different groups according to their length and the number of disulfide bridges [41]. The first group includes short disintegrins, single polypeptide composed of 49 - 51 amino acids with four disulfide bridges. The second group comprises medium disintegrins containing about 70 amino acids and six disulfide bridges. The third group includes long disintegrins of 83 residues linked by seven disulfide bridges. The disintegrin domains of PIII snake-venom metalloproteinases, containing approx. hundred amino acids with 16 Cysteine residues involved in the formation of eight disulfide bonds, constitute the fourth subgroup of the disintegrin family. Unlike short-, medium- and long-sized disintegrins, which are single-chain molecules, the fifth subgroup is composed of homo and heterodimers. The dimeric disintegrins subunits contain about 67 residues with four disulfide intra-chain bridges and two interchain bridges [42,43].

Although disintegrins are highly homologous, significant differences exist in their affinity and selectivity for integrins, which explains the multitude of effects of these molecules (Table 2).

Disintegrins were first identified as inhibitors of platelet aggregation and were subsequently shown to antagonize fibrinogen binding to platelet integrin αIIbβ3 [44,45]. After that, studies on disintegrins have revealed new uses in the diagnosis of cardiovascular diseases and the design of therapeutic agents in arterial thrombosis, osteoporosis, and angiogenesis-related tumor growth and metastasis (table 2). Triflavin from Trimeresurus flavoviridis venom was one of the first RGD-disintegrins shown to inhibit angiogenesis both in vitro and in vivo [46]. Triflavin strongly inhibited cell migration toward vitronectin and fibronectin nearly thirty orders of magnitude greater than anti-αvβ3 monoclonal antibodies [46]. Triflavin was also more effective in inhibiting TNF-α-induced angiogenesis in the chicken chorioallantoic membrane (CAM) assay. Similar results were obtained with another RGD-disintegrin, rhodostomin, from Agkistrodon rhodostoma venom, which inhibits endothelial cell migration, invasion and tube formation induced by bFGF in MatrigelTM both in vitro and in vivo [47]. Rhodostomin effects were inhibited by anti-αvβ3 but not by anti-αvβ5 antibodies, thus supporting the hypothesis that the effects of RGD-disintegrins are mediated by blockade of the vitronectin receptor.

ProteinsSnakeIntegrinsEffectsReferences
TriflavinTrimeresurus flavoviridisα5β1,αvβ3,
α3β1
Inhibits adhesion of tumor cells to matrix proteins, cell migration and angiogenesis in vitro and in vivo[46]
RhodostominAgikistrodon rhodostomaαvβ3,αvβ5Inhibits cell migration, invasion of endothelial cells; inhibits angiogenesis in vivo and in vitro[47]
ContortrostatinAgkistrodon contortrix contortrixαvβ3,α5β1,
αvβ5,
αIIββ3
Blocks adhesion, migration invasion of different type of tumor cells[48]
LebestatinMacrovipera lebetinaα1β1Inhibits migration and angiogenesis[56]
Accurhagin-CAgkistrodon acutusαvβ3Prevents migration and invasion of endothelial cells; anti-angiogenic activity in vitro and in vivo; elicites anoïkis[58]
EristostatinEritocophis macmahoniα4β1,other integrin not yet determinedInhibits cell motility; no effect on cell proliferation or angiogenesis[59,60]
DisBa-01Bothrops alternatusαvβ3Anti-angiogenic and anti-metastatic effect on melanoma cells[62]
Leberagin-CMacrovipera lebetinaαvβ3Inhibits cell adhesion of melanoma tumor cells[114]
AccutinAgkistrodon acutusαvβ3Inhibits angiogenesis in vitro and in vivo; induces apoptosis[115]

Table 2.

Effects of disintegrins on cancerous cells

Contortrostatin, a disintegrin isolated from the venom of the southern copperhead snake, exhibits anti-cancer activity in a variety of tumor cells [48-50]. It does not display cytotoxic activity in vitro nor animals upon injection. Contortrostatin inhibits adhesion, migration, invasion, metastatic and angiogenesis of tumor and endothelial cells mediated by αvβ3,α5β1 and αvβ5 [48,50-54]. Recently, contortrostatin showed an additive inhibitory effect in combination with docetaxel on the growth of xenograft tumors derived from prostate cancer cells [55].

Lebestatin is an example of a non toxic KTS-disintegrin isolated from Macrovipera lebetina that inhibits migration and VEGF-induced in vivo angiogenesis [56]. The presence of a WGD motif in CC8, a heterodimeric disintegrin from Echis carinatus, increases its inhibitory effect on αvβ3 and α5β1 integrins [57].

There are few reports regarding the effects of ECD-disintegrins on endothelial cell migration. Acurhagin-C, dose-dependently blocked HUVEC migration toward a vitronectin-coated membrane. Furthermore, acurhagin-C elicited endothelial anoïkis via disruption of the αvβ3/FAK/PI3K survival cascade and subsequent initiation of the procaspase-3 apoptotic signaling pathway [58].

Eristostatin, an RGD-disintegrin from Eristocophis macmahoni was tested on individual metastasis steps such as cell arrest, extravasation and migration [59]. Eristostatin treatment did not prevent tumor cell extravasation or migration [60]. However, it was shown later that eristostatin inhibited melanoma cell motility, an effect mediated by fibronectin-binding integrins [61]. Interestingly, this disintegrin, contrary to other RGD-disintegrins, did not inhibit angiogenesis, as stated before [61]. DisBa-01, a αvβ3 integrin-blocking RGD-disintegrin, inhibits not only migration of endothelial cells in vivo [62] but also in vitro migratory ability of fibroblasts and two tumor cell lines.

Since integrin receptors are also quite indiscriminate as they support cell adhesion to several substrates, it seems highly reasonable that the general RGD-disintegrin scaffold of the integrin-binding motif could be employed as a prototype for drug design for new anti-metastatic therapies via blocking both tumor cell adhesion and tumor angiogenesis.

2.3. The snake venom phospholipases

Snake venom is one of the most abundant sources of secretory phospholipases A2 (PLA2), which are one of the potent molecules in snake venoms [63-65].

PLA2 (EC 3.1.1.4)—are enzymes that catalyze the hydrolysis of sn-2-acyl bond of sn-3-phospholipids, generating free fatty acids and lysophospholipids as products [66]. They are currently classified in 15 groups and many subgroups that include five distinct types of enzymes, namely secreted PLA2 (sPLA2), cytosolic PLA2 (cPLA2), Ca2+ independent PLA2s (iPLA2), platelet-activating factor acetyl-hydrolases (PAF-AH), lysosomal PLA2, and a recently identified adipose-specific PLA2 [65,67]. PLA2 are low molecular weight proteins with molecular masses ranging from 13-19 kDa that generally require Ca2+ for their activities [69,70]. Snake venom sPLA2 are secreted enzymes belonging to only two groups that are based on their primary structure and disulfide bridge pattern [68,71,72]. Those of group I are similar to pancreatic sPLA2 present in mammals, were found in venom of Elapidae snakes, while group II PLA2s belong to the Viperidae and are similar to mammals nonpancreatic, inflammatory sPLA2s [73,74]. The group II can be subdivided mainly in two subgroups, depending on the residue at position 49 in the primary structure: Aspartic acid-49 PLA2s are enzymatically active, while Lysine 49 present low or no enzymatic activity [75]. There are other subgroups, such as Asparagine-49, Serine-49, Glutamine-49 and Arginine -49 [76-83]. Studies have found that catalytic activity is reduced or even abolished when an Aspartic acid of native PLA2 is replaced by another amino acid [80,84].

Despite a high identity of their amino acid sequences, sPLA2 exhibit a wide variety of pharmacological properties such as anticoagulant, haemolytic, neurotoxic, myotoxic, oedema-inducing, hemorrhagic, cytolytic, cardiotoxic, muscarinic inhibitor and antiplatelet activities [63,85-92].

Recently, PLA2s have been shown to possess anti-tumor and anti-angiogenic properties (Table 3). CC-PLA2-1 and CC-PLA2-2 from Cerastes cerastes viper are non-toxic and acidic proteins. They have high inhibitory effects on platelet aggregation and coagulation. In addition, CC-PLA2-1 and CC-PLA2-2 inhibit the adhesion of the human fibrosarcoma (HT1080) and melanoma (IGR39) cells to fibrinogen and fibronectin. In the same direction, CC-PLA2-1 and CC-PLA2-2 potently reduces HT1080 cell migration to fibrinogen and fibronectin with nearly similar IC50 values [93]. This anti-adhesive effect was due to the inhibition of α5β1 and αv-containing integrins [94]. A recent report demonstrated that Bth-A-I, a non-toxic PLA2 isolated from Bothrops jararacussu venom display an anti-tumoral effect upon breast adenocarcinoma as well as upon human leukaemia T and Erlich ascetic tumor [95].

ProteinsSnakeIntegrinsEffectsReferences
CCPLA2-1; CCPLA2-2Cerastes cerastesα5β1,αvInhibits migration and adhesion of fibrosarcoma and melanoma cells[93,94]
Bth-A-I-PLA2Bothrops jararacussu-Anti-tumor activity on adenocarcinoma and leukaemia cells[95]
MVL-PLA2Macrovipera lebetinaα5β1,αvInhibits adhesion and migration of human microvascular cells and inhibits angiogenesis in vivo and in vitro.[96]
BP IIProthobotrops flavoviridis-Induces cell death in human leukaemia cells[97]

Table 3.

PLA2s targeting tumor cells

MVL-PLA2 is a snake venom phospholipase purified from Macrovipera lebetina venom that inhibited adhesion and migration of human microvascular endothelial cells (HMEC-1) without being cytotoxic. Using MatrigelTM and chick chorioallantoic membrane assays, MVL-PLA2, as well as its catalytically inactivated form, significantly inhibited angiogenesis both in vitro and in vivo. Also, the actin cytoskeleton and the distribution of αvβ3 integrin, a critical regulator of angiogenesis and a major component of focal adhesions, were disturbed after MVL-PLA2 treatment. The enhancement of microtubule dynamics of HMEC-1 cells, in consequence of treatments by MVL-PLA2, may explain the alterations in the formation of focal adhesions, leading to inhibition of cell adhesion and migration [96].

A cell death activity was discovered in Lysine 49-PLA2 called BPII. It induces caspase-independent cell death in human leukaemia cells regardless of its depressed enzymatic activity [97].

2.4. The C-type lectins

The C-type lectins are abundant components of snake venom with various function. Typically, these proteins bind calcium and sugar residues. However, the C-type lectin like proteins from snake venom (termed actually snaclec) does not contain the classic calcium/sugar binding loop and have evolved to bind a wide range of physiologically important proteins and receptors [98].

Snaclecs have a basic heterodimeric structure with two subunits, nearly always linked covalently, via a disulphide bond. The heterodimers are often further multimerized either non-covalently or covalently via additional disulphide bonds, to form larger structures [99]. The two subunits form a concave surface between them [100] thus constituting the main site of ligand binding [101,102]. The subunits have a high structural degree of homology between them and with other snaclecs [103]. Despite their highly conserved primary structure, the snaclecs are characterized by various biological activities. They were and are still considered as modulators of platelet aggregation by targeting vWF, GPIb-IX-V, GPVI and possibly other platelet receptors.

Recently, novel activities of snaclecs were highlighted. They were described for their potential anti-tumor effect by blocking adhesion, migration, proliferation and invasion of different cancer cell lines (Table 4). Among these proteins, EMS16, a heterodimer isolated from the venom of Echis multisquamatus, inhibits the adhesion of HUVECs cells on ECM proteins and their migration by inhibiting the binding of integrin α2β1 to collagen [104].

Lebecetin and lebectin, purified from Macrovipera lebetina venom, are the only snaclecs, until today, with an evident anti-tumor effect in addition to their anti-aggregation activity on platelets. Indeed, these two non cytotoxic proteins inhibit the adhesion of various cancer cell lines: melanoma (IGR39), adenocarcinoma (HT29-D4), fibrosarcoma (HT1080) and leukemia cells (K562) on different ECM proteins. They also inhibit the proliferation, migration and invasion of HT1080 cells [105,106]. Lebectin also displays anti-angiogenic activity at very low concentrations both in vitro and in vivo [107]. Thus, lebectin presents the best anti-angiogenic efficacy yet described for snake venom-derived peptides [108,109]. These observed effects are mediated by α5β1 and αv integrins [107].

Extensive researches have been shown that cell adhesion activities in cancer disease are deregulated. According to this idea, it was also reported that lebectin inhibits these alterations by promoting N-cadherin/catenin complex reorganisation at cell-cell contacts, inducing a strengthening of intercellular adhesion [110].

Another snaclec, BJcuL isolated from Bothrops jararacussa venom, was also described for its anti-tumor, but the receptor or integrin implicated has not been determined yet. This homodimeric protein inhibits proliferation of several cell lines of renal, pancreatic, prostate and melanoma origin, but no effect was observed on colon or breast cancer cells [111]. BJcuL also affects the viability of some tumor cell lines of different origins, but has no effect on the growth of K562 and T24 cells, suggesting that these cells do not express the receptor recognized by the lectin. BJcuL induces apoptosis in human gastric carcinoma cells accompanied by inhibition of cell adhesion and actin cytoskeleton disassembly [112].

ProteinsSnakeIntegrinsEffectsReferences
Lebecetin, lebectinMacrovipera lebetinaα5β1,αvInhibits adhesion, migration and invasion of human tumor cells; inhibits angiogenesis[106]
BJcuLBothrops jararacussuInhibits tumor cell and endothelial cell growth; induces apoptosis of human gastric carcinoma cells; inhibits cell adhesion and actin cytoskeleton disassembly[111,112]
EM16Echis multisquamatusα2β1Inhibits adhesion and migration of HUVEC cells[104]

Table 4.

Snaclecs and their effects on tumor cells

Advertisement

3. Potential application of snake venom compounds

Venoms are a rich source of molecules endowed with diverse pharmacological effects. Most part of these molecules act via the adhesion molecules. The intervention of the scientists and the clinicians in the pharmaceutical development field would employ these molecules as therapeutic agents for several pathologies such as cancer, thrombosis, diabetes....

Until now, no medicine was produced from a native molecule purified from venom. However, several peptidomimetics were designed by basing on the structure of these molecules. The benefits of these peptidomimetics compared to antibodies that can be used for the treatment of certain diseases are: a shorter half-life, reversible inhibition, easier to control a problem and very low immunogenicity. For example, the antihypertensive drug captopril, modelled from the venom of the Brazilian arrowhead viper (Bothrops jaracusa); the anticoagulant Integrilin (eptifibatide), a heptapeptide derived from a protein found in the venom of the American southeastern pygmy rattlesnake (Sistrurus miliarius barbouri); Ancrod, a compound isolated from the venom of the Malaysian pit viper (Agkistrodon rhodostoma) for use in the treatment of heparin-induced thrombocytopenia and stroke and alfimeprase, a novel fibrinolytic metalloproteinase for thrombolysis derived from southern copperhead snake (Agkistrodon contortrix contortrix) venom (Table 5). Two venom proteins from the Australian brown snake, Pseudonaja textilis, are currently in development as human therapeutics (QRxPharma). The first is a single agent procoagulant that is a homolog of mammalian Factor Xa prothrombin activator, whereas the other is a plasmin inhibitor, named Textilinin-1, with antihemorrhagic properties.

NameSnakeTarget and function/treatmentClinical stage
Capoten ® (Captropil)Bothrops jaracusaAngiotensin converted enzyme (ACE) inhibitor/ high blood pressureGranted FDA approval
Integrilin ® (Eptifibatide)Sisturus miliarus barbouriPlatelet aggregation inhibitor/acute coronary syndromeGranted FDA approval
Aggrastat ® (tirofiban)Echis carinatusGPIIb-IIIa inhibitor/ myocardial infarct, refractory ischemiaApproved for use with heparin and aspirin for the treatment of acute coronary syndrome
ExantaCobraThrombin inhibitor/ arterial fibrillation and bloodSeeking FDA approval
Alfimeprase(Agkistrodon contortrix contortrix)Thrombolytic/ Acute ischemic stroke, acute peripheral arterial occlusionPhase III
Ancrod ® (viprinex)Agkistrodon rhodostomaFibrinogen inhibitor/ strokePhase III
hemocoagulaseBothrops atroxThrombin-like effect and thromboplastin activity/ prevention and treatment of haemorrhagePhase III
Protac/ Protein C activatorAgkistrodon contortrix contortrixProtein C activator/clinical diagnosis of haemostatic disorderGranted FDA approval
ReptilaseBothrops jaracaDiagnosis of blood coagulation disorderGranted FDA approval
EcarinEchis carinatusProthrombin activator/ diagnosticGranted FDA approval

Table 5.

Drugs and clinical diagnostic kits from snake venom

Actually, most of the current anticancer therapies (radiotherapy, chemotherapy) are not specific and are targeting at both tumor cells and healthy cells. However, in recent years, new treatments tend to focus on the tumor microenvironment and particularly on the inhibition of tumor angiogenesis. These treatments are based on several active and non toxic proteins from snake venom, as for example contortrostatin from Agkistrodon contortrix contortrix and eristostatin from Eristocophis macmahoo. Although all these molecules are still currently in clinical trials, they could in the future open new ways of healing and could be used as drugs.

Advertisement

4. Conclusions

From the initial discovery of captopril, the first oral ACE inhibitor, to the recent application of disintegrins for the potential treatment of cancer, the various components of snake venoms have never failed to reveal amazing new properties. While the original native snake venom compounds are usually unsuitable as therapeutics, interventions by medicinal chemists as well as scientists and clinicians in pharmaceutical R&D have made it possible to use the snake venom proteins as potential drugs for multiple disorders or scaffolds for drug design.

References

  1. 1. Felding-HabermannB.Integrin adhesion receptors in tumor metastasis. Clinical and Experimental Metastasis 2003203203213
  2. 2. Fidler IJ.Biological behavior of malignant melanoma cells correlated to their survival in vivo. Cancer Research 1975351218224
  3. 3. BarczykM.CarracedoS.GullbergD.IntegrinsCell Tissue Research 20103391269280
  4. 4. Berrier AL and Yamada KM. Cell Matrix.Journal of cellular physiology 20072133565573
  5. 5. KrenA.BaeriswylV.LehembreF.WunderlinC.StrittmatterK.AntoniadisH.FässlerR.CavallaroU.ChristoforiG.Increased tumor cell dissemination and cellular senescence in the absence of β1-integrin function. The EMBO Journal 2007261228322842
  6. 6. AlbeldaS. M.MetteS. A.ElderD. E.StewartR.DamjanovichL.HerlynM.CABuckIntegrin distribution in malignant melanoma: association of the beta 3 subunit with tumor progression. Cancer Research 1990502067576764
  7. 7. Brooks PC, Clark RA, Cheresh DA.Requirement of vascular integrin alpha v beta 3 for angiogenesis. Science 19942645158569571
  8. 8. EnensteinJ.WalehN. S.KramerR. H.BasicF. G. F.FbetaT. G.differentiallymodulate.integrinexpression.ofhuman.microvascularendothelial.cellsExperimental Cell Research 19922032499503
  9. 9. Voura EB, Ramjeesingh RA, Montgomery AM, Siu CH.Involvement of integrin alpha(v)beta(3) and cell adhesion molecule L1 in transendothelial migration of melanoma cells. Molecular Biology of the Cell 200112926992710
  10. 10. Cooper CR, Chay CH, Pienta KJ.The role of alpha(v)beta (3) in prostate cancer progression. Neoplasia 200243191194
  11. 11. HerseyP.SosmanJ.O’DayS. A.phaseI. I.randomizedopen-label.studyevaluating.theantitumor.activityof. M. E. D.I-52humanizeda.monoclonalantibody.directedagainst.thehuman.alphav.beta.(αvβintegrin±.dacarbazine. D. T. I. C.inpatients.withmetastatic.melanoma. M. M.A. S. C.ASCO Annual Meeting Proceedings, Journal of Clinical Oncology 2005
  12. 12. BeekmanK. W.ColevasA. D.CooneyK.DipaolaR.DunnR. L.GrossM.KellerE. T.PientaK. J.RyanC. J.SmithD.HussainM.PhaseI. I.evaluationsof.cilengitidein.asymptomaticpatients.withandrogen.independentprostate.cancerscientific.rationalestudydesign.Clinical Genitourinary Cancer 200644299302
  13. 13. Fox JW and Serrano SM.Structural considerations of the snake venom metalloproteinases, key members of the M12 reprolysin family of metalloproteinases, Toxicon 2005458969985
  14. 14. Fox JW and Serrano SM.Insights into and speculations about snake venom metalloproteinase (SVMP) synthesis, folding and disulfide bond formation and their contribution to venom complexity, FEBS Journal 20082751230163030
  15. 15. TakeyaH.MiyataT.NishinoN.Omori-SatohT.IwanagaS.Snake venom hemorrhagic and non hemorrhagic metalloendopeptidases, Methods in Enzymolgy 1993223365
  16. 16. ClemetsonK. J.MoritaT.ManjunathaKini. R.Scientific and standardization committee communications: classification and nomenclature of snake venom C-type lectins and related proteins, Journal of Thrombosis Haemostasis 2009360 EOF360 EOF
  17. 17. Baramova EN, Shannon JD, Bjarnason JB, Fox JW.Degradation of extracellular matrix proteins by hemorrhagic metalloproteinases. Archives of Biochemistry and Biophysics 198927516371
  18. 18. MoritaT.IwanagaS.Purification and properties of prothrombin activator from the venom of Echis carinatus. Journal of Biochemistry 1978832559570
  19. 19. NishidaS.FujitaT.KohnoN.AtodaH.MoritaT.TakeyaH.KidoI.MJPaineKawabata. S.IwanagaS. c. D. N. A.cloningdeducedamino.acidsequence.ofprothrombin.activator(ecarin.fromKenyan.Echiscarinatus.venomBiochemistry 199534517711778
  20. 20. YamadaD.SekiyaF.MoritaT.Isolation and characterization of carinactivase, a novel prothrombin activator in Echis carinatus venom with a unique catalytic mechanism. Journal Biological Chemistry 1996271952005207
  21. 21. Kress LF and Paroski EA.Enzymatic inactivation of human serum proteinase inhibitors by snake venom proteinases. Biochemical and Biophysics Research Communications 1978832649656
  22. 22. HamakoJ.MatsuiT.NishidaS.NomuraS.FujimuraY.ItoM.OzekiY.TitaniK.Purification and characterization of kaouthiagin, a von Willebrand factor-binding and-cleaving metalloproteinase from Naja kaouthia cobra venom, Thrombosis and Haemostasis 1998803499505
  23. 23. Laing GD, Moura-da-Silva AM.Jararhagin and its multiple effects on hemostasis. Toxicon 2005458987996
  24. 24. Kamiguti AS.Platelets as targets of snake venom metalloproteinases. Toxicon 200545810411049
  25. 25. Huang TF, Chang MC, Teng CM. Antiplatelet protease, kistomin, selectively cleaves human platelet glycoprotein Ib.Biochemica et Biophysica Acta 199311583293299
  26. 26. Ward CM, Andrews RK, Smith AI, Berndt MC. Mocarhagin, a novel cobra venom metalloproteinase, cleaves the platelet von Willebrand factor receptor glycoprotein Ib alpha.Identification of the sulfated tyrosine/anionic sequence Tyr-276-Glu-282 of glycoprotein Ib alpha as a binding site for von Willebrand factor and alpha-thrombin, Biochemistry 1996351549294938
  27. 27. Wu WB, Peng HC, Huang TF. Crotalin, a vWF and GP Ib cleaving metalloproteinase from venom of Crotalus atrox.Thrombosis and Haemostasis 200186615011511
  28. 28. HsuC. C.WuW. B.HuangT. F. A.snakevenom.metalloproteinasekistomin.cleavesplatelet.glycoproteinV. I.impairsplatelet.functionsJournal.ofThrombosis.Haemostasis20086915781585
  29. 29. WijeyewickremaL. C.EEGardinerMoroi. M.BerndtM. C.AndrewsR. K.Snakevenom.metalloproteinasescrotarhagin.alborhagininduce.ectodomainshedding.ofthe.plateletcollagen.receptorglycoprotein. V. I.Thrombosis and Haemostasis 200798612851290
  30. 30. Cominetti MR, Ribeiro JU, Fox JW, Selistre-de-Araujo HS. BaG, a new dimeric metalloproteinase/disintegrin from the Bothrops alternatus snake venom that interacts with alpha5beta1 integrin.Archives of Biochemistry and. Biophysics 20034162171179
  31. 31. MasudaS.ArakiS.KajiK.HayashiH.Purification of a vascular apoptosis-inducing factor from hemorrhagic snake venom. Biochemical and Biophysics Research Communications 199723515963
  32. 32. MasudaS.HayashiH.AtodaH.MoritaT.ArakiS.Purificationc. D. N. A.cloningcharacterizationof.thevascular.apoptosis-inducingprotein. H. V.fromTrimeresurus.flavoviridisEuropean Journal of Biochemistry 20012681133393345
  33. 33. YouW. K.SeoH. J.ChungK. H.DSKimA.novelmetalloprotease.fromGloydius.halysvenom.inducesendothelial.cellapoptosis.throughits.proteasedisintegrin-likedomains.Journal of Biochemistry 20031345739749
  34. 34. TrummalK.TonismagiK.SiigurE.AaspolluA.LoppA.SillatT.SaatR.KasakL.TammisteI.KogermanP.KalkkinenN.SiiguJ. A.novelmetalloprotease.fromVipera.lebetinavenom.induceshuman.endothelialcell.apoptosisToxicon 20054614661
  35. 35. ShinW. U. W. B.Ming-YiC. C.Tur-FuL.PurificationH.molecularcloning.mechanismof.actionof.graminelysinI. a.snake-venom-derivedmetalloproteinase.thatinduces.apoptosisof.humanendothelial.cellsBiochemical Journal 2001Pt 3) 719 EOF28 EOF
  36. 36. MasudaS.HayashiH.ArakiS.Two vascular apoptosis-inducing proteins from snake venom are members of the metalloprotease/disintegrin family. European Journal of Biochemistry 199825313641
  37. 37. MAMc LaneSanchez.EEWongA.Paquette-StraubC.PerezJ. C.DisintegrinsCurrent.DrugsTargets.CardiovascularHaematologicalDisorders.20044327
  38. 38. Calvete JJ.Structure-function correlations of snake venom disintegrins. Current Pharmaceutical Design 2005117829835
  39. 39. CalveteJ. J.MarcinkiewiczC.MonleonD.EsteveV.CeldaB.JuarezP.SanzL.Snakevenom.disintegrinsevolution.ofstructure.functionToxicon 200545810631074
  40. 40. KiniR. M.EvansH. J.Structural domains in venom proteins: evidence that metalloproteinases and nonenzymatic platelet aggregation inhibitors (disintegrins) from snake venoms are derived by proteolysis from a common precursor. Toxicon 1992303265293
  41. 41. CalveteJ. J.Moreno-MurcianoM. P.TheakstonR. D.KisielD. G.MarcinkiewiczC.Snakevenom.disintegrinsnovel.dimericdisintegrins.structuraldiversification.bydisulphide.bondengineering.Biochemical Journal 2003Pt 3) 725 EOF34 EOF
  42. 42. CalveteJ. J.JurgensM.MarcinkiewiczC.RomeroA.SchraderM.NiewiarowskiS.Disulphide-bond pattern and molecular modelling of the dimeric disintegrin EMF-10, a potent and selective integrin alpha5beta1 antagonist from Eristocophis macmahoni venom. Biochemical Journal 2000aPt 3) 573 EOF81 EOF
  43. 43. BilgramiS.TomarS.YadavS.KaurP.KumarJ.JabeenT.SharmaS.SinghT. P.Crystal structure of schistatin, a disintegrin homodimer from saw-scaled viper (Echis carinatus) at 2.5 A° resolution. Journal of Molecular Biology 20043413829837
  44. 44. GouldR. J.MAPolokoffFriedman. P. A.HuangT. F.HoltJ. C.CookJ. J.NiewiarowskiS.Disintegrinsa.familyof.integrininhibitory.proteinsfrom.vipervenoms.Proceeding of the Society for Experimental Biology and Medicine 19901952168171
  45. 45. OuyangC.YehH. I.HuangT. F. A.potentplatelet.aggregationinhibitor.purifiedfrom.Agkistrodonhalys.(mamushisnake.venomToxicon 1983216797804
  46. 46. Sheu JR, Yen MH, Kan YC, Hung WC, Chang PT, Luk HN.Inhibition of angiogenesis in vitro and in vivo: comparison of the relative activities of triflavin, an Arg-Gly-Asp-containing peptide and anti-alpha(beta3integrin monoclonal antibody. Biochemica et Biophysica Acta 1997445 EOF54 EOF
  47. 47. Yeh CH; Peng HC; Yang RS, Huang TF. Rhodostomin, a snake venom disintegrin, inhibits angiogenesis elicited by basic fibroblast growth factor and suppresses tumor growth by a selective alpha(v)beta(3) blockade of endothelial cells.Molecular Pharmacology 200159513331342
  48. 48. SwensonS.CostaF.ErnstW.FujiiG.MarklandF. S.Contortrostatina.snakevenom.disintegrinwith.anti-angiogenicanti-tumoractivity.Pathophysiology of Haemostasis and Thrombosis 2005169 EOF176 EOF
  49. 49. SwensonS.CostaF.MineaR.SherwinR. P.ErnstW.FujiiG.YangD.MarklandF. S.Jr Intravenous liposomal delivery of the snake venom disintegrin contortrostatin limits breast cancer progression. Molecular Cancer Therapeutics 200434499511
  50. 50. TrikhaM.De ClerckY. A.MarklandF. S.Contortrostatina.snakevenom.disintegrininhibits.beta.integrin-mediatedhuman.metastaticmelanoma.celladhesion.blocksexperimental.metastasisCancer Research 1994541849934998
  51. 51. ZhouQ.SherwinR. P.ParrishC.RichtersV.GroshenS. G.Tsao-WeiD.MarklandF. S.Contortrostatina.dimericdisintegrin.fromAgkistrodon.contortrixcontortrix.inhibitsbreast.cancerprogression.Breast Cancer Research and Treatment 2000a613249260
  52. 52. ZhouQ.NakadaM. T.BrooksP. C.SwensonS. D.RitterM. R.ArgounovaS.ArnoldC.MarklandF. S.Contortrostatina.homodimericdisintegrin.bindsto.integrinalphavbeta.Biochemical Biophysical Research Communications 2000b2671350355
  53. 53. TrikhaM.RoteW. E.ManleyP. J.LucchesiB. R.MarklandF. S.Purification and characterization of platelet aggregation inhibitors from snake venoms. Thrombosis Research 19947313952
  54. 54. RitterM. R.ZhouQ.MarklandF. S.Jr Contortrostatina.snakevenom.disintegrininduces.alphavbeta3-mediatedtyrosine.phosphorylationof. C. A. S.inF. A. K.tumorcells.Journal of Cellular Biochemistry 20007912837
  55. 55. LinE.WangQ.SwensonS.JadvarH.SusanG.YeW.MarklandF.PinskiJ.The disintegrin contortrostatinin combination with docetaxel is a Potent Inhibitor of Prostate cancer in vitro and in vivo. The Prostate 2010701213591370
  56. 56. OlfaK. Z.JoseL.SalmaD.BazaaA.SrairiN.NicolasA.MaximeL.ZouariR.MabroukK.MarvaldiJ.SabatierJ. M.El AyebM.MarrakchiN.Lebestatina.disintegrinfrom.Macroviperavenom.inhibitsintegrin-mediated.celladhesion.migrationangiogenesisLaboratory Investigation 2005851215071516
  57. 57. CalveteJ. J.FoxJ. W.AgelanA.NiewiarowskiS.MarcinkiewiczC.The presence of the WGD motif in CC8 heterodimeric disintegrin increases its inhibitory effect on alphaII(b)beta3, alpha(v)beta3, and alpha5beta1 integrins. Biochemistry 200241620142021
  58. 58. MorrisV. L.EESchmidtKoop. S.MacDonald. I. C.GrattanM.KhokhaR.MAMc LaneNiewiarowskiS.ChambersA. F.GroomA. C.Effects of the disintegrin eristostatin on individual steps of hematogenous metastasis. Experimental Cell Research 19952192571578
  59. 59. Wang WJ. Acurhagin-C, an ECD disintegrin, inhibits integrin alphavbeta3-mediated human endothelial cell functions by inducing apoptosis via caspase-3 activation.British Journal of Pharmacology 2010160613381351
  60. 60. Sohn YD, Cho KS, Sun SA, Sung HJ, Kwak KW, Hong SY, Kim DS, Chung KH.Suppressive effect and mechanism of saxatilin, a disintegrin from Korean snake (Gloydiussaxatilis), in vascular smooth muscle cells. Toxicon 2008523474480
  61. 61. TianJ.Paquette-StraubC.SageE. H.FunkS. E.PatelV.GalileoD.MAMc LaneInhibition of melanoma cell motility by the snake venom disintegrin eristostatin. Toxicon 2007497899908
  62. 62. RamosO. H.KauskotA.CominettiM. R.BechyneI.SallaPontes. C. L.ChareyreF.ManentJ.VassyR.GiovanniniM.LegrandC.Selistre-de-AraujoH. S.CrepinM.AABonnefoyNovel alpha(v)beta (3)-blocking disintegrin containing the RGD motive, DisBa-01, inhibits bFGF-induced angiogenesis and melanoma metastasis. Clinical & Experimental Metastasis 20082515364
  63. 63. Kini RM. Excitement ahead: structure, function and mechanism of snake venom phospholipase A2 enzymes.Toxicon 2003428827840
  64. 64. Burke JE and Dennis EA. Phospholipase A2 Biochemistry.Cardiovascular drugs and Therapy 2009a231122
  65. 65. RamarP. S.GopalakrishnakoneP.BowH.PuspharajP. N.ChowV. T.Identification and characterization of a phospholipase A2 from the venom of the Saw-scaled viper: Novel bactericidal and membrane damaging activities. Biochimie 2010921218541866
  66. 66. RitonjaA.GubensekF.AmmodytoxinA. a.highlylethal.phospholipaseA.fromVipera.ammodytesammodytes.venomBiochemica et Biophysica Acta 1985828393306
  67. 67. Maung-MaungT.GopalakrishnakoneP.YuenR.TanC. H. A.majorlethal.factorof.thevenom.ofBurmese.Russell’sviper. .Daboiarusselli.siamensisisolationN-terminal.sequencingbiologicalactivities.ofdaboiatoxin.Toxicon 19953316376
  68. 68. ChakrabartyD.DattaK.GomesA.BhattacharyyaD.Haemorrhagic protein of Russell’s viper venom with fibrinolytic and esterolytic activities. Toxicon 2000381114751490
  69. 69. Kini RM.Phospholipase A2-a complex multifunctional protein puzzle. In: Kini, R M (ed) Enzymes: Structure, Function and Mechanism. John Wiley and Sons, Chichester, England;1997128
  70. 70. ValentinE.LambeauG.Increasing molecular diversity of secreted phospholipases A(2) and their receptors and binding proteins. Biochemica et Biophysica Acta 2000
  71. 71. Six DA and Dennis EA.The expanding superfamily of phospholipase A(2) enzymes: classification and characterization. Biochemica et Biophysica Acta 2000
  72. 72. RouaultM.BollingerJ. G.LazdunskiM.GelbM. H.LambeauG.Novel mammalian group XII secreted phospholipase A2 lacking enzymatic activity. Biochemistry 200342391149411503
  73. 73. LambeauG.LazdunskiM.Receptors for a growing family of secreted phospholipases A2. Trends in Pharmacological Sciences 1999204162170
  74. 74. Dennis EA. Phospholipase A2 in ecicosanoid generation.American Journal of Respiratory and Critical Care Medicine 2000Pt 2) S32S35.
  75. 75. LomonteB.AnguloY.CalderonL.Angulo Y, Calderon L. An overview of lysine-49 phospholipase A2 myotoxins from crotalid snake venoms and their structural determinants of myotoxic action. Toxicon 2003428885901
  76. 76. Tsai IH, Wang YM, Chen YH, Tsai TS. Venom phospholipases A2 of bamboo viper (Trimeresurus stejnegeri): molecular characterization, geographic variations and evidence of multiple ancestries.Biochemical Journal 2004Pt 1) 215 EOF23 EOF
  77. 77. WeiJ. F.WeiX. L.ChenQ. Y.HuangT.QiaoL. Y.WangW. Y.XiongY. L.HeS. H. N.phospholipaseA.uniquea.subgroupof.snakevenom.groupI. I.phospholipaseA.Biochemica et Biophysica Acta 200617603462471
  78. 78. KrizajI.BieberA. L.RitonjaA.GubensekF.The primary structure of ammodytin L, a myotoxic phospholipase A2 homologue from Vipera ammodytes venom. European Journal of Biochemistry 1991202311651168
  79. 79. PolgarJ.MagnenatE. M.PeitschM. C.WellsT. N.ClemetsonK. J.Asp-isnot.anabsolute.prerequisitefor.theenzymic.activityof.low-M(rphospholipases. A.purificationcharacterization.computermodelling.ofan.enzymicallyactive.Ser-phospholipaseA.ecarpholinS.fromthe.venomof.Echiscarinatus.sochureki(saw-scaled.viperBiochemical Journal 1996Pt 3) 961-968.
  80. 80. BaoY.BuP.JinL.WangH.YangQ.AnL.Purificationcharacterization.genecloning.ofa.novelphospholipase. A.fromthe.venomof.Agkistrodonblomhoffii.ussurensisThe International Journal of Biochemistry & Cell Biology Cell Biol 2005373558565
  81. 81. ChijiwaT.TokunagaE.IkedaR.TeradaK.OgawaT.Oda-UedaN.HattoriS.NozakiM.OhnoM.Discovery of novel [Arg49] phospholipase A2 isozymes from Protobothrops elegans venom and regional evolution of Crotalinae snake venom phospholipase A2 isozymes in the southwestern islands of Japan and Taiwan. Toxicon 2006486672682
  82. 82. MebsD.KuchU.CoronasF. I. V.BatistaC. V. F.GumprechtA.PossaniL. D.Biochemical and biological activities of the venom of the Chinese pitviper Zhaoermia mangshanensis, with the complete amino acid sequence and phylogenetic analysis of a novel Arg49 phospholipase A2 myotoxin. Toxicon 2006477797811
  83. 83. WeiJ. F.LiT.WeiX. L.SunQ. Y.YangF. M.ChenQ. Y.WangW. Y.XiongY. L.HeS. H.Purificationcharacterization.cytokinerelease.functionof. a.novelArg-.phospholipaseA.fromthe.venomof.Protobothropsmucrosquamatus.Biochimie 2006881013311342
  84. 84. LiY.YuB. Z.ZhuH.JainM. K.MDTsaiPhospholipase. A.engineeringStructural and functional roles of the highly conserved active site residue aspartate-49. Biochemistry 199433491471414722
  85. 85. Kini RM and Evans HJ.Correlation between the enzymatic activity, anticoagulant and antiplatelet effects of phospholipase A2 isoenzymes from Naja nigricollis venom. Thrombosis and Haemostasis 1988602170173
  86. 86. KasturiS.GowdaT. V.Purification and characterization of a major phospholipase A2 from Russell’s viper (Vipera russelli) venom. Toxicon 1989272229237
  87. 87. StefanssonS.KiniR. M.EvansH. J.The inhibition of clotting complexes of the extrinsic coagulation cascade by the phospholipase A2 isoenzymes from Naja nigricollis venom. Thrombosis Research 1989554481491
  88. 88. Maung-MaungT.GopalakrishnakoneP.YuenR.TanC. H. A.majorlethal.factorof.thevenom.ofBurmese.Russell’sviper. .Daboiarusselli.siamensisisolationN-terminal.sequencingbiologicalactivities.ofdaboiatoxin.Toxicon 19953316376
  89. 89. HuangM. Z.GopalakrishnakoneP.KiniR. M.Role of enzymatic activity in the antiplatelet effects of a phospholipase A2 from Ophiophagus hannah snake venom. Life Sciences 1997612222112217
  90. 90. KoleL.ChakrabartyD.DattaK.BhattacharyyaD.Purification and characterization of an organ specific haemorrhagic toxin from Vipera russelli russelli (Russell’s viper) venom. Indian Journal of Biochemistry & Biophysics 2000372114120
  91. 91. Chakrabarty AK, Hall RH, Ghose AC.Purification and characterization of a potent hemolytic toxin with phospholipase A2 activity from the venom of Indian Russell’s viper. Molecular and Cellular Biochemistry 2002
  92. 92. DongM.GudaK.NambiarP. R.RezaieA.BelinskyG. S.LambeauG.GiardinaC.RosenbergD. W.Inverse association between phospholipase A2 and COX-2 expression during mouse colon tumorigenesis. Carcinogenesis 2003242307315
  93. 93. Zouari-KessentiniR.LuisJ.KarrayA.Kallech-ZiriO.Srairi-AbidN.BazaaA.LoretE.BezzineS.El AyebM.MarrakchiN.Two purified and characterized phospholipases A2 from Cerastes cerastes venom that inhibits cancerous cell adhesion and migration. Toxicon 2009534444453
  94. 94. Zouari-KessentiniR.JebaliJ.TaboubiS.Srairi-AbidN.MorjenM.Kallech-ZiriO.BezzineS.MarvaldiJ.El AyebM.Marrakchi-PN. C. C.A2L.-PC. C.A2-L.twocerastes.cerastesvenom.derivedphospholipases. A.inhibitangiogenesis.bothin.vitroinvivo.Laboratory Investigation 2010904510519
  95. 95. RobertoP. G.KashimaS.MarcussiS.PereiraJ. O.Astolfi-FilhoS.NomizoA.GiglioJ. R.FontesM. R.SoaresA. M.FançaS. C.Cloning and identification of a complete cDNA coding for a bactericidal and anti-tumoral acidic phospholipase A2 from Bothrops jararacussu venom. Protein Journal 2004234273285
  96. 96. BazaaA.PasquierE.DefillesC.LimamI.Kessentini-ZouariR.Kallech-ZiriO.El BattariA.BraguerD.El AyebM.MarrakchiN.Luis-PJ. M. V. L.L. A.snakea.venomphospholipase. A.inhibitsangiogenesis.throughan.increasein.microtubuledynamics.disorganizationof.focaladhesions. P.LoPLoS One 2010e10124 EOF
  97. 97. MurakamiT.KamikadoN.FujimotiR.HamaguchiK.NakamuraH.ChijiwaT.OhnoM.Oda-UedaA. [.Lys49]phospholipase. A.fromProtobothrops.flavoviridisvenom.inducescaspase-independent.apoptoticcell.deathaccompanied.byrapid.plasma-membranerupture.inhuman.leukemiacells.Biosciences, Biotechnology and Biochemistry 2011755864870
  98. 98. LuQ.NavdaevA.ClemetsonJ. M.ClemetsonK. J.Snake venom C-type lectins interacting with platelet receptors: structure-function relationships and effects on haemostasis. Toxicon 200545810891098
  99. 99. EbleJ. A.NilandS.BrachtT.MormannM.Peter-KatalinicJ.PohlentzG.StetefeldJ.Thealpha2beta.integrin-specificantagonist.rhodocetinis. a.cruciformheterotetrameric.moleculeF. A. S. E.FASEB Journal 200923929172927
  100. 100. MizunoH.FujimotoZ.KoizumiM.KanoH.AtodaH.MoritaT.Crystal structure of coagulation factor IX-binding protein from habu snake venom at 2.6 A: implication of central loop swapping based on deletion in the linker region. Journal of Molecular Biology 19992891103112
  101. 101. HoriiK.OkudaD.MoritaT.MizunoH.Crystal structure of EMS16 in complex with the integrin alpha2-I domain. Journal of Molecular Biology 20043412519527
  102. 102. MaitaN.NishioK.NishimotoE.MatsuiT.ShikamotoY.MoritaT.SadlerJ. E.MizunoH.Crystal structure of von willebrand factor A1 domain complexed with snake venom, bitiscetin: insight into glycoprotein Ibα binding mechanism induced by snake venom proteins. The Journal of Biological Chemistry 2003278393777737781
  103. 103. RunhuaW. R.ManjunathaK.MaxC. M. C.Rhodocetina.novelplatelet.aggregationinhibitor.fromthe.venomof.Calloselasmarhodostoma. .Malayanpit.vipersynergisticnoncovalent.interactionbetween.subunitsBiochemistry 1999382375847593
  104. 104. MarcinkiewiczC.LobbR. R.MMMarcinkiewiczDaniel. J. L.SmithJ. B.DangelmaierC.WeinrebP. H.BeachamD. A.NiewiarowskiS.Isolation and characterization of EMS16, a C-lectin type protein from Echis multisquamatus venom, a potent and selective inhibitor of the α2β1 integrin. Biochemistry 2000393298599867
  105. 105. SarrayS.BerthetV.CalveteJ. J.SecchiJ.MarvaldiJ.El AyebM.MarrakchiN.LuisJ.Lebectina.novelC-type.lectinfrom.Macroviperalebetina.venominhibits.integrinmediated.adhesionmigration.invasionof.humantumour.cellsLaboratory Investigation 2004845573581
  106. 106. SarrayS.DelamarreE.MarvaldiJ.El AyebM.MarrakchiN.LuisJ.Lebectinlebecetintwo.C-typelectins.fromsnake.venominhibit.alphabeta.alphaV-containing.integrinsMatrix Biology 2007264306313
  107. 107. PilorgetA.ConesaM.SarrayS.Michaud-LevesqueJ.DaoudS.KimK. S.DemeuleM.MarvaldiJ.El AyebM.MarrakchiN.BeliveauR.LuisJ.Lebectina.Macroviperalebetina.venom-derivedC-type.lectininhibits.angiogenesisboth.invitro.invivo.Journal of Cellular Physiology 200720072112307
  108. 108. GolubkovV.HawesD.MarklandF. S.Anti-angiogenic activity of contortrostatin, a disintegrin from Agkistrodon contortrix contortrix snake venom. Angiogenesis 200363213224
  109. 109. MarcinkiewiczC.WeinrebP. H.CalveteJ. J.KisielD. G.MousaS. A.TuszynskiG. P.LobbR. R.ObtustatinA.potentselective.inhibitorof.alpha1betaintegrin.invitro.angiogenesisin.vivoCancer Research 200363920202023
  110. 110. SarrayS.SiretC.LehmannM.MarrakchiN.LuisJ.El AyebM.AndreF.Lebectin increases N-cadherin-mediated adhesion through PI3K/AKT pathway. Cancer Letters 20092852174181
  111. 111. Pereira-BittencourtM.CarvalhoD. D.GagliardA. R.CollinsD. C.The effect of a lectin from the venom of the snake, Bothrops jararacussu, on tumor cell proliferation. Anticancer Research 1999B) 40234025
  112. 112. NolteS.de CastroDamasio. D.BaréaA. C.GomesJ.MagalhãesA.MelloZischler. L. F.Stuelp-CampeloP. M.Elífio-EspositoS. L.Roque-BarreiraM. C.CAReis-AmaralMoreno.JcuA. N. B.BJcuL, a lectin purified from Bothrops jararacussu venom, induces apoptosis in human gastric carcinoma cells accompanied by inhibition of cell adhesion and actin cytoskeleton disassembly. Toxicon 20125918185
  113. 113. WanS. G.JinY.LeeW. H.ZhangY. A.snakevenom.metalloproteinasethat.inhibitedcell.proliferationinducedmorphological.changesof. E. C.V3cellsToxicon 2006474480489
  114. 114. LimamI.BazaaA.Srairi-AbidN.TaboubiS.JebaliJ.Zouari-KessentiniR.Kallech-ZiriO.MejdoubH.HammamiA.El AyebM.LuisJ.MarrakchiN.Leberagin-CA.disintegrin-like/cysteine-richprotein.fromMacrovipera.lebetinatransmediterranea.venominhibits.alphavbetaintegrin-mediated.celladhesion.Matrix Biology 2010292117126
  115. 115. YehC. H.PengH. C.YihJ. B.HuangT. F. A.newshort.chainR. G.D-containingdisintegrin.accutininhibits.thecommon.pathwayof.humanplatelet.aggregationBiochemica Biophysica Acta 199814253493504

Written By

Sameh Sarray, Jose Luis, Mohamed El Ayeb and Naziha Marrakchi

Submitted: 31 May 2012 Published: 30 January 2013