Acessibilidade / Reportar erro

Antineoplastic properties and pharmacological applications of Crotalus durissus terrificus snake venom

ABSTRACT

Snake toxins are widely studied owing to their importance in snakebite accidents, a serious public health issue in tropical countries, and their broad therapeutic potential. Isolated fractions from venom produced by snakes of the genus Crotalus sp. present a wide variety of pharmacological uses such as antifungal, antiviral, antibacterial, and antitumor properties, among other therapeutic potentialities. Given the direct effect of this venom on tumor cells, isolation of its compounds is important for the characterization of its anticarcinogenic actions. Crotalus durissus terrificus venom and its toxins have been widely evaluated as potential candidates for the development of new antineoplastic therapies that are efficient against different tumor lines and cellular targets. This review highlights the venom toxins of this species, with a focus on their antineoplastic properties.

Keywords:
Snake Venom; Cancer; Antitumor; Crotalid Venoms; Crotalus

INTRODUCTION

Currently, approximately 11,341 reptile species are recognized worldwide11. Uetz P, Hošek J. The Reptile Database. [S.I.]: Peter Uetz; 2020 [updated 2021 may 22; cited 2020 october 27]. Available from: Available from: http://www.reptile-database.org .
http://www.reptile-database.org...
, with 1,116 species found in Australia, 974 in Mexico, and 830 in Brazil. Crotalus comprises of the venomous Viperidae snakes22. Fraga R, Lima AP, Prudente ALC, Magnusson WE. Guide to the snakes of the Manaus region - Central Amazonia, 1st ed. Manaus: Editora Inpa, 2013. 303 p.

3. Mader DR. Reptile Medicine and Surgery, 2nd ed. [S. I.]: Elsevier; 2005. 1264 p.

4. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.

5. Marques OAV, Medeiros CR. Our Incredible Serpents: Characterization, Biology, Accidents and Conservation. 1st ed. Cotia: Ponto A; 2018. 80 p.

6. Frare BT, Resende YKS, Dornelas BC, Jorge MT, Ricarte VAS, Alves LM, et al. Clinical, Laboratory, and Therapeutic Aspects of Crotalus durissus (South American Rattlesnake) Victims: A Literature Review. Biomed Res Int. 2019; Article ID 1345923:1-7. Available from: https://doi.org/10.1155/2019/1345923
https://doi.org/10.1155/2019/1345923...
-77. Tasima LJ, Serino-Silva C, Hatakeyama DM, Nishiduka ES, Tashima AK, Sant’Anna SS, et al. Crotamine in Crotalus durissus: distribution according to subspecies and geographic origin, in captivity or nature. J Venom Anim Toxins Incl Trop Dis. 2020;26:e20190053. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2019-0053
https://doi.org/10.1590/1678-9199-jvatit...
from the subfamily Crotalinae, also known as rattlesnakes. These are distributed across South America, mainly from Colombia to Argentina55. Marques OAV, Medeiros CR. Our Incredible Serpents: Characterization, Biology, Accidents and Conservation. 1st ed. Cotia: Ponto A; 2018. 80 p.,77. Tasima LJ, Serino-Silva C, Hatakeyama DM, Nishiduka ES, Tashima AK, Sant’Anna SS, et al. Crotamine in Crotalus durissus: distribution according to subspecies and geographic origin, in captivity or nature. J Venom Anim Toxins Incl Trop Dis. 2020;26:e20190053. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2019-0053
https://doi.org/10.1590/1678-9199-jvatit...

8. Rangel-Santos A, Dos-Santos EC, Lopes-Ferreira M, Lima C, Cardoso DF, Mota I. A comparative study of biological activities of crotoxin and CB fraction of venoms from Crotalus durissus terrificus, Crotalus durissus cascavella and Crotalus durissus collilineatus. Toxicon. 2004;43(7):801-10. Available from: https://doi.org/10.1016/j.toxicon.2004.03.011.
https://doi.org/10.1016/j.toxicon.2004.0...
-99. Wüster W, Ferguson JE, Quijada-Mascareñas JA, Pook CE, Salomão MDG, Thorpe RS. Tracing an invasion: Landbridges, refugia, and the phylogeography of the Neotropical rattlesnake (Serpentes: Viperidae: Crotalus durissus). Mol Ecol. 2005;14(4):1095-80. Available from: https://doi.org/10.1111/j.1365-294X.2005.02471.x.
https://doi.org/10.1111/j.1365-294X.2005...
, with the following six subspecies found in Brazil: Crotalus durissus cascavella, C. d. collilineatus, C. d. terrificus, C. d. marajoensis, C. d. ruruima, and C. d. durissus44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,66. Frare BT, Resende YKS, Dornelas BC, Jorge MT, Ricarte VAS, Alves LM, et al. Clinical, Laboratory, and Therapeutic Aspects of Crotalus durissus (South American Rattlesnake) Victims: A Literature Review. Biomed Res Int. 2019; Article ID 1345923:1-7. Available from: https://doi.org/10.1155/2019/1345923
https://doi.org/10.1155/2019/1345923...
,99. Wüster W, Ferguson JE, Quijada-Mascareñas JA, Pook CE, Salomão MDG, Thorpe RS. Tracing an invasion: Landbridges, refugia, and the phylogeography of the Neotropical rattlesnake (Serpentes: Viperidae: Crotalus durissus). Mol Ecol. 2005;14(4):1095-80. Available from: https://doi.org/10.1111/j.1365-294X.2005.02471.x.
https://doi.org/10.1111/j.1365-294X.2005...
.

These snakes are primarily nocturnal55. Marques OAV, Medeiros CR. Our Incredible Serpents: Characterization, Biology, Accidents and Conservation. 1st ed. Cotia: Ponto A; 2018. 80 p. and solenoglyphic dentition55. Marques OAV, Medeiros CR. Our Incredible Serpents: Characterization, Biology, Accidents and Conservation. 1st ed. Cotia: Ponto A; 2018. 80 p.,1010. Fry BG, Sunagar K, Casewell NR, Kochva E, Roelants K, Scheib H, et al. The origin and evolution of the toxicofera reptile venom system. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press; 2015. p. 1-31. presents loreal pits, a thermoreceptor organ of viperid species, visible as openings between the eye and the nostril of the animal head, which are of great importance for the detection of temperature variations, particularly of prey and predators55. Marques OAV, Medeiros CR. Our Incredible Serpents: Characterization, Biology, Accidents and Conservation. 1st ed. Cotia: Ponto A; 2018. 80 p.. The most striking characteristic of Crotalus snakes is the presence of a rattle at the end of their tails (Figure 1)55. Marques OAV, Medeiros CR. Our Incredible Serpents: Characterization, Biology, Accidents and Conservation. 1st ed. Cotia: Ponto A; 2018. 80 p.,66. Frare BT, Resende YKS, Dornelas BC, Jorge MT, Ricarte VAS, Alves LM, et al. Clinical, Laboratory, and Therapeutic Aspects of Crotalus durissus (South American Rattlesnake) Victims: A Literature Review. Biomed Res Int. 2019; Article ID 1345923:1-7. Available from: https://doi.org/10.1155/2019/1345923
https://doi.org/10.1155/2019/1345923...
.

FIGURE 1:
(A)Crotalus durissus terrificus; (B) Rattle detail; (C) Loreal Pit (yellow circle).

Crotalus snakes cause frequent and severe accidents, represent a serious public health problem in tropical countries, and the snakebites are considered a neglected disease by the World Health Organization66. Frare BT, Resende YKS, Dornelas BC, Jorge MT, Ricarte VAS, Alves LM, et al. Clinical, Laboratory, and Therapeutic Aspects of Crotalus durissus (South American Rattlesnake) Victims: A Literature Review. Biomed Res Int. 2019; Article ID 1345923:1-7. Available from: https://doi.org/10.1155/2019/1345923
https://doi.org/10.1155/2019/1345923...
,1111. WHO - World Health Organization. Report of the Tenth Meeting of the WHO Strategic and Technical Advisory Group for Neglected Tropical Diseases. Geneva: WHO; 2017 [cited 2020 Oct 27]. Available from: Available from: http://www.who.int/neglected_diseases/NTD_STAG_report_2017.pdf?ua=1
http://www.who.int/neglected_diseases/NT...

12. Grego KF, Vieira SEM, Vidueiros JP, Serapicos EO, Barbarini CC, Silveira GPM, et al. Maintenance of venomous snakes in captivity for venom production at Butantan Institute from 1908 to the present: a scoping history. J Venom Anim Toxins Incl Trop Dis . 2021;27:e20200068. Available from: https://dx.doi.org/10.1590/1678-9199-jvatitd-2020-0068
https://doi.org/10.1590/1678-9199-jvatit...
-1313. Chippaux JP. Snakebite envenomation turns again into a neglected tropical disease. J Venom Anim Toxins Incl Trop Dis . 2017;8;23:38. Available from: https://doi.org/10.1186/s40409-017-0127-6
https://doi.org/10.1186/s40409-017-0127-...
. However, venom is an important biotechnological tool because of the specialization and efficiency of its components, which affect a large number of targets with high selectivity and affinity1414. El-Aziz TMA, Soares AG, Stockand JD. Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins. 2019;11(10):1-25. Available from: https://doi.org/10.3390/toxins11100564
https://doi.org/10.3390/toxins11100564...

15. Almeida JR, Resende LM, Watanabe RK, Carregari VC, Huancahuire-Vega S, Caldeira CAS, et al. Snake Venom Peptides and Low Mass Proteins: Molecular Tools and Therapeutic Agents. Curr Med Chem. 2017;24(30):3254-82. Available from: https://doi.org/10.2174/0929867323666161028155611
https://doi.org/10.2174/0929867323666161...
-1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
.

CROTALUS DURISSUS TERRIFICUS VENOM: COMPOSITION, GENERAL PHARMACOLOGICAL ACTIONS AND ANTINEOPLASTIC APPLICATIONS

Snake venom is one of the richest sources of bioactive substances in nature and is therefore of great interest for the development of new drugs44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,1414. El-Aziz TMA, Soares AG, Stockand JD. Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins. 2019;11(10):1-25. Available from: https://doi.org/10.3390/toxins11100564
https://doi.org/10.3390/toxins11100564...

15. Almeida JR, Resende LM, Watanabe RK, Carregari VC, Huancahuire-Vega S, Caldeira CAS, et al. Snake Venom Peptides and Low Mass Proteins: Molecular Tools and Therapeutic Agents. Curr Med Chem. 2017;24(30):3254-82. Available from: https://doi.org/10.2174/0929867323666161028155611
https://doi.org/10.2174/0929867323666161...

16. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...

17. Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B. Anticancer potential of animal venoms and toxins. Indian J Exp Biol. 2010;48(2):93-103.

18. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...

19. Soares MA, Pujatti PB, Fortes-Dias CI, Antonelli L, Santos RG. Crotalus durissus terrificus venom as a source of antitumoral agents. J Venom Anim Toxins Incl Trop Dis . 2010;16(3):480-92. Available from: https://doi.org/10.1590/S1678-91992010000300015
https://doi.org/10.1590/S1678-9199201000...

20. Vonk FJ, Jackson K, Doley R, Madaras F, Mirtschin PJ, Vidal N. Snake venom: From fieldwork to the clinic: Recent insights into snake biology, together with new technology allowing high-throughput screening of venom, bring new hope for drug discovery. Bioessays. 2011;33(4):269-79. Available from: http://dx.doi.org/10.1002/bies.201000117
https://doi.org/10.1002/bies.201000117...

21. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...

22. Kollipara PS, Won DH, Hwang CJ, Jung YY, Yoon HS, Park MH, et al. Enhanced Anti-Cancer Effect of Snake Venom Activated NK Cells on Lung Cancer Cells by Inactivation of NF-κB. Biomol Ther. 2014;22(2):106-13. Available from: http://dx.doi.org/10.4062/biomolther.2013.103
https://doi.org/10.4062/biomolther.2013....

23. Melani RD, Araujo GDT, Carvalho PC, Goto L, Nogueira FCS, Junqueira M, et al. Seeing beyond the tip of the icebeg: A deep analysis of the venome of the Brazilian Rattlesnake, Crotalus durissus terrificus. EuPA Open Proteom. 2015;8:144-56. Available from: https://doi.org/10.1016/j.euprot.2015.05.006
https://doi.org/10.1016/j.euprot.2015.05...

24. Boldrini-França J, Cologna CT, Pucca MB, Bordon KCF, Amorim FG, Anjolette FAP, et al. Minor snake venom proteins: Structure, function and potential applications. Biochim Biophys Acta Gen Subj. 2017;1861(4):824-38. Available from: http://dx.doi.org/10.1016/j.bbagen.2016.12.022
https://doi.org/10.1016/j.bbagen.2016.12...

25. Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, et al. Secondary homeostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus. Int J Biol Macromol. 2019;131:127-33. Available from: https://doi.org/10.1016/j.ijbiomac.2019.03.059
https://doi.org/10.1016/j.ijbiomac.2019....

26. Pérez-Peinado C, Defaus S, Andreu D. Hitchhiking with Nature: Snake Venom Peptides to Fight Cancer and Superbugs. Toxins. 2020;12(4):1-23. Available from: https://doi.org/10.3390/toxins12040255
https://doi.org/10.3390/toxins12040255...

27. Santos L, Oliveira C, Vasconcelos BM, Vilela D, Melo L, Ambrósio L, et al. Good management practices of venomous snakes in captivity to produce biological venom-based medicines: achieving replicability and contributing to pharmaceutical industry. J Toxicol Environ Health B Crit Rev. 2021;24(1):30-50. Available from: http://dx.doi.org/10.1080/10937404.2020.1855279
https://doi.org/10.1080/10937404.2020.18...
-2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
. Snake venoms are composed of a mixture of proteins, organic compounds, inorganic ions, carbohydrates, lipid fractions, and other substances44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,1414. El-Aziz TMA, Soares AG, Stockand JD. Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins. 2019;11(10):1-25. Available from: https://doi.org/10.3390/toxins11100564
https://doi.org/10.3390/toxins11100564...
,1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,1717. Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B. Anticancer potential of animal venoms and toxins. Indian J Exp Biol. 2010;48(2):93-103.,2020. Vonk FJ, Jackson K, Doley R, Madaras F, Mirtschin PJ, Vidal N. Snake venom: From fieldwork to the clinic: Recent insights into snake biology, together with new technology allowing high-throughput screening of venom, bring new hope for drug discovery. Bioessays. 2011;33(4):269-79. Available from: http://dx.doi.org/10.1002/bies.201000117
https://doi.org/10.1002/bies.201000117...
,2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,2727. Santos L, Oliveira C, Vasconcelos BM, Vilela D, Melo L, Ambrósio L, et al. Good management practices of venomous snakes in captivity to produce biological venom-based medicines: achieving replicability and contributing to pharmaceutical industry. J Toxicol Environ Health B Crit Rev. 2021;24(1):30-50. Available from: http://dx.doi.org/10.1080/10937404.2020.1855279
https://doi.org/10.1080/10937404.2020.18...
,2929. Vyas VK, Brahmbhatt K, Bhatt H, Parmar U. Therapeutic potential of snake venom in cancer therapy: current perspectives. Asian Pac J Trop Biomed. 2013;3(2):156-62. Available from: http://dx.doi.org/10.1016/S2221-1691(13)60042-8
https://doi.org/10.1016/S2221-1691(13)60...
,3030. Shanbhag VKL. Applications of snake venoms in treatment of cancer. Asian Pac J Trop Biomed . 2015;5(4):275-6. Available from: http://dx.doi.org/10.1016/S2221-1691(15)30344-0
https://doi.org/10.1016/S2221-1691(15)30...
.

Proteins account for approximately 90% of the dry weight of snake venom44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,2929. Vyas VK, Brahmbhatt K, Bhatt H, Parmar U. Therapeutic potential of snake venom in cancer therapy: current perspectives. Asian Pac J Trop Biomed. 2013;3(2):156-62. Available from: http://dx.doi.org/10.1016/S2221-1691(13)60042-8
https://doi.org/10.1016/S2221-1691(13)60...
,3131. Devi A. The Protein and Nonprotein Constituents of Snake Venoms. In: Bucherl W, Buckley EE, Deulofeu V, editors. Venomous Animals and their Venoms. 1st ed. New York: Academic Press; 1968. p. 119-65. Available from: https://doi.org/10.1016/B978-1-4832-2949-2.50014-X
https://doi.org/10.1016/B978-1-4832-2949...
,3232. Sarkar NK, Devi A. Enzymes in Snake Venoms. In: Bucherl W, Buckley EE, Deulofeu V, editors. Venomous Animals and their Venoms. 1st ed. New York: Academic Press ; 1968. p. 167-216. Available from: https://doi.org/10.1016/B978-1-4832-2949-2.50015-1
https://doi.org/10.1016/B978-1-4832-2949...
. C. d. terrificus (Cdt) venom is mainly composed of phospholipase A2 (PLA2), serinoproteases, hyaluronidases, L-amino acid oxidases, peptides, low molecular weight organic compounds, inorganic ions, and enzyme inhibitors44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,3333. Fusco LS, Neto EB, Francisco AF, Alfonso J, Soares A, Pimenta DC, et al. Fast venomic analysis of Crotalus durissus terrificus from northeastern Argentina. Toxicon X. 2020;7:100047. Available from: https://doi.org/10.1016/j.toxcx.2020.100047
https://doi.org/10.1016/j.toxcx.2020.100...
. The main toxins found in Cdt venom are Crotoxin, Convulxin, Gyroxin, and Crotamine44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,66. Frare BT, Resende YKS, Dornelas BC, Jorge MT, Ricarte VAS, Alves LM, et al. Clinical, Laboratory, and Therapeutic Aspects of Crotalus durissus (South American Rattlesnake) Victims: A Literature Review. Biomed Res Int. 2019; Article ID 1345923:1-7. Available from: https://doi.org/10.1155/2019/1345923
https://doi.org/10.1155/2019/1345923...

7. Tasima LJ, Serino-Silva C, Hatakeyama DM, Nishiduka ES, Tashima AK, Sant’Anna SS, et al. Crotamine in Crotalus durissus: distribution according to subspecies and geographic origin, in captivity or nature. J Venom Anim Toxins Incl Trop Dis. 2020;26:e20190053. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2019-0053
https://doi.org/10.1590/1678-9199-jvatit...
-88. Rangel-Santos A, Dos-Santos EC, Lopes-Ferreira M, Lima C, Cardoso DF, Mota I. A comparative study of biological activities of crotoxin and CB fraction of venoms from Crotalus durissus terrificus, Crotalus durissus cascavella and Crotalus durissus collilineatus. Toxicon. 2004;43(7):801-10. Available from: https://doi.org/10.1016/j.toxicon.2004.03.011.
https://doi.org/10.1016/j.toxicon.2004.0...
,3434. Lourenço Jr A, Creste CFZ, Barros LC, dos Santos LC, Pimenta DC, Barraviera B, et al. Individual venom profiling of Crotalus durissus terrificus specimens from a geographically limited region: Crotamine assessment and captivity evaluation on the biological activities. Toxicon. 2013;69:75-81. Available from: https://doi.org/10.1016/j.toxicon.2013.01.006
https://doi.org/10.1016/j.toxicon.2013.0...

35. Montoni F, Andreotti DZ, Eichler RAS, Santos WS, Kisaki CY, Arcos SSS, et al. The impact of rattlesnake venom on mice cerebellum proteomics points to synaptic inhibition and tissue damage. J Proteomics. 2020;221:103779. Available from: https://doi.org/10.1016/j.jprot.2020.103779
https://doi.org/10.1016/j.jprot.2020.103...

36. Lima TS, Neves CL, Zambelli VO, Lopes F, Sampaio SC, Cirillo MC. Crotoxin, a rattlesnake toxin, down-modulates functions of bone marrow neutrophils and impairs the Syk-GTPase pathway. Toxicon. 2017;136:44-55. Available from: https://doi.org/10.1016/j.toxicon.2017.07.002
https://doi.org/10.1016/j.toxicon.2017.0...

37. Tasima LJ, Hatakeyama DM, Serino-Silva C, Rodrigues C, de Lima E, Sant'Anna SS, et al. Comparative proteomic profiling and functional characterization of venom pooled from captive Crotalus durissus terrificus specimens and the Brazilian crotalic reference venom. Toxicon. 2020;185:26-35. Available from: https://doi.org/10.1016/j.toxicon.2020.07.001
https://doi.org/10.1016/j.toxicon.2020.0...
-3838. Corin RE, Viskatis LJ, Vidal JC, Etcheverry MA. Cytotoxicity of crotoxin on murine erythroleukemia cells in vitro. Invest New Drugs. 1993;11(1):11-5. Available from: https://doi.org/10.1007/BF00873905
https://doi.org/10.1007/BF00873905...
. This venom also contains more than 60 different protein families2323. Melani RD, Araujo GDT, Carvalho PC, Goto L, Nogueira FCS, Junqueira M, et al. Seeing beyond the tip of the icebeg: A deep analysis of the venome of the Brazilian Rattlesnake, Crotalus durissus terrificus. EuPA Open Proteom. 2015;8:144-56. Available from: https://doi.org/10.1016/j.euprot.2015.05.006
https://doi.org/10.1016/j.euprot.2015.05...
. Envenomation generates local manifestations of pain, edema, erythema, paresthesia, and systemic manifestations such as eyelid ptosis, facial muscle paralysis, and myoglobinuria, among other clinical signs44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,66. Frare BT, Resende YKS, Dornelas BC, Jorge MT, Ricarte VAS, Alves LM, et al. Clinical, Laboratory, and Therapeutic Aspects of Crotalus durissus (South American Rattlesnake) Victims: A Literature Review. Biomed Res Int. 2019; Article ID 1345923:1-7. Available from: https://doi.org/10.1155/2019/1345923
https://doi.org/10.1155/2019/1345923...
,2525. Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, et al. Secondary homeostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus. Int J Biol Macromol. 2019;131:127-33. Available from: https://doi.org/10.1016/j.ijbiomac.2019.03.059
https://doi.org/10.1016/j.ijbiomac.2019....
,3131. Devi A. The Protein and Nonprotein Constituents of Snake Venoms. In: Bucherl W, Buckley EE, Deulofeu V, editors. Venomous Animals and their Venoms. 1st ed. New York: Academic Press; 1968. p. 119-65. Available from: https://doi.org/10.1016/B978-1-4832-2949-2.50014-X
https://doi.org/10.1016/B978-1-4832-2949...
,3535. Montoni F, Andreotti DZ, Eichler RAS, Santos WS, Kisaki CY, Arcos SSS, et al. The impact of rattlesnake venom on mice cerebellum proteomics points to synaptic inhibition and tissue damage. J Proteomics. 2020;221:103779. Available from: https://doi.org/10.1016/j.jprot.2020.103779
https://doi.org/10.1016/j.jprot.2020.103...
,3939. Bordon KCF, Perino MG, Giglio JR, Arantes EC. Isolation, enzymatic characterization and antiedematogenic activity of the first reported rattlesnake hyaluronidase from Crotalus durissus terrificus venom. Biochimie, 2012;94(12):2740-8. Available from: https://doi.org/10.1016/j.biochi.2012.08.014.
https://doi.org/10.1016/j.biochi.2012.08...
, because of its neurotoxic, coagulant, and myotoxic actions44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,2525. Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, et al. Secondary homeostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus. Int J Biol Macromol. 2019;131:127-33. Available from: https://doi.org/10.1016/j.ijbiomac.2019.03.059
https://doi.org/10.1016/j.ijbiomac.2019....
,3131. Devi A. The Protein and Nonprotein Constituents of Snake Venoms. In: Bucherl W, Buckley EE, Deulofeu V, editors. Venomous Animals and their Venoms. 1st ed. New York: Academic Press; 1968. p. 119-65. Available from: https://doi.org/10.1016/B978-1-4832-2949-2.50014-X
https://doi.org/10.1016/B978-1-4832-2949...
,3333. Fusco LS, Neto EB, Francisco AF, Alfonso J, Soares A, Pimenta DC, et al. Fast venomic analysis of Crotalus durissus terrificus from northeastern Argentina. Toxicon X. 2020;7:100047. Available from: https://doi.org/10.1016/j.toxcx.2020.100047
https://doi.org/10.1016/j.toxcx.2020.100...
,3535. Montoni F, Andreotti DZ, Eichler RAS, Santos WS, Kisaki CY, Arcos SSS, et al. The impact of rattlesnake venom on mice cerebellum proteomics points to synaptic inhibition and tissue damage. J Proteomics. 2020;221:103779. Available from: https://doi.org/10.1016/j.jprot.2020.103779
https://doi.org/10.1016/j.jprot.2020.103...
.

There is a wide variety of pharmacological uses of the different fractions of Crotalus sp. venom, including antifungal, antiviral, antibacterial, antitumor, and antiprotozoal activities44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,1515. Almeida JR, Resende LM, Watanabe RK, Carregari VC, Huancahuire-Vega S, Caldeira CAS, et al. Snake Venom Peptides and Low Mass Proteins: Molecular Tools and Therapeutic Agents. Curr Med Chem. 2017;24(30):3254-82. Available from: https://doi.org/10.2174/0929867323666161028155611
https://doi.org/10.2174/0929867323666161...
,2626. Pérez-Peinado C, Defaus S, Andreu D. Hitchhiking with Nature: Snake Venom Peptides to Fight Cancer and Superbugs. Toxins. 2020;12(4):1-23. Available from: https://doi.org/10.3390/toxins12040255
https://doi.org/10.3390/toxins12040255...

27. Santos L, Oliveira C, Vasconcelos BM, Vilela D, Melo L, Ambrósio L, et al. Good management practices of venomous snakes in captivity to produce biological venom-based medicines: achieving replicability and contributing to pharmaceutical industry. J Toxicol Environ Health B Crit Rev. 2021;24(1):30-50. Available from: http://dx.doi.org/10.1080/10937404.2020.1855279
https://doi.org/10.1080/10937404.2020.18...
-2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,3737. Tasima LJ, Hatakeyama DM, Serino-Silva C, Rodrigues C, de Lima E, Sant'Anna SS, et al. Comparative proteomic profiling and functional characterization of venom pooled from captive Crotalus durissus terrificus specimens and the Brazilian crotalic reference venom. Toxicon. 2020;185:26-35. Available from: https://doi.org/10.1016/j.toxicon.2020.07.001
https://doi.org/10.1016/j.toxicon.2020.0...
,4040. Faure G, Bakouh N, Lourdel S, Odolczyk N, Premchandar A, Servel N, et al. Rattlesnake Phospholipase A2 increases CFTR-chloride channel current and corrects F508CFTR dysfunction: impact in Cystic Fibrosis. J Mol Biol. 2016;428(14):2898-915. Available from: http://dx.doi.org/10.1016/j.jmb.2016.05.016
https://doi.org/10.1016/j.jmb.2016.05.01...

41. Marcussi S, Santos PRS, Menaldo DL, Silveira LB, Santos-Filho NA, Mazzi MV, et al. Evaluation of the genotoxicity of Crotalus durissus terrificus snake venom and its isolated toxins on human lymphocytes. Mutat Res. 2011;724(1-2):59-63. Available from: https://doi.org/10.1016/j.mrgentox.2011.06.004
https://doi.org/10.1016/j.mrgentox.2011....

42. Muller VDM, Russo RR, Cintra ACO, Sartim MA, Alves-Paiva RM, Figueiredo LTM, et al. Crotoxin and phospholipases A₂ from Crotalus durissus terrificus showed antiviral activity against dengue and yellow fever viruses. Toxicon. 2012;59(4):507-15. Available from: https://doi.org/10.1016/j.toxicon.2011.05.021
https://doi.org/10.1016/j.toxicon.2011.0...
-4343. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins Incl Trop Dis . 2018;24:1-13. Available from: https://doi.org/10.1186/s40409-018-0178-3.
https://doi.org/10.1186/s40409-018-0178-...
.

CROTOXIN

Crotoxin represents approximately 40%-60% of the dry weight of the Cdt venom44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,88. Rangel-Santos A, Dos-Santos EC, Lopes-Ferreira M, Lima C, Cardoso DF, Mota I. A comparative study of biological activities of crotoxin and CB fraction of venoms from Crotalus durissus terrificus, Crotalus durissus cascavella and Crotalus durissus collilineatus. Toxicon. 2004;43(7):801-10. Available from: https://doi.org/10.1016/j.toxicon.2004.03.011.
https://doi.org/10.1016/j.toxicon.2004.0...
,1919. Soares MA, Pujatti PB, Fortes-Dias CI, Antonelli L, Santos RG. Crotalus durissus terrificus venom as a source of antitumoral agents. J Venom Anim Toxins Incl Trop Dis . 2010;16(3):480-92. Available from: https://doi.org/10.1590/S1678-91992010000300015
https://doi.org/10.1590/S1678-9199201000...
,3333. Fusco LS, Neto EB, Francisco AF, Alfonso J, Soares A, Pimenta DC, et al. Fast venomic analysis of Crotalus durissus terrificus from northeastern Argentina. Toxicon X. 2020;7:100047. Available from: https://doi.org/10.1016/j.toxcx.2020.100047
https://doi.org/10.1016/j.toxcx.2020.100...
,3636. Lima TS, Neves CL, Zambelli VO, Lopes F, Sampaio SC, Cirillo MC. Crotoxin, a rattlesnake toxin, down-modulates functions of bone marrow neutrophils and impairs the Syk-GTPase pathway. Toxicon. 2017;136:44-55. Available from: https://doi.org/10.1016/j.toxicon.2017.07.002
https://doi.org/10.1016/j.toxicon.2017.0...
,4242. Muller VDM, Russo RR, Cintra ACO, Sartim MA, Alves-Paiva RM, Figueiredo LTM, et al. Crotoxin and phospholipases A₂ from Crotalus durissus terrificus showed antiviral activity against dengue and yellow fever viruses. Toxicon. 2012;59(4):507-15. Available from: https://doi.org/10.1016/j.toxicon.2011.05.021
https://doi.org/10.1016/j.toxicon.2011.0...

43. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins Incl Trop Dis . 2018;24:1-13. Available from: https://doi.org/10.1186/s40409-018-0178-3.
https://doi.org/10.1186/s40409-018-0178-...
-4444. Pimenta LA, Almeida MES, Bretones ML, Cirillo MC, Curi R, Sampaio SC. Crotoxin promotes macrophage reprogramming towards an antiangiogenic phenotype. Sci Rep. 2019;9(1):1-15. Available from: https://doi.org/10.1038/s41598-019-40903-0
https://doi.org/10.1038/s41598-019-40903...
and is a potent neurotoxin formed by PLA2 and crotapotin, forming a complex of high toxicity44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,88. Rangel-Santos A, Dos-Santos EC, Lopes-Ferreira M, Lima C, Cardoso DF, Mota I. A comparative study of biological activities of crotoxin and CB fraction of venoms from Crotalus durissus terrificus, Crotalus durissus cascavella and Crotalus durissus collilineatus. Toxicon. 2004;43(7):801-10. Available from: https://doi.org/10.1016/j.toxicon.2004.03.011.
https://doi.org/10.1016/j.toxicon.2004.0...
,3232. Sarkar NK, Devi A. Enzymes in Snake Venoms. In: Bucherl W, Buckley EE, Deulofeu V, editors. Venomous Animals and their Venoms. 1st ed. New York: Academic Press ; 1968. p. 167-216. Available from: https://doi.org/10.1016/B978-1-4832-2949-2.50015-1
https://doi.org/10.1016/B978-1-4832-2949...
,3838. Corin RE, Viskatis LJ, Vidal JC, Etcheverry MA. Cytotoxicity of crotoxin on murine erythroleukemia cells in vitro. Invest New Drugs. 1993;11(1):11-5. Available from: https://doi.org/10.1007/BF00873905
https://doi.org/10.1007/BF00873905...
,4242. Muller VDM, Russo RR, Cintra ACO, Sartim MA, Alves-Paiva RM, Figueiredo LTM, et al. Crotoxin and phospholipases A₂ from Crotalus durissus terrificus showed antiviral activity against dengue and yellow fever viruses. Toxicon. 2012;59(4):507-15. Available from: https://doi.org/10.1016/j.toxicon.2011.05.021
https://doi.org/10.1016/j.toxicon.2011.0...

43. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins Incl Trop Dis . 2018;24:1-13. Available from: https://doi.org/10.1186/s40409-018-0178-3.
https://doi.org/10.1186/s40409-018-0178-...

44. Pimenta LA, Almeida MES, Bretones ML, Cirillo MC, Curi R, Sampaio SC. Crotoxin promotes macrophage reprogramming towards an antiangiogenic phenotype. Sci Rep. 2019;9(1):1-15. Available from: https://doi.org/10.1038/s41598-019-40903-0
https://doi.org/10.1038/s41598-019-40903...

45. Silva-Júnior LN, Abreu LS, Rodrigues CFB, Galizio NC, Aguiar WS, Serino-Silva C, et al. Geographic variation of individual venom profile of Crotalus durissus snakes. J Venom Anim Toxins Incl Trop Dis . 2020;26:e20200016. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2020-0016
https://doi.org/10.1590/1678-9199-jvatit...

46. Sampaio SC, Brigatte P, Sousa-e-Silva MCC, dos-Santos EC, Rangel-Santos AC, Curi R, et al. Contribution of crotoxin for the inhibitory effect of Crotalus durissus terrificus snake venom on macrophage function. Toxicon. 2003;41(7):899-907. Available from: https://doi.org/10.1016/S0041-0101(03)00069-2.
https://doi.org/10.1016/S0041-0101(03)00...

47. Yang C, Yang Y, Qin Z, Gu Z, Reid P, Liang Z. Autophagy is involved in cytotoxic effects of crotoxin in human breast cancer cell line MCF-7 cells. Acta Pharmacol Sin. 2007;28(4):540-8. Available from: http://dx.doi.org/10.1111/j.1745-7254.2007.00530.x
https://doi.org/10.1111/j.1745-7254.2007...

48. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...

49. Ye B, Xie Y, Qin ZH, Wu JC, Han R, He JK. Anti-tumor activity of CrTX in human lung adenocarcinoma cell line A549. Acta Pharmacol Sinica . 2011;32(11):1397-401. Available from: https://doi.org/10.1038/aps.2011.116
https://doi.org/10.1038/aps.2011.116...

50. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...

51. Brigatte P, Faiad OJ, Nocelli RCF, Landgraf RG, Palma MS, Cury Y, et al. Walker 256 Tumor Growth Suppression by Crotoxin Involves Formyl Peptide Receptors and Lipoxin A₄. Mediators Inflamm. 2016;2016:2457532. Available from: http://dx.doi.org/10.1155/2016/2457532
https://doi.org/10.1155/2016/2457532...

52. Cavalcante WLG, Matos JBN, Timoteo MA, Fontes MRM, Gallacci M, de Sa PC. Neuromuscular paralysis by the basic phospholipase A2 subunit crotoxin from Crotalus durissus terrificus snake venom needs its acid chaperone to con-currently inhibit acetylcholine release and produce muscle blockage. Toxicol Appl Pharmacol. 2017;334:8-17. Available from: https://doi.org/10.1016/j.taap.2017.08.021
https://doi.org/10.1016/j.taap.2017.08.0...

53. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
-5454. Andrade CM, Rey FM, Cintra A, Sampaio SV, Torqueti MR. Effects of crotoxin, a neurotoxin from Crotalus durissus terrificus snake venom, on human endothelial cells. Int J Biol Macromol . 2019;134:613-21. Available from: https://doi.org/10.1016/j.ijbiomac.2019.05.019
https://doi.org/10.1016/j.ijbiomac.2019....
, and exhibits myotoxic, nephrotoxic, and cardiotoxic effects44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,3737. Tasima LJ, Hatakeyama DM, Serino-Silva C, Rodrigues C, de Lima E, Sant'Anna SS, et al. Comparative proteomic profiling and functional characterization of venom pooled from captive Crotalus durissus terrificus specimens and the Brazilian crotalic reference venom. Toxicon. 2020;185:26-35. Available from: https://doi.org/10.1016/j.toxicon.2020.07.001
https://doi.org/10.1016/j.toxicon.2020.0...
,3838. Corin RE, Viskatis LJ, Vidal JC, Etcheverry MA. Cytotoxicity of crotoxin on murine erythroleukemia cells in vitro. Invest New Drugs. 1993;11(1):11-5. Available from: https://doi.org/10.1007/BF00873905
https://doi.org/10.1007/BF00873905...
,4343. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins Incl Trop Dis . 2018;24:1-13. Available from: https://doi.org/10.1186/s40409-018-0178-3.
https://doi.org/10.1186/s40409-018-0178-...
,4444. Pimenta LA, Almeida MES, Bretones ML, Cirillo MC, Curi R, Sampaio SC. Crotoxin promotes macrophage reprogramming towards an antiangiogenic phenotype. Sci Rep. 2019;9(1):1-15. Available from: https://doi.org/10.1038/s41598-019-40903-0
https://doi.org/10.1038/s41598-019-40903...
,4646. Sampaio SC, Brigatte P, Sousa-e-Silva MCC, dos-Santos EC, Rangel-Santos AC, Curi R, et al. Contribution of crotoxin for the inhibitory effect of Crotalus durissus terrificus snake venom on macrophage function. Toxicon. 2003;41(7):899-907. Available from: https://doi.org/10.1016/S0041-0101(03)00069-2.
https://doi.org/10.1016/S0041-0101(03)00...
,4848. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...
,5050. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...
.

This neurotoxic action is mainly attributed to the inhibitory mechanism of acetylcholine release in presynaptic neurons4848. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...
,5252. Cavalcante WLG, Matos JBN, Timoteo MA, Fontes MRM, Gallacci M, de Sa PC. Neuromuscular paralysis by the basic phospholipase A2 subunit crotoxin from Crotalus durissus terrificus snake venom needs its acid chaperone to con-currently inhibit acetylcholine release and produce muscle blockage. Toxicol Appl Pharmacol. 2017;334:8-17. Available from: https://doi.org/10.1016/j.taap.2017.08.021
https://doi.org/10.1016/j.taap.2017.08.0...
,5454. Andrade CM, Rey FM, Cintra A, Sampaio SV, Torqueti MR. Effects of crotoxin, a neurotoxin from Crotalus durissus terrificus snake venom, on human endothelial cells. Int J Biol Macromol . 2019;134:613-21. Available from: https://doi.org/10.1016/j.ijbiomac.2019.05.019
https://doi.org/10.1016/j.ijbiomac.2019....
. Desensitization of postsynaptic nicotinic receptors is another mechanism that reduces the response to acetylcholine4848. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...
,5252. Cavalcante WLG, Matos JBN, Timoteo MA, Fontes MRM, Gallacci M, de Sa PC. Neuromuscular paralysis by the basic phospholipase A2 subunit crotoxin from Crotalus durissus terrificus snake venom needs its acid chaperone to con-currently inhibit acetylcholine release and produce muscle blockage. Toxicol Appl Pharmacol. 2017;334:8-17. Available from: https://doi.org/10.1016/j.taap.2017.08.021
https://doi.org/10.1016/j.taap.2017.08.0...
. Thus, crotoxin acts by blocking potassium channels and prolonging the action potential at neuromuscular junctions, thereby increasing calcium influx into the channels, mainly due to the presence and high activity of PLA2 in its composition88. Rangel-Santos A, Dos-Santos EC, Lopes-Ferreira M, Lima C, Cardoso DF, Mota I. A comparative study of biological activities of crotoxin and CB fraction of venoms from Crotalus durissus terrificus, Crotalus durissus cascavella and Crotalus durissus collilineatus. Toxicon. 2004;43(7):801-10. Available from: https://doi.org/10.1016/j.toxicon.2004.03.011.
https://doi.org/10.1016/j.toxicon.2004.0...
,4848. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...
,5252. Cavalcante WLG, Matos JBN, Timoteo MA, Fontes MRM, Gallacci M, de Sa PC. Neuromuscular paralysis by the basic phospholipase A2 subunit crotoxin from Crotalus durissus terrificus snake venom needs its acid chaperone to con-currently inhibit acetylcholine release and produce muscle blockage. Toxicol Appl Pharmacol. 2017;334:8-17. Available from: https://doi.org/10.1016/j.taap.2017.08.021
https://doi.org/10.1016/j.taap.2017.08.0...
.

Crotoxin has been widely studied for its immunomodulatory, anti-inflammatory, antitumor, antimicrobial, and analgesic actions44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,4040. Faure G, Bakouh N, Lourdel S, Odolczyk N, Premchandar A, Servel N, et al. Rattlesnake Phospholipase A2 increases CFTR-chloride channel current and corrects F508CFTR dysfunction: impact in Cystic Fibrosis. J Mol Biol. 2016;428(14):2898-915. Available from: http://dx.doi.org/10.1016/j.jmb.2016.05.016
https://doi.org/10.1016/j.jmb.2016.05.01...
,4343. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins Incl Trop Dis . 2018;24:1-13. Available from: https://doi.org/10.1186/s40409-018-0178-3.
https://doi.org/10.1186/s40409-018-0178-...
,4444. Pimenta LA, Almeida MES, Bretones ML, Cirillo MC, Curi R, Sampaio SC. Crotoxin promotes macrophage reprogramming towards an antiangiogenic phenotype. Sci Rep. 2019;9(1):1-15. Available from: https://doi.org/10.1038/s41598-019-40903-0
https://doi.org/10.1038/s41598-019-40903...
,4646. Sampaio SC, Brigatte P, Sousa-e-Silva MCC, dos-Santos EC, Rangel-Santos AC, Curi R, et al. Contribution of crotoxin for the inhibitory effect of Crotalus durissus terrificus snake venom on macrophage function. Toxicon. 2003;41(7):899-907. Available from: https://doi.org/10.1016/S0041-0101(03)00069-2.
https://doi.org/10.1016/S0041-0101(03)00...
,4848. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...
,5050. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...

51. Brigatte P, Faiad OJ, Nocelli RCF, Landgraf RG, Palma MS, Cury Y, et al. Walker 256 Tumor Growth Suppression by Crotoxin Involves Formyl Peptide Receptors and Lipoxin A₄. Mediators Inflamm. 2016;2016:2457532. Available from: http://dx.doi.org/10.1155/2016/2457532
https://doi.org/10.1155/2016/2457532...

52. Cavalcante WLG, Matos JBN, Timoteo MA, Fontes MRM, Gallacci M, de Sa PC. Neuromuscular paralysis by the basic phospholipase A2 subunit crotoxin from Crotalus durissus terrificus snake venom needs its acid chaperone to con-currently inhibit acetylcholine release and produce muscle blockage. Toxicol Appl Pharmacol. 2017;334:8-17. Available from: https://doi.org/10.1016/j.taap.2017.08.021
https://doi.org/10.1016/j.taap.2017.08.0...

53. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
-5454. Andrade CM, Rey FM, Cintra A, Sampaio SV, Torqueti MR. Effects of crotoxin, a neurotoxin from Crotalus durissus terrificus snake venom, on human endothelial cells. Int J Biol Macromol . 2019;134:613-21. Available from: https://doi.org/10.1016/j.ijbiomac.2019.05.019
https://doi.org/10.1016/j.ijbiomac.2019....
. In vivo studies have demonstrated its ability to inhibit the production of pro-inflammatory and anti-inflammatory cytokines from the injection of the toxin in rats, including IL-10, IL-4, IL-6, and tumor necrosis factor3636. Lima TS, Neves CL, Zambelli VO, Lopes F, Sampaio SC, Cirillo MC. Crotoxin, a rattlesnake toxin, down-modulates functions of bone marrow neutrophils and impairs the Syk-GTPase pathway. Toxicon. 2017;136:44-55. Available from: https://doi.org/10.1016/j.toxicon.2017.07.002
https://doi.org/10.1016/j.toxicon.2017.0...
,4343. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins Incl Trop Dis . 2018;24:1-13. Available from: https://doi.org/10.1186/s40409-018-0178-3.
https://doi.org/10.1186/s40409-018-0178-...
. This immunomodulatory activity may be associated with the production of anti-inflammatory mediators via the lipoxygenase pathway, such as lipoxin A4 (LXA4), and the activation of formyl peptide receptors, in addition to its regulatory role in macrophages3636. Lima TS, Neves CL, Zambelli VO, Lopes F, Sampaio SC, Cirillo MC. Crotoxin, a rattlesnake toxin, down-modulates functions of bone marrow neutrophils and impairs the Syk-GTPase pathway. Toxicon. 2017;136:44-55. Available from: https://doi.org/10.1016/j.toxicon.2017.07.002
https://doi.org/10.1016/j.toxicon.2017.0...
,4343. Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins Incl Trop Dis . 2018;24:1-13. Available from: https://doi.org/10.1186/s40409-018-0178-3.
https://doi.org/10.1186/s40409-018-0178-...
,4444. Pimenta LA, Almeida MES, Bretones ML, Cirillo MC, Curi R, Sampaio SC. Crotoxin promotes macrophage reprogramming towards an antiangiogenic phenotype. Sci Rep. 2019;9(1):1-15. Available from: https://doi.org/10.1038/s41598-019-40903-0
https://doi.org/10.1038/s41598-019-40903...
,5151. Brigatte P, Faiad OJ, Nocelli RCF, Landgraf RG, Palma MS, Cury Y, et al. Walker 256 Tumor Growth Suppression by Crotoxin Involves Formyl Peptide Receptors and Lipoxin A₄. Mediators Inflamm. 2016;2016:2457532. Available from: http://dx.doi.org/10.1155/2016/2457532
https://doi.org/10.1155/2016/2457532...
.

In vitro and in vivo studies have described activating mechanisms of cell apoptosis in different cancer cell lines1919. Soares MA, Pujatti PB, Fortes-Dias CI, Antonelli L, Santos RG. Crotalus durissus terrificus venom as a source of antitumoral agents. J Venom Anim Toxins Incl Trop Dis . 2010;16(3):480-92. Available from: https://doi.org/10.1590/S1678-91992010000300015
https://doi.org/10.1590/S1678-9199201000...
,4747. Yang C, Yang Y, Qin Z, Gu Z, Reid P, Liang Z. Autophagy is involved in cytotoxic effects of crotoxin in human breast cancer cell line MCF-7 cells. Acta Pharmacol Sin. 2007;28(4):540-8. Available from: http://dx.doi.org/10.1111/j.1745-7254.2007.00530.x
https://doi.org/10.1111/j.1745-7254.2007...

48. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...

49. Ye B, Xie Y, Qin ZH, Wu JC, Han R, He JK. Anti-tumor activity of CrTX in human lung adenocarcinoma cell line A549. Acta Pharmacol Sinica . 2011;32(11):1397-401. Available from: https://doi.org/10.1038/aps.2011.116
https://doi.org/10.1038/aps.2011.116...

50. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...
-5151. Brigatte P, Faiad OJ, Nocelli RCF, Landgraf RG, Palma MS, Cury Y, et al. Walker 256 Tumor Growth Suppression by Crotoxin Involves Formyl Peptide Receptors and Lipoxin A₄. Mediators Inflamm. 2016;2016:2457532. Available from: http://dx.doi.org/10.1155/2016/2457532
https://doi.org/10.1155/2016/2457532...
,5353. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
,5555. He J, Wu X, Wang Y, Han R, Qin Z, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta pharmacol Sin. 2013;34(2):295-300. Available from: https://doi.org/10.1038/aps.2012.156
https://doi.org/10.1038/aps.2012.156...
induced by cellular autophagy mechanisms4747. Yang C, Yang Y, Qin Z, Gu Z, Reid P, Liang Z. Autophagy is involved in cytotoxic effects of crotoxin in human breast cancer cell line MCF-7 cells. Acta Pharmacol Sin. 2007;28(4):540-8. Available from: http://dx.doi.org/10.1111/j.1745-7254.2007.00530.x
https://doi.org/10.1111/j.1745-7254.2007...
,5353. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
. Both cell death pathways activated by crotoxin (apoptosis and autophagy) can occur simultaneously or sequentially through mechanisms such as changes in mitochondrial membrane potential and release of intracellular cytochrome C. Another important factor related to the mechanism of action of crotoxin is its apparent selectivity for cells with high expression of epidermal growth factor receptors (EGFR) 1919. Soares MA, Pujatti PB, Fortes-Dias CI, Antonelli L, Santos RG. Crotalus durissus terrificus venom as a source of antitumoral agents. J Venom Anim Toxins Incl Trop Dis . 2010;16(3):480-92. Available from: https://doi.org/10.1590/S1678-91992010000300015
https://doi.org/10.1590/S1678-9199201000...
,2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,4747. Yang C, Yang Y, Qin Z, Gu Z, Reid P, Liang Z. Autophagy is involved in cytotoxic effects of crotoxin in human breast cancer cell line MCF-7 cells. Acta Pharmacol Sin. 2007;28(4):540-8. Available from: http://dx.doi.org/10.1111/j.1745-7254.2007.00530.x
https://doi.org/10.1111/j.1745-7254.2007...
,5050. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...
,5656. Donato NJ, Martin CA, Perez M, Newman RA, Vidal JC, Etcheverry M. Regulation of epidermal growth factor receptor activity by crotoxin, a snake venom phospholipase A2 toxin. A novel growth inhibitory mechanism. Biochem pharmacol. 1996;51(11):1535-43. Available from: https://doi.org/10.1016/0006-2952(96)00097-4
https://doi.org/10.1016/0006-2952(96)000...
.

The cytotoxic action on glioblastoma and benign pituitary adenoma cells was partially attributed to crotoxin, which is also cytotoxic to human mammary duct carcinoma and human lung adenocarcinoma cell lines44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,1919. Soares MA, Pujatti PB, Fortes-Dias CI, Antonelli L, Santos RG. Crotalus durissus terrificus venom as a source of antitumoral agents. J Venom Anim Toxins Incl Trop Dis . 2010;16(3):480-92. Available from: https://doi.org/10.1590/S1678-91992010000300015
https://doi.org/10.1590/S1678-9199201000...
,4747. Yang C, Yang Y, Qin Z, Gu Z, Reid P, Liang Z. Autophagy is involved in cytotoxic effects of crotoxin in human breast cancer cell line MCF-7 cells. Acta Pharmacol Sin. 2007;28(4):540-8. Available from: http://dx.doi.org/10.1111/j.1745-7254.2007.00530.x
https://doi.org/10.1111/j.1745-7254.2007...

48. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...

49. Ye B, Xie Y, Qin ZH, Wu JC, Han R, He JK. Anti-tumor activity of CrTX in human lung adenocarcinoma cell line A549. Acta Pharmacol Sinica . 2011;32(11):1397-401. Available from: https://doi.org/10.1038/aps.2011.116
https://doi.org/10.1038/aps.2011.116...

50. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...
-5151. Brigatte P, Faiad OJ, Nocelli RCF, Landgraf RG, Palma MS, Cury Y, et al. Walker 256 Tumor Growth Suppression by Crotoxin Involves Formyl Peptide Receptors and Lipoxin A₄. Mediators Inflamm. 2016;2016:2457532. Available from: http://dx.doi.org/10.1155/2016/2457532
https://doi.org/10.1155/2016/2457532...
,5555. He J, Wu X, Wang Y, Han R, Qin Z, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta pharmacol Sin. 2013;34(2):295-300. Available from: https://doi.org/10.1038/aps.2012.156
https://doi.org/10.1038/aps.2012.156...
,5757. Han R, Liang H, Qin ZH, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin . 2014;35(10):1323-32. Available from: https://doi.org/10.1038/aps.2014.62
https://doi.org/10.1038/aps.2014.62...
. The application of portions of the toxin to murine erythroleukemia cells demonstrated the potential to reduce the viability of the strain3838. Corin RE, Viskatis LJ, Vidal JC, Etcheverry MA. Cytotoxicity of crotoxin on murine erythroleukemia cells in vitro. Invest New Drugs. 1993;11(1):11-5. Available from: https://doi.org/10.1007/BF00873905
https://doi.org/10.1007/BF00873905...
. To observe cytotoxicity, the B subunit of crotoxin was separated from PLA2 and used alone3838. Corin RE, Viskatis LJ, Vidal JC, Etcheverry MA. Cytotoxicity of crotoxin on murine erythroleukemia cells in vitro. Invest New Drugs. 1993;11(1):11-5. Available from: https://doi.org/10.1007/BF00873905
https://doi.org/10.1007/BF00873905...
.

The isolated crotoxin is cytotoxic to different cell lines, with different cell response5353. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
. The mechanisms evaluated involved changes in the mitochondrial membrane potential, release of cytochrome C, and activation of caspase-3, a protease essential for the process of cell apoptosis4747. Yang C, Yang Y, Qin Z, Gu Z, Reid P, Liang Z. Autophagy is involved in cytotoxic effects of crotoxin in human breast cancer cell line MCF-7 cells. Acta Pharmacol Sin. 2007;28(4):540-8. Available from: http://dx.doi.org/10.1111/j.1745-7254.2007.00530.x
https://doi.org/10.1111/j.1745-7254.2007...

48. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...

49. Ye B, Xie Y, Qin ZH, Wu JC, Han R, He JK. Anti-tumor activity of CrTX in human lung adenocarcinoma cell line A549. Acta Pharmacol Sinica . 2011;32(11):1397-401. Available from: https://doi.org/10.1038/aps.2011.116
https://doi.org/10.1038/aps.2011.116...
-5050. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...
,5252. Cavalcante WLG, Matos JBN, Timoteo MA, Fontes MRM, Gallacci M, de Sa PC. Neuromuscular paralysis by the basic phospholipase A2 subunit crotoxin from Crotalus durissus terrificus snake venom needs its acid chaperone to con-currently inhibit acetylcholine release and produce muscle blockage. Toxicol Appl Pharmacol. 2017;334:8-17. Available from: https://doi.org/10.1016/j.taap.2017.08.021
https://doi.org/10.1016/j.taap.2017.08.0...
,5353. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
,5555. He J, Wu X, Wang Y, Han R, Qin Z, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta pharmacol Sin. 2013;34(2):295-300. Available from: https://doi.org/10.1038/aps.2012.156
https://doi.org/10.1038/aps.2012.156...
. Furthermore, it was possible to conclude that the toxin did not interfere with the viability of keratinocytes, which are highly affected by current antineoplastic therapies5353. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
.

Crotoxin provokes possible damage to the cellular DNA of PANC-1 cells, associated with pancreatic tumors, by upregulating protein expression5353. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
. DNA damage has also been observed in glioma cell lines, leading to an increase in the percentage of cells undergoing apoptosis. Some in vitro studies have also reported a higher percentage of apoptosis among SK-MES-1 cells, a lung cancer cell line, in addition to damage such as nuclear condensation and fragmentation5050. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...
,5757. Han R, Liang H, Qin ZH, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin . 2014;35(10):1323-32. Available from: https://doi.org/10.1038/aps.2014.62
https://doi.org/10.1038/aps.2014.62...
.

When associated with tyrosine kinase inhibitors, crotoxin potentiates the antitumor effect of the drug against lung tumor cell lines5050. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...
,5353. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
,5757. Han R, Liang H, Qin ZH, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin . 2014;35(10):1323-32. Available from: https://doi.org/10.1038/aps.2014.62
https://doi.org/10.1038/aps.2014.62...
. In a dose-dependent manner, the toxin prevents DNA synthesis and interrupts the cell cycle in the S phase, suppressing the proliferation of SK-MES-1 cells both in vitro and in vivo5252. Cavalcante WLG, Matos JBN, Timoteo MA, Fontes MRM, Gallacci M, de Sa PC. Neuromuscular paralysis by the basic phospholipase A2 subunit crotoxin from Crotalus durissus terrificus snake venom needs its acid chaperone to con-currently inhibit acetylcholine release and produce muscle blockage. Toxicol Appl Pharmacol. 2017;334:8-17. Available from: https://doi.org/10.1016/j.taap.2017.08.021
https://doi.org/10.1016/j.taap.2017.08.0...
,5757. Han R, Liang H, Qin ZH, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin . 2014;35(10):1323-32. Available from: https://doi.org/10.1038/aps.2014.62
https://doi.org/10.1038/aps.2014.62...
. One of the mechanisms identified was the increased expression and cleavage of caspase-3, which is responsible for inducing cell apoptosis5050. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...
,5757. Han R, Liang H, Qin ZH, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin . 2014;35(10):1323-32. Available from: https://doi.org/10.1038/aps.2014.62
https://doi.org/10.1038/aps.2014.62...
. Another mechanism observed was the induction of cytochrome C release, which increased the occurrence of cellular autophagy, a mechanism also observed in MCF-7 breast cancer lines4747. Yang C, Yang Y, Qin Z, Gu Z, Reid P, Liang Z. Autophagy is involved in cytotoxic effects of crotoxin in human breast cancer cell line MCF-7 cells. Acta Pharmacol Sin. 2007;28(4):540-8. Available from: http://dx.doi.org/10.1111/j.1745-7254.2007.00530.x
https://doi.org/10.1111/j.1745-7254.2007...
,4949. Ye B, Xie Y, Qin ZH, Wu JC, Han R, He JK. Anti-tumor activity of CrTX in human lung adenocarcinoma cell line A549. Acta Pharmacol Sinica . 2011;32(11):1397-401. Available from: https://doi.org/10.1038/aps.2011.116
https://doi.org/10.1038/aps.2011.116...
,5353. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
.

Crotoxin also induces the release of LXA4, pro-inflammatory eicosanoid lipoxin, and its analogs through the induction of its synthesis in macrophages3636. Lima TS, Neves CL, Zambelli VO, Lopes F, Sampaio SC, Cirillo MC. Crotoxin, a rattlesnake toxin, down-modulates functions of bone marrow neutrophils and impairs the Syk-GTPase pathway. Toxicon. 2017;136:44-55. Available from: https://doi.org/10.1016/j.toxicon.2017.07.002
https://doi.org/10.1016/j.toxicon.2017.0...
,4444. Pimenta LA, Almeida MES, Bretones ML, Cirillo MC, Curi R, Sampaio SC. Crotoxin promotes macrophage reprogramming towards an antiangiogenic phenotype. Sci Rep. 2019;9(1):1-15. Available from: https://doi.org/10.1038/s41598-019-40903-0
https://doi.org/10.1038/s41598-019-40903...
,4646. Sampaio SC, Brigatte P, Sousa-e-Silva MCC, dos-Santos EC, Rangel-Santos AC, Curi R, et al. Contribution of crotoxin for the inhibitory effect of Crotalus durissus terrificus snake venom on macrophage function. Toxicon. 2003;41(7):899-907. Available from: https://doi.org/10.1016/S0041-0101(03)00069-2.
https://doi.org/10.1016/S0041-0101(03)00...
,4848. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...
,5151. Brigatte P, Faiad OJ, Nocelli RCF, Landgraf RG, Palma MS, Cury Y, et al. Walker 256 Tumor Growth Suppression by Crotoxin Involves Formyl Peptide Receptors and Lipoxin A₄. Mediators Inflamm. 2016;2016:2457532. Available from: http://dx.doi.org/10.1155/2016/2457532
https://doi.org/10.1155/2016/2457532...
. In vivo studies of Walker 256 carcinoma cells concluded that this mechanism is responsible for the antineoplastic action of crotoxin on the lineage, and the concentration of lipoxin was 74% higher in the plasma of animals treated with crotoxin than in those treated with saline solution5151. Brigatte P, Faiad OJ, Nocelli RCF, Landgraf RG, Palma MS, Cury Y, et al. Walker 256 Tumor Growth Suppression by Crotoxin Involves Formyl Peptide Receptors and Lipoxin A₄. Mediators Inflamm. 2016;2016:2457532. Available from: http://dx.doi.org/10.1155/2016/2457532
https://doi.org/10.1155/2016/2457532...
. Lipoxins have been shown to be antineoplastic owing to their ability to inhibit tumor growth by inhibiting endothelial cell proliferation and reducing the production of angiogenic growth factors4646. Sampaio SC, Brigatte P, Sousa-e-Silva MCC, dos-Santos EC, Rangel-Santos AC, Curi R, et al. Contribution of crotoxin for the inhibitory effect of Crotalus durissus terrificus snake venom on macrophage function. Toxicon. 2003;41(7):899-907. Available from: https://doi.org/10.1016/S0041-0101(03)00069-2.
https://doi.org/10.1016/S0041-0101(03)00...
,5151. Brigatte P, Faiad OJ, Nocelli RCF, Landgraf RG, Palma MS, Cury Y, et al. Walker 256 Tumor Growth Suppression by Crotoxin Involves Formyl Peptide Receptors and Lipoxin A₄. Mediators Inflamm. 2016;2016:2457532. Available from: http://dx.doi.org/10.1155/2016/2457532
https://doi.org/10.1155/2016/2457532...
.

Macrophages cultured in vitro in the presence of crotoxin secreted 47% less angiogenesis mediators than macrophages from a control group4646. Sampaio SC, Brigatte P, Sousa-e-Silva MCC, dos-Santos EC, Rangel-Santos AC, Curi R, et al. Contribution of crotoxin for the inhibitory effect of Crotalus durissus terrificus snake venom on macrophage function. Toxicon. 2003;41(7):899-907. Available from: https://doi.org/10.1016/S0041-0101(03)00069-2.
https://doi.org/10.1016/S0041-0101(03)00...
, confirming the role of the toxin in reducing tumor blood vessel neoformation.

The efficacy of crotoxin in dose-dependent inhibition of human esophageal carcinoma tumor growth (Eca-109 cells) was demonstrated in vivo5555. He J, Wu X, Wang Y, Han R, Qin Z, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta pharmacol Sin. 2013;34(2):295-300. Available from: https://doi.org/10.1038/aps.2012.156
https://doi.org/10.1038/aps.2012.156...
,5757. Han R, Liang H, Qin ZH, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin . 2014;35(10):1323-32. Available from: https://doi.org/10.1038/aps.2014.62
https://doi.org/10.1038/aps.2014.62...
. The toxin causes cellular damage to the lineage, such as formation of pyknotic cell nuclei, cell lysis, and DNA damage5555. He J, Wu X, Wang Y, Han R, Qin Z, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta pharmacol Sin. 2013;34(2):295-300. Available from: https://doi.org/10.1038/aps.2012.156
https://doi.org/10.1038/aps.2012.156...
. Exposure of tumor cells to crotoxin also resulted in an increase in the number of stagnant cells in the G1 phase of cell division5353. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
,5555. He J, Wu X, Wang Y, Han R, Qin Z, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta pharmacol Sin. 2013;34(2):295-300. Available from: https://doi.org/10.1038/aps.2012.156
https://doi.org/10.1038/aps.2012.156...
,5757. Han R, Liang H, Qin ZH, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin . 2014;35(10):1323-32. Available from: https://doi.org/10.1038/aps.2014.62
https://doi.org/10.1038/aps.2014.62...
. Increased expression of caspase 3, p17, and p15 proteins and reduced production of Bcl-25 protein can be envolved5555. He J, Wu X, Wang Y, Han R, Qin Z, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta pharmacol Sin. 2013;34(2):295-300. Available from: https://doi.org/10.1038/aps.2012.156
https://doi.org/10.1038/aps.2012.156...
.

In vivo studies on the HL-60 leukemia cell line showed lower tumor growth inhibitory activity, suggesting that it acts preferentially on solid tumors2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,4747. Yang C, Yang Y, Qin Z, Gu Z, Reid P, Liang Z. Autophagy is involved in cytotoxic effects of crotoxin in human breast cancer cell line MCF-7 cells. Acta Pharmacol Sin. 2007;28(4):540-8. Available from: http://dx.doi.org/10.1111/j.1745-7254.2007.00530.x
https://doi.org/10.1111/j.1745-7254.2007...
,4848. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...
,5050. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...
. The treatment of patients with solid tumors refractory to conventional antineoplastic therapies with the administration of different doses of crotoxin has demonstrated efficacy in reducing different types of carcinomas2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
. Mechanisms of mitochondrial collapse, cytochrome C release, and caspase 3 activation induced cell death in the human leukemia-associated K562 cell line, with the induction of apoptosis and autophagy observed5050. Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
https://doi.org/10.3892/or.2012.1677...
,5757. Han R, Liang H, Qin ZH, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin . 2014;35(10):1323-32. Available from: https://doi.org/10.1038/aps.2014.62
https://doi.org/10.1038/aps.2014.62...
.

Crotoxin has been shown to be more cytotoxic than standard chemotherapeutic agents for the treatment of glioma, pancreatic cancer, esophageal cancer, and cervical cancer. Therefore, novel antineoplastic therapies are of great interest, particularly against leukemia, lung cancer, colon cancer, renal cancer, ovarian cancer, esophageal carcinoma, breast carcinoma, melanoma, and brain tumors, whose proliferation is already known to be preventable by the toxin1919. Soares MA, Pujatti PB, Fortes-Dias CI, Antonelli L, Santos RG. Crotalus durissus terrificus venom as a source of antitumoral agents. J Venom Anim Toxins Incl Trop Dis . 2010;16(3):480-92. Available from: https://doi.org/10.1590/S1678-91992010000300015
https://doi.org/10.1590/S1678-9199201000...
,5353. Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
https://doi.org/10.1016/j.toxicon.2018.1...
,5757. Han R, Liang H, Qin ZH, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin . 2014;35(10):1323-32. Available from: https://doi.org/10.1038/aps.2014.62
https://doi.org/10.1038/aps.2014.62...
. New drugs derived from the toxin, such as VRCTC-310 and CB24, have already been studied in murine and human cell lines1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,1717. Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B. Anticancer potential of animal venoms and toxins. Indian J Exp Biol. 2010;48(2):93-103.,2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,4141. Marcussi S, Santos PRS, Menaldo DL, Silveira LB, Santos-Filho NA, Mazzi MV, et al. Evaluation of the genotoxicity of Crotalus durissus terrificus snake venom and its isolated toxins on human lymphocytes. Mutat Res. 2011;724(1-2):59-63. Available from: https://doi.org/10.1016/j.mrgentox.2011.06.004
https://doi.org/10.1016/j.mrgentox.2011....
,4848. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...
.

PHOSPHOLIPASES A2

PLA2 are type 1 and type 2 enzymes associated with the induction of inflammatory processes, lipid membrane metabolism, and release of substances such as prostaglandins, prostacyclins, thromboxanes, and leukotrienes1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,5858. Cotrim CA, Oliveira SCB, Filho EBSD, Fonseca FV, Jr LB, Antunes E, et al. Quercetin as an inhibitor of snake venom secretory phospholipase A2. Chem Biol Interact. 2011;189(1-2):9-16. Available from: https://doi.org/10.1016/j.cbi.2010.10.016
https://doi.org/10.1016/j.cbi.2010.10.01...

59. Sunagar K, Jackson TNW, Reeks T, Fry BG. Group I phospholipase A2 enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 327-34.
-6060. Sunagar K, Tsai IH, Lomonte B, Jackson TNW, Fry BG. Group II phospholipase A2 enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 335-40..

These enzymes represent the largest family of proteins contained in the venom2323. Melani RD, Araujo GDT, Carvalho PC, Goto L, Nogueira FCS, Junqueira M, et al. Seeing beyond the tip of the icebeg: A deep analysis of the venome of the Brazilian Rattlesnake, Crotalus durissus terrificus. EuPA Open Proteom. 2015;8:144-56. Available from: https://doi.org/10.1016/j.euprot.2015.05.006
https://doi.org/10.1016/j.euprot.2015.05...
,5858. Cotrim CA, Oliveira SCB, Filho EBSD, Fonseca FV, Jr LB, Antunes E, et al. Quercetin as an inhibitor of snake venom secretory phospholipase A2. Chem Biol Interact. 2011;189(1-2):9-16. Available from: https://doi.org/10.1016/j.cbi.2010.10.016
https://doi.org/10.1016/j.cbi.2010.10.01...
, accounting for up to 80% of total proteins2424. Boldrini-França J, Cologna CT, Pucca MB, Bordon KCF, Amorim FG, Anjolette FAP, et al. Minor snake venom proteins: Structure, function and potential applications. Biochim Biophys Acta Gen Subj. 2017;1861(4):824-38. Available from: http://dx.doi.org/10.1016/j.bbagen.2016.12.022
https://doi.org/10.1016/j.bbagen.2016.12...
. PLA2 induces processes such as edema, blockage of neuromuscular junctions, platelet aggregation, and muscle necrosis2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,5959. Sunagar K, Jackson TNW, Reeks T, Fry BG. Group I phospholipase A2 enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 327-34.. It has a substantial pharmacological interest owing to a wide range of biological actions6060. Sunagar K, Tsai IH, Lomonte B, Jackson TNW, Fry BG. Group II phospholipase A2 enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 335-40.. Some enzymes have anticoagulant activity through mechanisms of hydrolysis of procoagulant phospholipids, antagonistic effects with coagulant proteins, and interaction with factor X2525. Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, et al. Secondary homeostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus. Int J Biol Macromol. 2019;131:127-33. Available from: https://doi.org/10.1016/j.ijbiomac.2019.03.059
https://doi.org/10.1016/j.ijbiomac.2019....
. Cotrim et al. (2011) suggested that PLA2 activity is attributable to its actions at different pharmacological sites, which are responsible for platelet aggregation, myotoxicity, and antibacterial activity, as well as anti-inflammatory and neurotoxic effects5858. Cotrim CA, Oliveira SCB, Filho EBSD, Fonseca FV, Jr LB, Antunes E, et al. Quercetin as an inhibitor of snake venom secretory phospholipase A2. Chem Biol Interact. 2011;189(1-2):9-16. Available from: https://doi.org/10.1016/j.cbi.2010.10.016
https://doi.org/10.1016/j.cbi.2010.10.01...
.

PLA2 has shown anticancer properties by acting on epithelial growth factor receptors (EGFR), reducing the production of tumor necrosis factor, and inhibiting neoplastic growth in lung carcinoma, human breast carcinoma, and leukemia.

The Cdt crotoxin and Naja naja atra cardiotoxin association has been conducted to develop “VRCTC-310-Onco,” which aims to interfere with the signaling of EGRFs, reduce the production of tumor necrosis factor, and exert cytotoxic action on tumor cells1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,4848. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...
. The development of EGFR receptor inhibitor drugs represents a new type of therapy against epithelial neoplasms6161. Herbst RS. Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Phys. 2004;59(2):2-26. Available from: http://dx.doi.org/10.1016/j.ijrobp.2003.11.041
https://doi.org/10.1016/j.ijrobp.2003.11...
,6262. Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007;7:169-81. Available from: https://doi.org/10.1038/nrc2088
https://doi.org/10.1038/nrc2088...
given that the receptors act in the signaling responsible for the formation of epithelial cell tumors6161. Herbst RS. Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Phys. 2004;59(2):2-26. Available from: http://dx.doi.org/10.1016/j.ijrobp.2003.11.041
https://doi.org/10.1016/j.ijrobp.2003.11...
.

Reduction in tumor necrosis factor production is also an important mechanism of anticancer action, since the presence of necrosis stimulates tumor phosphorescence mediators, favoring angiogenesis and tumor metastasis.

GYROXIN

Gyroxin, a member of the serinoprotease family, is a neurotoxic enzyme with coagulant action44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,66. Frare BT, Resende YKS, Dornelas BC, Jorge MT, Ricarte VAS, Alves LM, et al. Clinical, Laboratory, and Therapeutic Aspects of Crotalus durissus (South American Rattlesnake) Victims: A Literature Review. Biomed Res Int. 2019; Article ID 1345923:1-7. Available from: https://doi.org/10.1155/2019/1345923
https://doi.org/10.1155/2019/1345923...
,2525. Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, et al. Secondary homeostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus. Int J Biol Macromol. 2019;131:127-33. Available from: https://doi.org/10.1016/j.ijbiomac.2019.03.059
https://doi.org/10.1016/j.ijbiomac.2019....
,4545. Silva-Júnior LN, Abreu LS, Rodrigues CFB, Galizio NC, Aguiar WS, Serino-Silva C, et al. Geographic variation of individual venom profile of Crotalus durissus snakes. J Venom Anim Toxins Incl Trop Dis . 2020;26:e20200016. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2020-0016
https://doi.org/10.1590/1678-9199-jvatit...
,6363. Ferreira RS, de Barros LC, Abbade LPF, Barraviera SRCS, Silvares MRC, de Pontes LG, et al. Heterologous fibrin sealant derived from snake venom: from bench to bedside - an overview. J Venom Anim Toxins Incl Trop Dis . 2017;23:21. Available from: http://dx.doi.org/10.1186/s40409-017-0109-8
https://doi.org/http://dx.doi.org/10.118...
, including thrombin-like action44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,3737. Tasima LJ, Hatakeyama DM, Serino-Silva C, Rodrigues C, de Lima E, Sant'Anna SS, et al. Comparative proteomic profiling and functional characterization of venom pooled from captive Crotalus durissus terrificus specimens and the Brazilian crotalic reference venom. Toxicon. 2020;185:26-35. Available from: https://doi.org/10.1016/j.toxicon.2020.07.001
https://doi.org/10.1016/j.toxicon.2020.0...
,4545. Silva-Júnior LN, Abreu LS, Rodrigues CFB, Galizio NC, Aguiar WS, Serino-Silva C, et al. Geographic variation of individual venom profile of Crotalus durissus snakes. J Venom Anim Toxins Incl Trop Dis . 2020;26:e20200016. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2020-0016
https://doi.org/10.1590/1678-9199-jvatit...
,6363. Ferreira RS, de Barros LC, Abbade LPF, Barraviera SRCS, Silvares MRC, de Pontes LG, et al. Heterologous fibrin sealant derived from snake venom: from bench to bedside - an overview. J Venom Anim Toxins Incl Trop Dis . 2017;23:21. Available from: http://dx.doi.org/10.1186/s40409-017-0109-8
https://doi.org/http://dx.doi.org/10.118...
,6464. Abbade LPF, Barraviera SRCS, Silvares MRC, Lima ABBCO, Haddad GR, Gatti MAN, et al. Treatment of Chronic Venous Ulcers with Heterologous Fibrin Sealant: A Phase I/II Clinical Trial. Front Immunol. 2021;12:627541. Available from: http://dx.doi.org/10.3389/fimmu.2021.627541
https://doi.org/10.3389/fimmu.2021.62754...
, and represents the second most commonly found family of venoms3737. Tasima LJ, Hatakeyama DM, Serino-Silva C, Rodrigues C, de Lima E, Sant'Anna SS, et al. Comparative proteomic profiling and functional characterization of venom pooled from captive Crotalus durissus terrificus specimens and the Brazilian crotalic reference venom. Toxicon. 2020;185:26-35. Available from: https://doi.org/10.1016/j.toxicon.2020.07.001
https://doi.org/10.1016/j.toxicon.2020.0...
. Montoni et al. (2020) demonstrated that the toxin also has the ability to cross the blood-brain barrier3535. Montoni F, Andreotti DZ, Eichler RAS, Santos WS, Kisaki CY, Arcos SSS, et al. The impact of rattlesnake venom on mice cerebellum proteomics points to synaptic inhibition and tissue damage. J Proteomics. 2020;221:103779. Available from: https://doi.org/10.1016/j.jprot.2020.103779
https://doi.org/10.1016/j.jprot.2020.103...
.

In vitro studies have revealed that the enzyme generates clotting in human plasma samples with citrate, with the speed of clot formation being directly proportional to the amount of gyroxin2525. Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, et al. Secondary homeostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus. Int J Biol Macromol. 2019;131:127-33. Available from: https://doi.org/10.1016/j.ijbiomac.2019.03.059
https://doi.org/10.1016/j.ijbiomac.2019....
, causing the breakdown of fibrinogen into fibrinopeptide A2525. Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, et al. Secondary homeostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus. Int J Biol Macromol. 2019;131:127-33. Available from: https://doi.org/10.1016/j.ijbiomac.2019.03.059
https://doi.org/10.1016/j.ijbiomac.2019....
. Gyroxin is the enzyme responsible for the coagulant activity of Cdt venom as it rapidly consumes circulating fibrinogen, making the blood incoagulable.

Brazilian researchers have used this activity to develop a biopolymer (Heterologous Fibrin Sealant, HFS), which consists of a fibrinogen-rich cryoprecipitate extracted from buffalo blood and a thrombin-like enzyme (gyroxin) purified from Crotalus durissus terrificus snake venom2727. Santos L, Oliveira C, Vasconcelos BM, Vilela D, Melo L, Ambrósio L, et al. Good management practices of venomous snakes in captivity to produce biological venom-based medicines: achieving replicability and contributing to pharmaceutical industry. J Toxicol Environ Health B Crit Rev. 2021;24(1):30-50. Available from: http://dx.doi.org/10.1080/10937404.2020.1855279
https://doi.org/10.1080/10937404.2020.18...
,6363. Ferreira RS, de Barros LC, Abbade LPF, Barraviera SRCS, Silvares MRC, de Pontes LG, et al. Heterologous fibrin sealant derived from snake venom: from bench to bedside - an overview. J Venom Anim Toxins Incl Trop Dis . 2017;23:21. Available from: http://dx.doi.org/10.1186/s40409-017-0109-8
https://doi.org/http://dx.doi.org/10.118...

64. Abbade LPF, Barraviera SRCS, Silvares MRC, Lima ABBCO, Haddad GR, Gatti MAN, et al. Treatment of Chronic Venous Ulcers with Heterologous Fibrin Sealant: A Phase I/II Clinical Trial. Front Immunol. 2021;12:627541. Available from: http://dx.doi.org/10.3389/fimmu.2021.627541
https://doi.org/10.3389/fimmu.2021.62754...
-6565. Abbade LPF, Ferreira Jr RS, dos Santos LD, Barraviera B. Chronic venous ulcers: a review on treatment with fibrin sealant and prognostic advances using proteomic strategies. J Venom Anim Toxins Incl Trop Dis . 2020;26:e20190101. Available from: http://dx.doi.org/10.1590/1678-9199-jvatitd-2019-0101
https://doi.org/10.1590/1678-9199-jvatit...
. They successfully evaluated the safety and immunogenicity of HFS for the first time, estimated the optimum dose, and assessed its preliminary efficacy in the treatment of chronic venous ulcers (CVU) in a phase 2 clinical trial2727. Santos L, Oliveira C, Vasconcelos BM, Vilela D, Melo L, Ambrósio L, et al. Good management practices of venomous snakes in captivity to produce biological venom-based medicines: achieving replicability and contributing to pharmaceutical industry. J Toxicol Environ Health B Crit Rev. 2021;24(1):30-50. Available from: http://dx.doi.org/10.1080/10937404.2020.1855279
https://doi.org/10.1080/10937404.2020.18...
,6363. Ferreira RS, de Barros LC, Abbade LPF, Barraviera SRCS, Silvares MRC, de Pontes LG, et al. Heterologous fibrin sealant derived from snake venom: from bench to bedside - an overview. J Venom Anim Toxins Incl Trop Dis . 2017;23:21. Available from: http://dx.doi.org/10.1186/s40409-017-0109-8
https://doi.org/http://dx.doi.org/10.118...

64. Abbade LPF, Barraviera SRCS, Silvares MRC, Lima ABBCO, Haddad GR, Gatti MAN, et al. Treatment of Chronic Venous Ulcers with Heterologous Fibrin Sealant: A Phase I/II Clinical Trial. Front Immunol. 2021;12:627541. Available from: http://dx.doi.org/10.3389/fimmu.2021.627541
https://doi.org/10.3389/fimmu.2021.62754...
-6565. Abbade LPF, Ferreira Jr RS, dos Santos LD, Barraviera B. Chronic venous ulcers: a review on treatment with fibrin sealant and prognostic advances using proteomic strategies. J Venom Anim Toxins Incl Trop Dis . 2020;26:e20190101. Available from: http://dx.doi.org/10.1590/1678-9199-jvatitd-2019-0101
https://doi.org/10.1590/1678-9199-jvatit...
.

As gyroxin can cross the blood-brain barrier, it can be an important tool for studies of tumors of the brain and central nervous system.

CONVULXIN

Convulxin is a high-molecular-mass glycoprotein of the C-type lectin family, which has potent platelet activating and aggregating action44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,66. Frare BT, Resende YKS, Dornelas BC, Jorge MT, Ricarte VAS, Alves LM, et al. Clinical, Laboratory, and Therapeutic Aspects of Crotalus durissus (South American Rattlesnake) Victims: A Literature Review. Biomed Res Int. 2019; Article ID 1345923:1-7. Available from: https://doi.org/10.1155/2019/1345923
https://doi.org/10.1155/2019/1345923...
,6666. Francischetti IMB, Ghazaleh FA, Reis RAM, Carlini CR, Guimarães JA. Convulxin induces platelet activation by a tyrosine-kinase-dependent pathway and stimulates tyrosine phosphorylation of platelet proteins, including PLCγ2, independently of integrin αIIBβ3. Arch biochem biophys. 1998;353(2):239-50.,6767. Arlinghaus FT, Fry BG, Sunagar K, Jackson TNW, Eble JA, Reeks T, et al. Lectin proteins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 299-311., with high affinity for platelets6666. Francischetti IMB, Ghazaleh FA, Reis RAM, Carlini CR, Guimarães JA. Convulxin induces platelet activation by a tyrosine-kinase-dependent pathway and stimulates tyrosine phosphorylation of platelet proteins, including PLCγ2, independently of integrin αIIBβ3. Arch biochem biophys. 1998;353(2):239-50.. However, its effect on human peripheral blood mononuclear cells (PBMCs) and the immune system remains unclear.

The mechanism of action of convulxin involves the activation of phospholipase C and its rapid phosphorylation, which is similar to the mechanism induced by collagens in mediating platelet aggregation6666. Francischetti IMB, Ghazaleh FA, Reis RAM, Carlini CR, Guimarães JA. Convulxin induces platelet activation by a tyrosine-kinase-dependent pathway and stimulates tyrosine phosphorylation of platelet proteins, including PLCγ2, independently of integrin αIIBβ3. Arch biochem biophys. 1998;353(2):239-50..

In in vitro studies utilizing citrated human plasma samples, the protein generated clot formation without interfering with factors of the coagulation cascade2525. Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, et al. Secondary homeostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus. Int J Biol Macromol. 2019;131:127-33. Available from: https://doi.org/10.1016/j.ijbiomac.2019.03.059
https://doi.org/10.1016/j.ijbiomac.2019....
.

CROTAMINE

Crotamine is a non-enzymatic polypeptide with myotoxic and neurotoxic actions, responsible for causing cell death in skeletal muscles due to alterations in their sodium channels44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,77. Tasima LJ, Serino-Silva C, Hatakeyama DM, Nishiduka ES, Tashima AK, Sant’Anna SS, et al. Crotamine in Crotalus durissus: distribution according to subspecies and geographic origin, in captivity or nature. J Venom Anim Toxins Incl Trop Dis. 2020;26:e20190053. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2019-0053
https://doi.org/10.1590/1678-9199-jvatit...
,1010. Fry BG, Sunagar K, Casewell NR, Kochva E, Roelants K, Scheib H, et al. The origin and evolution of the toxicofera reptile venom system. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press; 2015. p. 1-31.,1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,3737. Tasima LJ, Hatakeyama DM, Serino-Silva C, Rodrigues C, de Lima E, Sant'Anna SS, et al. Comparative proteomic profiling and functional characterization of venom pooled from captive Crotalus durissus terrificus specimens and the Brazilian crotalic reference venom. Toxicon. 2020;185:26-35. Available from: https://doi.org/10.1016/j.toxicon.2020.07.001
https://doi.org/10.1016/j.toxicon.2020.0...
,6868. Kerkis I, Hayashi MAF, da Silva ARBP, Pereira A, Júnior PLS, Zaharenko AJ, et al. State of the art in the studies on crotamine, a cell penetrating peptide from South American rattlesnake. Biomed Res Int . 2014;2014:675985. Available from: https://doi.org/10.1155/2014/675985
https://doi.org/10.1155/2014/675985...

69. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.

70. Falcao CB, Rádis-Baptista G. Crotamine and crotalicidin, membrane active peptides from Crotalus durissus terrificus rattlesnake venom, and their structurally-minimized fragments for applications in medicine and biotechnology. Peptides. 2020;126:170234. Available from: https://doi.org/10.1016/j.peptides.2019.170234.
https://doi.org/10.1016/j.peptides.2019....
-7171. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38..

A great curiosity is that this myotoxin is not present in all individuals of the species, being thus classified as crotamine-positive or crotamine-negative venom-producing animals77. Tasima LJ, Serino-Silva C, Hatakeyama DM, Nishiduka ES, Tashima AK, Sant’Anna SS, et al. Crotamine in Crotalus durissus: distribution according to subspecies and geographic origin, in captivity or nature. J Venom Anim Toxins Incl Trop Dis. 2020;26:e20190053. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2019-0053
https://doi.org/10.1590/1678-9199-jvatit...
,1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,2323. Melani RD, Araujo GDT, Carvalho PC, Goto L, Nogueira FCS, Junqueira M, et al. Seeing beyond the tip of the icebeg: A deep analysis of the venome of the Brazilian Rattlesnake, Crotalus durissus terrificus. EuPA Open Proteom. 2015;8:144-56. Available from: https://doi.org/10.1016/j.euprot.2015.05.006
https://doi.org/10.1016/j.euprot.2015.05...
,2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,3333. Fusco LS, Neto EB, Francisco AF, Alfonso J, Soares A, Pimenta DC, et al. Fast venomic analysis of Crotalus durissus terrificus from northeastern Argentina. Toxicon X. 2020;7:100047. Available from: https://doi.org/10.1016/j.toxcx.2020.100047
https://doi.org/10.1016/j.toxcx.2020.100...
,3434. Lourenço Jr A, Creste CFZ, Barros LC, dos Santos LC, Pimenta DC, Barraviera B, et al. Individual venom profiling of Crotalus durissus terrificus specimens from a geographically limited region: Crotamine assessment and captivity evaluation on the biological activities. Toxicon. 2013;69:75-81. Available from: https://doi.org/10.1016/j.toxicon.2013.01.006
https://doi.org/10.1016/j.toxicon.2013.0...
,4545. Silva-Júnior LN, Abreu LS, Rodrigues CFB, Galizio NC, Aguiar WS, Serino-Silva C, et al. Geographic variation of individual venom profile of Crotalus durissus snakes. J Venom Anim Toxins Incl Trop Dis . 2020;26:e20200016. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2020-0016
https://doi.org/10.1590/1678-9199-jvatit...
,7171. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38.. In crotamine-positive venom-producing animals, the toxin comprises approximately 10%-15% of the venom3131. Devi A. The Protein and Nonprotein Constituents of Snake Venoms. In: Bucherl W, Buckley EE, Deulofeu V, editors. Venomous Animals and their Venoms. 1st ed. New York: Academic Press; 1968. p. 119-65. Available from: https://doi.org/10.1016/B978-1-4832-2949-2.50014-X
https://doi.org/10.1016/B978-1-4832-2949...
,3333. Fusco LS, Neto EB, Francisco AF, Alfonso J, Soares A, Pimenta DC, et al. Fast venomic analysis of Crotalus durissus terrificus from northeastern Argentina. Toxicon X. 2020;7:100047. Available from: https://doi.org/10.1016/j.toxcx.2020.100047
https://doi.org/10.1016/j.toxcx.2020.100...
,7171. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38.,7272. Campeiro JD, Marinovic MP, Carapeto FC, Mas CD, Monte GG, Porta LC, et al. Oral treatment with a rattlesnake native polypeptide crotamine efficiently inhibits the tumor growth with no potential toxicity for the host animal and with suggestive positive effects on animal metabolic profile. Amino Acids. 2018;50(2):267-78. Available from: http://dx.doi.org/10.1007/s00726-017-2513-3
https://doi.org/10.1007/s00726-017-2513-...
.

This toxin induces skeletal muscle contraction through its action on sodium channels, interfering with ion permeability in the sarcolemma and reducing the resting potential of the membranes1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,6969. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.. The changes in permeability cause a greater influx of sodium and calcium ions, which are responsible for depolarization, muscle contraction, vacuolization of sarcoplasmic reticulum, rupture of actin and myosin muscle filaments, and muscle necrosis1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,6969. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.,7171. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38..

Crotamine displays analgesic, antibacterial, antifungal, antiparasitic, and antitumor actions44. Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima. Sci Amazon. 2017;6(1):42-53.,77. Tasima LJ, Serino-Silva C, Hatakeyama DM, Nishiduka ES, Tashima AK, Sant’Anna SS, et al. Crotamine in Crotalus durissus: distribution according to subspecies and geographic origin, in captivity or nature. J Venom Anim Toxins Incl Trop Dis. 2020;26:e20190053. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2019-0053
https://doi.org/10.1590/1678-9199-jvatit...
,1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,2626. Pérez-Peinado C, Defaus S, Andreu D. Hitchhiking with Nature: Snake Venom Peptides to Fight Cancer and Superbugs. Toxins. 2020;12(4):1-23. Available from: https://doi.org/10.3390/toxins12040255
https://doi.org/10.3390/toxins12040255...
,2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,7171. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38.,7373. Vu TTT, Jeong B, Yu J, Koo B, Jo S, Robinson RC, et al. Soluble prokaryotic expression and purification of crotamine using an N-terminal maltose-binding protein tag. Toxicon. 2014;92:157-65. Available from: http://dx.doi.org/10.1016/j.toxicon.2014.10.017
https://doi.org/10.1016/j.toxicon.2014.1...

74. Dal Mas C, Pinheiro DA, Campeiro JD, Mattei B, Oliveira V, Oliveira EB, et al. Biophysical and biological properties of small linear peptides derived from crotamine, a cationic antimicrobial/antitumoral toxin with cell penetrating and cargo delivery abilities. Biochim Biophys Acta Biomembr. 2017;1859(12):2340-9. Available from: https://doi.org/10.1016/j.bbamem.2017.09.006
https://doi.org/10.1016/j.bbamem.2017.09...

75. Yamane ES, Bizerra FC, Oliveira EB, Moreira JT, Rajabi M, Nunes GL, et al. Unraveling the antifungal activity of a South American rattlesnake toxin crotamine. Biochimie. 2013;95(2):231-40. Available from: https://doi.org/10.1016/j.biochi.2012.09.019
https://doi.org/10.1016/j.biochi.2012.09...
-7676. Nascimento FD, Sancey L, Pereira A, Rome C, Oliveira V, Oliveira EB, et al. The natural cell-penetrating peptide crotamine targets tumor tissue in vivo and triggers a lethal calcium-dependent pathway in cultured cells. Mol Pharm. 2012;9(2):211-21. Available from: https://doi.org/10.1021/mp2000605
https://doi.org/10.1021/mp2000605...
. It can be classified as a cell-penetrating peptide, a protein transduction domain, a Trojan peptide, or a membrane translocation sequence1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,2626. Pérez-Peinado C, Defaus S, Andreu D. Hitchhiking with Nature: Snake Venom Peptides to Fight Cancer and Superbugs. Toxins. 2020;12(4):1-23. Available from: https://doi.org/10.3390/toxins12040255
https://doi.org/10.3390/toxins12040255...
,2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,6868. Kerkis I, Hayashi MAF, da Silva ARBP, Pereira A, Júnior PLS, Zaharenko AJ, et al. State of the art in the studies on crotamine, a cell penetrating peptide from South American rattlesnake. Biomed Res Int . 2014;2014:675985. Available from: https://doi.org/10.1155/2014/675985
https://doi.org/10.1155/2014/675985...
,7272. Campeiro JD, Marinovic MP, Carapeto FC, Mas CD, Monte GG, Porta LC, et al. Oral treatment with a rattlesnake native polypeptide crotamine efficiently inhibits the tumor growth with no potential toxicity for the host animal and with suggestive positive effects on animal metabolic profile. Amino Acids. 2018;50(2):267-78. Available from: http://dx.doi.org/10.1007/s00726-017-2513-3
https://doi.org/10.1007/s00726-017-2513-...
.

Translocation across cell membranes occurs by binding between crotamine and cell surface heparan sulfate proteoglycans, endocytosis, and accumulation of the toxin in intracellular vesicles1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,6969. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.

70. Falcao CB, Rádis-Baptista G. Crotamine and crotalicidin, membrane active peptides from Crotalus durissus terrificus rattlesnake venom, and their structurally-minimized fragments for applications in medicine and biotechnology. Peptides. 2020;126:170234. Available from: https://doi.org/10.1016/j.peptides.2019.170234.
https://doi.org/10.1016/j.peptides.2019....

71. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38.
-7272. Campeiro JD, Marinovic MP, Carapeto FC, Mas CD, Monte GG, Porta LC, et al. Oral treatment with a rattlesnake native polypeptide crotamine efficiently inhibits the tumor growth with no potential toxicity for the host animal and with suggestive positive effects on animal metabolic profile. Amino Acids. 2018;50(2):267-78. Available from: http://dx.doi.org/10.1007/s00726-017-2513-3
https://doi.org/10.1007/s00726-017-2513-...
,7575. Yamane ES, Bizerra FC, Oliveira EB, Moreira JT, Rajabi M, Nunes GL, et al. Unraveling the antifungal activity of a South American rattlesnake toxin crotamine. Biochimie. 2013;95(2):231-40. Available from: https://doi.org/10.1016/j.biochi.2012.09.019
https://doi.org/10.1016/j.biochi.2012.09...
,7676. Nascimento FD, Sancey L, Pereira A, Rome C, Oliveira V, Oliveira EB, et al. The natural cell-penetrating peptide crotamine targets tumor tissue in vivo and triggers a lethal calcium-dependent pathway in cultured cells. Mol Pharm. 2012;9(2):211-21. Available from: https://doi.org/10.1021/mp2000605
https://doi.org/10.1021/mp2000605...
. To reach the cytoplasm, crotamine induces changes in the permeability of vesicles, causing it to be released and dispersed in the cell1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,6969. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.,7272. Campeiro JD, Marinovic MP, Carapeto FC, Mas CD, Monte GG, Porta LC, et al. Oral treatment with a rattlesnake native polypeptide crotamine efficiently inhibits the tumor growth with no potential toxicity for the host animal and with suggestive positive effects on animal metabolic profile. Amino Acids. 2018;50(2):267-78. Available from: http://dx.doi.org/10.1007/s00726-017-2513-3
https://doi.org/10.1007/s00726-017-2513-...
,7676. Nascimento FD, Sancey L, Pereira A, Rome C, Oliveira V, Oliveira EB, et al. The natural cell-penetrating peptide crotamine targets tumor tissue in vivo and triggers a lethal calcium-dependent pathway in cultured cells. Mol Pharm. 2012;9(2):211-21. Available from: https://doi.org/10.1021/mp2000605
https://doi.org/10.1021/mp2000605...
. In the cytoplasm, it can bind to centrosomes in the G1 phase of cell proliferation and enables the diagnosis of cell division phases by functioning as a molecular marker1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,6868. Kerkis I, Hayashi MAF, da Silva ARBP, Pereira A, Júnior PLS, Zaharenko AJ, et al. State of the art in the studies on crotamine, a cell penetrating peptide from South American rattlesnake. Biomed Res Int . 2014;2014:675985. Available from: https://doi.org/10.1155/2014/675985
https://doi.org/10.1155/2014/675985...

69. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.

70. Falcao CB, Rádis-Baptista G. Crotamine and crotalicidin, membrane active peptides from Crotalus durissus terrificus rattlesnake venom, and their structurally-minimized fragments for applications in medicine and biotechnology. Peptides. 2020;126:170234. Available from: https://doi.org/10.1016/j.peptides.2019.170234.
https://doi.org/10.1016/j.peptides.2019....
-7171. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38..

The antitumor and antimicrobial properties of crotamine are due to its ability to bind to surfaces and acidic cellular compartments such as lysosomes2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,7474. Dal Mas C, Pinheiro DA, Campeiro JD, Mattei B, Oliveira V, Oliveira EB, et al. Biophysical and biological properties of small linear peptides derived from crotamine, a cationic antimicrobial/antitumoral toxin with cell penetrating and cargo delivery abilities. Biochim Biophys Acta Biomembr. 2017;1859(12):2340-9. Available from: https://doi.org/10.1016/j.bbamem.2017.09.006
https://doi.org/10.1016/j.bbamem.2017.09...

75. Yamane ES, Bizerra FC, Oliveira EB, Moreira JT, Rajabi M, Nunes GL, et al. Unraveling the antifungal activity of a South American rattlesnake toxin crotamine. Biochimie. 2013;95(2):231-40. Available from: https://doi.org/10.1016/j.biochi.2012.09.019
https://doi.org/10.1016/j.biochi.2012.09...
-7676. Nascimento FD, Sancey L, Pereira A, Rome C, Oliveira V, Oliveira EB, et al. The natural cell-penetrating peptide crotamine targets tumor tissue in vivo and triggers a lethal calcium-dependent pathway in cultured cells. Mol Pharm. 2012;9(2):211-21. Available from: https://doi.org/10.1021/mp2000605
https://doi.org/10.1021/mp2000605...
. In tumor cells, the prevalence of negatively charged surface molecules, such as phospholipids and mucins, allows preferential binding with the toxin compared to that in healthy cells with electrically neutral surfaces7070. Falcao CB, Rádis-Baptista G. Crotamine and crotalicidin, membrane active peptides from Crotalus durissus terrificus rattlesnake venom, and their structurally-minimized fragments for applications in medicine and biotechnology. Peptides. 2020;126:170234. Available from: https://doi.org/10.1016/j.peptides.2019.170234.
https://doi.org/10.1016/j.peptides.2019....
.

To develop new drugs, synthetic analogs of crotamine were produced, composed of peptides with smaller chains, and were used to study their functions in comparison to natural crotamine, revealing the possibility of producing crotamine derivatives with important antimicrobial and antineoplastic functions1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,7474. Dal Mas C, Pinheiro DA, Campeiro JD, Mattei B, Oliveira V, Oliveira EB, et al. Biophysical and biological properties of small linear peptides derived from crotamine, a cationic antimicrobial/antitumoral toxin with cell penetrating and cargo delivery abilities. Biochim Biophys Acta Biomembr. 2017;1859(12):2340-9. Available from: https://doi.org/10.1016/j.bbamem.2017.09.006
https://doi.org/10.1016/j.bbamem.2017.09...
.

Crotamine possesses preferential selectivity for proliferating cells and for certain phases of the cell cycle1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,6969. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.,7171. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38.,7272. Campeiro JD, Marinovic MP, Carapeto FC, Mas CD, Monte GG, Porta LC, et al. Oral treatment with a rattlesnake native polypeptide crotamine efficiently inhibits the tumor growth with no potential toxicity for the host animal and with suggestive positive effects on animal metabolic profile. Amino Acids. 2018;50(2):267-78. Available from: http://dx.doi.org/10.1007/s00726-017-2513-3
https://doi.org/10.1007/s00726-017-2513-...
,7474. Dal Mas C, Pinheiro DA, Campeiro JD, Mattei B, Oliveira V, Oliveira EB, et al. Biophysical and biological properties of small linear peptides derived from crotamine, a cationic antimicrobial/antitumoral toxin with cell penetrating and cargo delivery abilities. Biochim Biophys Acta Biomembr. 2017;1859(12):2340-9. Available from: https://doi.org/10.1016/j.bbamem.2017.09.006
https://doi.org/10.1016/j.bbamem.2017.09...

75. Yamane ES, Bizerra FC, Oliveira EB, Moreira JT, Rajabi M, Nunes GL, et al. Unraveling the antifungal activity of a South American rattlesnake toxin crotamine. Biochimie. 2013;95(2):231-40. Available from: https://doi.org/10.1016/j.biochi.2012.09.019
https://doi.org/10.1016/j.biochi.2012.09...

76. Nascimento FD, Sancey L, Pereira A, Rome C, Oliveira V, Oliveira EB, et al. The natural cell-penetrating peptide crotamine targets tumor tissue in vivo and triggers a lethal calcium-dependent pathway in cultured cells. Mol Pharm. 2012;9(2):211-21. Available from: https://doi.org/10.1021/mp2000605
https://doi.org/10.1021/mp2000605...
-7777. Cura JE, Blanzaco DP, Brisson C, Cura MA, Cabrol R, Larrateguy L, et al. Phase I and pharmacokinetics study of crotoxin (cytotoxic PLA(2), NSC-624244) in patients with advanced cancer. Clin Cancer Res. 2002;8(4):1033-41. PMID: 11948110.
https://doi.org/11948110...
. Both in vitro and in vivo studies have demonstrated specific and aggressive cytotoxicity against different tumor types6969. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61..

The role of crotamine against murine melanoma cells, human melanoma cells, and primary human pancreatic adenocarcinoma cells has been extensively studied2626. Pérez-Peinado C, Defaus S, Andreu D. Hitchhiking with Nature: Snake Venom Peptides to Fight Cancer and Superbugs. Toxins. 2020;12(4):1-23. Available from: https://doi.org/10.3390/toxins12040255
https://doi.org/10.3390/toxins12040255...
,6868. Kerkis I, Hayashi MAF, da Silva ARBP, Pereira A, Júnior PLS, Zaharenko AJ, et al. State of the art in the studies on crotamine, a cell penetrating peptide from South American rattlesnake. Biomed Res Int . 2014;2014:675985. Available from: https://doi.org/10.1155/2014/675985
https://doi.org/10.1155/2014/675985...

69. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.

70. Falcao CB, Rádis-Baptista G. Crotamine and crotalicidin, membrane active peptides from Crotalus durissus terrificus rattlesnake venom, and their structurally-minimized fragments for applications in medicine and biotechnology. Peptides. 2020;126:170234. Available from: https://doi.org/10.1016/j.peptides.2019.170234.
https://doi.org/10.1016/j.peptides.2019....
-7171. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38.. Although it is cytotoxic to normal cells when administered at high doses, it is non-toxic at low doses1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
.

Crotamine administered via the intraperitoneal route at a concentration of one microgram per animal per day for 21 days demonstrated efficacy in reducing tumors in rats with subcutaneous melanoma6868. Kerkis I, Hayashi MAF, da Silva ARBP, Pereira A, Júnior PLS, Zaharenko AJ, et al. State of the art in the studies on crotamine, a cell penetrating peptide from South American rattlesnake. Biomed Res Int . 2014;2014:675985. Available from: https://doi.org/10.1155/2014/675985
https://doi.org/10.1155/2014/675985...

69. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.

70. Falcao CB, Rádis-Baptista G. Crotamine and crotalicidin, membrane active peptides from Crotalus durissus terrificus rattlesnake venom, and their structurally-minimized fragments for applications in medicine and biotechnology. Peptides. 2020;126:170234. Available from: https://doi.org/10.1016/j.peptides.2019.170234.
https://doi.org/10.1016/j.peptides.2019....

71. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38.
-7272. Campeiro JD, Marinovic MP, Carapeto FC, Mas CD, Monte GG, Porta LC, et al. Oral treatment with a rattlesnake native polypeptide crotamine efficiently inhibits the tumor growth with no potential toxicity for the host animal and with suggestive positive effects on animal metabolic profile. Amino Acids. 2018;50(2):267-78. Available from: http://dx.doi.org/10.1007/s00726-017-2513-3
https://doi.org/10.1007/s00726-017-2513-...
.

Crotamine’s action mechanisms to induce cell apoptosis include the activation of caspases, the reduction of mitochondrial membrane potential, and consequent alteration of organelle membrane permeability, inducing the release of intracellular calcium ions and the influx of extracellular calcium2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,6868. Kerkis I, Hayashi MAF, da Silva ARBP, Pereira A, Júnior PLS, Zaharenko AJ, et al. State of the art in the studies on crotamine, a cell penetrating peptide from South American rattlesnake. Biomed Res Int . 2014;2014:675985. Available from: https://doi.org/10.1155/2014/675985
https://doi.org/10.1155/2014/675985...
,7070. Falcao CB, Rádis-Baptista G. Crotamine and crotalicidin, membrane active peptides from Crotalus durissus terrificus rattlesnake venom, and their structurally-minimized fragments for applications in medicine and biotechnology. Peptides. 2020;126:170234. Available from: https://doi.org/10.1016/j.peptides.2019.170234.
https://doi.org/10.1016/j.peptides.2019....
,7171. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38.,7373. Vu TTT, Jeong B, Yu J, Koo B, Jo S, Robinson RC, et al. Soluble prokaryotic expression and purification of crotamine using an N-terminal maltose-binding protein tag. Toxicon. 2014;92:157-65. Available from: http://dx.doi.org/10.1016/j.toxicon.2014.10.017
https://doi.org/10.1016/j.toxicon.2014.1...
,7676. Nascimento FD, Sancey L, Pereira A, Rome C, Oliveira V, Oliveira EB, et al. The natural cell-penetrating peptide crotamine targets tumor tissue in vivo and triggers a lethal calcium-dependent pathway in cultured cells. Mol Pharm. 2012;9(2):211-21. Available from: https://doi.org/10.1021/mp2000605
https://doi.org/10.1021/mp2000605...
. The activation of caspases is one of the mechanisms responsible for cell apoptosis signaling7878. Anazetti MC, Melo PS. Apoptosis Cell Death: biochemistry and molecular aspects. Metrocamp Pesquisa. 2007;1(1):37-58.. Its activation can occur by alterations in mitochondrial membrane permeability, which generates cytochrome C release that amplifies apoptosis signals6969. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61. in HL-60 cells from human leukemia and urinary bladder tumors6969. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61..

Owing to the ability of the toxin to penetrate cells, it is a potential delivery mechanism for other drugs and antitumor agents1818. Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
https://doi.org/10.1517/13543784.2010.53...
,2828. Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
https://doi.org/10.1016/j.toxicon.2018.0...
,6868. Kerkis I, Hayashi MAF, da Silva ARBP, Pereira A, Júnior PLS, Zaharenko AJ, et al. State of the art in the studies on crotamine, a cell penetrating peptide from South American rattlesnake. Biomed Res Int . 2014;2014:675985. Available from: https://doi.org/10.1155/2014/675985
https://doi.org/10.1155/2014/675985...
,6969. Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.,7171. Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38.

72. Campeiro JD, Marinovic MP, Carapeto FC, Mas CD, Monte GG, Porta LC, et al. Oral treatment with a rattlesnake native polypeptide crotamine efficiently inhibits the tumor growth with no potential toxicity for the host animal and with suggestive positive effects on animal metabolic profile. Amino Acids. 2018;50(2):267-78. Available from: http://dx.doi.org/10.1007/s00726-017-2513-3
https://doi.org/10.1007/s00726-017-2513-...

73. Vu TTT, Jeong B, Yu J, Koo B, Jo S, Robinson RC, et al. Soluble prokaryotic expression and purification of crotamine using an N-terminal maltose-binding protein tag. Toxicon. 2014;92:157-65. Available from: http://dx.doi.org/10.1016/j.toxicon.2014.10.017
https://doi.org/10.1016/j.toxicon.2014.1...
-7474. Dal Mas C, Pinheiro DA, Campeiro JD, Mattei B, Oliveira V, Oliveira EB, et al. Biophysical and biological properties of small linear peptides derived from crotamine, a cationic antimicrobial/antitumoral toxin with cell penetrating and cargo delivery abilities. Biochim Biophys Acta Biomembr. 2017;1859(12):2340-9. Available from: https://doi.org/10.1016/j.bbamem.2017.09.006
https://doi.org/10.1016/j.bbamem.2017.09...
. In addition to representing a possible antineoplastic or adjuvant therapy, crotamine can be used as a diagnostic marker for cancer7070. Falcao CB, Rádis-Baptista G. Crotamine and crotalicidin, membrane active peptides from Crotalus durissus terrificus rattlesnake venom, and their structurally-minimized fragments for applications in medicine and biotechnology. Peptides. 2020;126:170234. Available from: https://doi.org/10.1016/j.peptides.2019.170234.
https://doi.org/10.1016/j.peptides.2019....
,7373. Vu TTT, Jeong B, Yu J, Koo B, Jo S, Robinson RC, et al. Soluble prokaryotic expression and purification of crotamine using an N-terminal maltose-binding protein tag. Toxicon. 2014;92:157-65. Available from: http://dx.doi.org/10.1016/j.toxicon.2014.10.017
https://doi.org/10.1016/j.toxicon.2014.1...
,7676. Nascimento FD, Sancey L, Pereira A, Rome C, Oliveira V, Oliveira EB, et al. The natural cell-penetrating peptide crotamine targets tumor tissue in vivo and triggers a lethal calcium-dependent pathway in cultured cells. Mol Pharm. 2012;9(2):211-21. Available from: https://doi.org/10.1021/mp2000605
https://doi.org/10.1021/mp2000605...
. Crotamine can be used as a diagnostic marker in human epithelial carcinoma (HeLa), human pancreatic adenocarcinoma (BxPC-3), human breast carcinoma (BT-474), and human colorectal carcinoma (Caco2) cells.

L-AMINO OXIDASES (LAAOS)

LAAOs are flavoenzymes responsible for catalyzing amino acids, which generate alpha-keto acids, ammonia, and hydrogen peroxide1414. El-Aziz TMA, Soares AG, Stockand JD. Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins. 2019;11(10):1-25. Available from: https://doi.org/10.3390/toxins11100564
https://doi.org/10.3390/toxins11100564...
,1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,1717. Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B. Anticancer potential of animal venoms and toxins. Indian J Exp Biol. 2010;48(2):93-103.,2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,3232. Sarkar NK, Devi A. Enzymes in Snake Venoms. In: Bucherl W, Buckley EE, Deulofeu V, editors. Venomous Animals and their Venoms. 1st ed. New York: Academic Press ; 1968. p. 167-216. Available from: https://doi.org/10.1016/B978-1-4832-2949-2.50015-1
https://doi.org/10.1016/B978-1-4832-2949...
,7979. Tan NH, Fry BG, Sunagar K, Jackson TNW, Reeks T, Fung SY. L-Amino acid oxidase enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 291-8,8080. Teixeira TL, Silva VAO, Cunha DB, Polettini FL, Thomaz CD, Pianca AA, et al. Isolation, characterization and screening of the in vitro cytotoxic activity of a novel L-amino acid oxidase (LAAOcdt) from Crotalus durissus terrificus venom on human cancer cell lines. Toxicon. 2016;119:203-17. Available from: https://doi.org/10.1016/j.toxicon.2016.06.009
https://doi.org/10.1016/j.toxicon.2016.0...
. Members of this enzyme class are highly toxic and have great pharmacological importance1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,7979. Tan NH, Fry BG, Sunagar K, Jackson TNW, Reeks T, Fung SY. L-Amino acid oxidase enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 291-8, as they can cause platelet aggregation, hemorrhage, edema, cytotoxicity, and induction of apoptosis1414. El-Aziz TMA, Soares AG, Stockand JD. Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins. 2019;11(10):1-25. Available from: https://doi.org/10.3390/toxins11100564
https://doi.org/10.3390/toxins11100564...
,1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,1717. Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B. Anticancer potential of animal venoms and toxins. Indian J Exp Biol. 2010;48(2):93-103.,3737. Tasima LJ, Hatakeyama DM, Serino-Silva C, Rodrigues C, de Lima E, Sant'Anna SS, et al. Comparative proteomic profiling and functional characterization of venom pooled from captive Crotalus durissus terrificus specimens and the Brazilian crotalic reference venom. Toxicon. 2020;185:26-35. Available from: https://doi.org/10.1016/j.toxicon.2020.07.001
https://doi.org/10.1016/j.toxicon.2020.0...
,7979. Tan NH, Fry BG, Sunagar K, Jackson TNW, Reeks T, Fung SY. L-Amino acid oxidase enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 291-8,8080. Teixeira TL, Silva VAO, Cunha DB, Polettini FL, Thomaz CD, Pianca AA, et al. Isolation, characterization and screening of the in vitro cytotoxic activity of a novel L-amino acid oxidase (LAAOcdt) from Crotalus durissus terrificus venom on human cancer cell lines. Toxicon. 2016;119:203-17. Available from: https://doi.org/10.1016/j.toxicon.2016.06.009
https://doi.org/10.1016/j.toxicon.2016.0...
.

These enzymes induce apoptosis in human leukemia cells. Their toxicity is mainly attributed to the formation of hydrogen peroxide during oxidative reactions, among other mechanisms1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,3030. Shanbhag VKL. Applications of snake venoms in treatment of cancer. Asian Pac J Trop Biomed . 2015;5(4):275-6. Available from: http://dx.doi.org/10.1016/S2221-1691(15)30344-0
https://doi.org/10.1016/S2221-1691(15)30...
,7979. Tan NH, Fry BG, Sunagar K, Jackson TNW, Reeks T, Fung SY. L-Amino acid oxidase enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 291-8,8080. Teixeira TL, Silva VAO, Cunha DB, Polettini FL, Thomaz CD, Pianca AA, et al. Isolation, characterization and screening of the in vitro cytotoxic activity of a novel L-amino acid oxidase (LAAOcdt) from Crotalus durissus terrificus venom on human cancer cell lines. Toxicon. 2016;119:203-17. Available from: https://doi.org/10.1016/j.toxicon.2016.06.009
https://doi.org/10.1016/j.toxicon.2016.0...
. Although cytotoxic to tumor cells, LAAOs do not affect healthy cells2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,8080. Teixeira TL, Silva VAO, Cunha DB, Polettini FL, Thomaz CD, Pianca AA, et al. Isolation, characterization and screening of the in vitro cytotoxic activity of a novel L-amino acid oxidase (LAAOcdt) from Crotalus durissus terrificus venom on human cancer cell lines. Toxicon. 2016;119:203-17. Available from: https://doi.org/10.1016/j.toxicon.2016.06.009
https://doi.org/10.1016/j.toxicon.2016.0...
.

The species-specific cytotoxicity of LAAOcdt was evaluated using nine human cancer cell lines, including pancreatic, esophageal, cervical, and glioblastoma tumors8080. Teixeira TL, Silva VAO, Cunha DB, Polettini FL, Thomaz CD, Pianca AA, et al. Isolation, characterization and screening of the in vitro cytotoxic activity of a novel L-amino acid oxidase (LAAOcdt) from Crotalus durissus terrificus venom on human cancer cell lines. Toxicon. 2016;119:203-17. Available from: https://doi.org/10.1016/j.toxicon.2016.06.009
https://doi.org/10.1016/j.toxicon.2016.0...
.

Purified LAAOs can act on different targets of cellular mechanisms such as DNA fragmentation, chromatin condensation, and nuclear fragmentation. Another mechanism is the induction of P53 protein expression, which is synthesized from a tumor suppressor gene that is functionally deficient in more than half of the human tumors7878. Anazetti MC, Melo PS. Apoptosis Cell Death: biochemistry and molecular aspects. Metrocamp Pesquisa. 2007;1(1):37-58.,7979. Tan NH, Fry BG, Sunagar K, Jackson TNW, Reeks T, Fung SY. L-Amino acid oxidase enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 291-8,8181. Fett-Conte AC, Salles ABCF. The importance of the p53 gene in human carcinogenesis. Rev Bras Hematol Hemoter. 2002;24(2):85-9. Available from: https://doi.org/10.1590/S1516-84842002000200004
https://doi.org/10.1590/S1516-8484200200...
. Moreover, the induction of protein expression would be relevant to stimulate the monitoring of genome integrity, allowing the identification of damage and repair, resulting in the reduced proliferation of cells with genetic mutations.

LECTINS

Lectins belong to a family of proteins and glycoproteins that generate platelet aggregation1010. Fry BG, Sunagar K, Casewell NR, Kochva E, Roelants K, Scheib H, et al. The origin and evolution of the toxicofera reptile venom system. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press; 2015. p. 1-31.,1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,2020. Vonk FJ, Jackson K, Doley R, Madaras F, Mirtschin PJ, Vidal N. Snake venom: From fieldwork to the clinic: Recent insights into snake biology, together with new technology allowing high-throughput screening of venom, bring new hope for drug discovery. Bioessays. 2011;33(4):269-79. Available from: http://dx.doi.org/10.1002/bies.201000117
https://doi.org/10.1002/bies.201000117...
,6767. Arlinghaus FT, Fry BG, Sunagar K, Jackson TNW, Eble JA, Reeks T, et al. Lectin proteins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 299-311.. C-type lectins are non-enzymatic calcium-dependent proteins that affect cell adhesion, endocytosis, and neutralization of pathogens6767. Arlinghaus FT, Fry BG, Sunagar K, Jackson TNW, Eble JA, Reeks T, et al. Lectin proteins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 299-311.. These proteins may also interfere with tumor proliferation, which has been observed using lectins from venom of other species, offering potential for antineoplastic therapy1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
.

METALLOPROTEASES

Metalloproteases present hemorrhagic action and induce coagulation alterations in the prey1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,8282. Markland JR, Francis S, Swenson S. Snake venom metalloproteinases. Toxicon. 2013;62:3-18. Available from: https://doi.org/10.1016/j.toxicon.2012.09.004
https://doi.org/10.1016/j.toxicon.2012.0...
. These proteins are copious in crotalid venom8282. Markland JR, Francis S, Swenson S. Snake venom metalloproteinases. Toxicon. 2013;62:3-18. Available from: https://doi.org/10.1016/j.toxicon.2012.09.004
https://doi.org/10.1016/j.toxicon.2012.0...
but are present in low quantities in Cdt venom, thus conferring low proteolytic and hemorrhagic activity3333. Fusco LS, Neto EB, Francisco AF, Alfonso J, Soares A, Pimenta DC, et al. Fast venomic analysis of Crotalus durissus terrificus from northeastern Argentina. Toxicon X. 2020;7:100047. Available from: https://doi.org/10.1016/j.toxcx.2020.100047
https://doi.org/10.1016/j.toxcx.2020.100...
.

This class of enzymes is composed of endopeptidases that degrade extracellular matrix proteins, blood components, and endothelial cells2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
. In addition, metalloproteases play a fibrinolytic role and act as prothrombin activators, blood coagulation factor X activators, apoptosis inducers, platelet aggregation inhibitors, pro-inflammatory agents, and inactivators of serinoprotease inhibitors8282. Markland JR, Francis S, Swenson S. Snake venom metalloproteinases. Toxicon. 2013;62:3-18. Available from: https://doi.org/10.1016/j.toxicon.2012.09.004
https://doi.org/10.1016/j.toxicon.2012.0...
. Different groups of metalloproteases found in viperid and crotalid venoms are involved in tumor proliferation and angiogenesis processes1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
. However, specific studies on Cdt venom metalloproteinases have not yet been conducted.

DISINTEGRINS

Disintegrins are also important for the inhibition of tumor cells, together with metalloproteases, by acting against angiogenesis and metastasis1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
. This group of non-enzymatic proteins of low molecular mass can interact with integrins expressed by different cells1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,1717. Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B. Anticancer potential of animal venoms and toxins. Indian J Exp Biol. 2010;48(2):93-103.,2020. Vonk FJ, Jackson K, Doley R, Madaras F, Mirtschin PJ, Vidal N. Snake venom: From fieldwork to the clinic: Recent insights into snake biology, together with new technology allowing high-throughput screening of venom, bring new hope for drug discovery. Bioessays. 2011;33(4):269-79. Available from: http://dx.doi.org/10.1002/bies.201000117
https://doi.org/10.1002/bies.201000117...
,8383. Galán JA, Sánchez EE, Rodríguez-Acosta A, Soto JG, Bashir S, McLane MA, et al. Inhibition of lung tumor colonization and cell migration with the disintegrin crotatroxin 2 isolated from the venom of Crotalus atrox. Toxicon. 2008;51(7):1186-96. Available from: https://doi.org/10.1016/j.toxicon.2008.02.004
https://doi.org/10.1016/j.toxicon.2008.0...
, important cell surface receptors that are involved in interactions between cells and between cells and the extracellular matrix1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,1717. Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B. Anticancer potential of animal venoms and toxins. Indian J Exp Biol. 2010;48(2):93-103.,2020. Vonk FJ, Jackson K, Doley R, Madaras F, Mirtschin PJ, Vidal N. Snake venom: From fieldwork to the clinic: Recent insights into snake biology, together with new technology allowing high-throughput screening of venom, bring new hope for drug discovery. Bioessays. 2011;33(4):269-79. Available from: http://dx.doi.org/10.1002/bies.201000117
https://doi.org/10.1002/bies.201000117...
,2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,8383. Galán JA, Sánchez EE, Rodríguez-Acosta A, Soto JG, Bashir S, McLane MA, et al. Inhibition of lung tumor colonization and cell migration with the disintegrin crotatroxin 2 isolated from the venom of Crotalus atrox. Toxicon. 2008;51(7):1186-96. Available from: https://doi.org/10.1016/j.toxicon.2008.02.004
https://doi.org/10.1016/j.toxicon.2008.0...
.

Aggrastat® (Tirofiban, Merck & Co.) and Integrilin® (Eptifibatide, Cor Therapeutics, now part of Millennium Pharmaceuticals) were developed based on snake disintegrins such as echistatin from the saw-scaled viper Echis carinatus and barbourin from the southeastern pygmy rattlesnake Sistrusus miliarius barbouiri1414. El-Aziz TMA, Soares AG, Stockand JD. Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins. 2019;11(10):1-25. Available from: https://doi.org/10.3390/toxins11100564
https://doi.org/10.3390/toxins11100564...
,2020. Vonk FJ, Jackson K, Doley R, Madaras F, Mirtschin PJ, Vidal N. Snake venom: From fieldwork to the clinic: Recent insights into snake biology, together with new technology allowing high-throughput screening of venom, bring new hope for drug discovery. Bioessays. 2011;33(4):269-79. Available from: http://dx.doi.org/10.1002/bies.201000117
https://doi.org/10.1002/bies.201000117...
,8484. Pennington MW, Czerwinski A, Norton RS. Peptide therapeutics from venom: Current status and potential. Bioorg Med Chem. 2018;26(10):2738-58. Available from: https://doi.org/10.1016/j.bmc.2017.09.029
https://doi.org/10.1016/j.bmc.2017.09.02...
.

Integrins, one of the most important targets of antineoplastic action, are cell surface adhesion molecules that function as receptors and transmitters of cellular signals for migration, invasion and cell proliferation1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,8383. Galán JA, Sánchez EE, Rodríguez-Acosta A, Soto JG, Bashir S, McLane MA, et al. Inhibition of lung tumor colonization and cell migration with the disintegrin crotatroxin 2 isolated from the venom of Crotalus atrox. Toxicon. 2008;51(7):1186-96. Available from: https://doi.org/10.1016/j.toxicon.2008.02.004
https://doi.org/10.1016/j.toxicon.2008.0...
. Inhibition of integrins is important because it affects cell proliferation, angiogenesis, and metastasis and is a widely studied antineoplastic treatment option1616. Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
https://doi.org/10.1155/2014/203639...
,8383. Galán JA, Sánchez EE, Rodríguez-Acosta A, Soto JG, Bashir S, McLane MA, et al. Inhibition of lung tumor colonization and cell migration with the disintegrin crotatroxin 2 isolated from the venom of Crotalus atrox. Toxicon. 2008;51(7):1186-96. Available from: https://doi.org/10.1016/j.toxicon.2008.02.004
https://doi.org/10.1016/j.toxicon.2008.0...
.

Disintegrins isolated from Cdt venom inhibit the interaction between tumor cells, impairing their motility, and preventing the invasion of other tissues2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
. One of the mechanisms involved is the deposition of fibrin around the tumor, which limits its growth.

PHOSPHODIESTERASES

These enzymes are less abundant in the venom, representing only approximately 2% of its total3333. Fusco LS, Neto EB, Francisco AF, Alfonso J, Soares A, Pimenta DC, et al. Fast venomic analysis of Crotalus durissus terrificus from northeastern Argentina. Toxicon X. 2020;7:100047. Available from: https://doi.org/10.1016/j.toxcx.2020.100047
https://doi.org/10.1016/j.toxcx.2020.100...
. Despite being present in the venom in low quantities, it is responsible for inducing important clinical signs of intoxication3333. Fusco LS, Neto EB, Francisco AF, Alfonso J, Soares A, Pimenta DC, et al. Fast venomic analysis of Crotalus durissus terrificus from northeastern Argentina. Toxicon X. 2020;7:100047. Available from: https://doi.org/10.1016/j.toxcx.2020.100047
https://doi.org/10.1016/j.toxcx.2020.100...
, and its antitumor activity has not yet been evaluated.

Figure 2, Figure 3, and Figure 4 summarize the main mechanisms of antineoplastic action for each component of Crotalus durissus terrificus venom.

FIGURE 2:
Main antineoplastic actions of Crotalus durissus terrificus venom associated with Crotoxin.

FIGURE 3:
Main antineoplastic actions of Crotalus durissus terrificus venom associated with Crotamin.

FIGURE 4:
Main antineoplastic actions of Crotalus durissus terrificus venom associated with other venom compounds.

CLINICAL TRIALS

Crotoxin was administered to patients with solid tumors that were refractory to standard therapy in a phase 1 clinical trial that observed a partial response of more than 50% reduction in tumor mass and a complete response in three of the 23 evaluated patients2121. Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
https://doi.org/10.7314/APJCP.2012.13.10...
,4848. Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
https://doi.org/10.1016/j.toxicon.2010.0...
,7777. Cura JE, Blanzaco DP, Brisson C, Cura MA, Cabrol R, Larrateguy L, et al. Phase I and pharmacokinetics study of crotoxin (cytotoxic PLA(2), NSC-624244) in patients with advanced cancer. Clin Cancer Res. 2002;8(4):1033-41. PMID: 11948110.
https://doi.org/11948110...
. The authors concluded that crotoxin is a new class of anticancer agents that acts through a novel mechanism of action and thought that neurotoxicity could be the principal toxic effect and appears to be manageable. They recommended 0.18 mg/m2 a therapeutic dose for Phase II studies7777. Cura JE, Blanzaco DP, Brisson C, Cura MA, Cabrol R, Larrateguy L, et al. Phase I and pharmacokinetics study of crotoxin (cytotoxic PLA(2), NSC-624244) in patients with advanced cancer. Clin Cancer Res. 2002;8(4):1033-41. PMID: 11948110.
https://doi.org/11948110...
.

The same research team proposed an innovative design for a phase 1 trial with intra-patient dose escalation to study crotoxin8585. Medioni J, Brizard M, Elaidi R, Reid PF, Benlhassan K, Bray D. Innovative design for a phase 1 trial with intra-patient dose escalation: The Crotoxin study. Contemp Clin Trials Commun. 2017;7:186-8. Available from: http://dx.doi.org/10.1016/j.conctc.2017.07.008
https://doi.org/10.1016/j.conctc.2017.07...
. As recorded on the clinical trial platform ClinicalTrial.gov, 18 patients were recruited for this study between 2015 and 2018. The researchers stated that the results would be published shortly8686. ClinicalTrials.gov [Internet]. Gil-Delgado MA (MD): National Library of Medicine (US). 2011 Nov 28. Identifier NCT01481532, Open Label Clinical Trial of Intravenous Crotoxin; 2018 Jan 31 [cited 2022 Sep 21]. Available from: Available from: https://clinicaltrials.gov/ct2/history/NCT01481532?V_5=View#StudyPageTop
https://clinicaltrials.gov/ct2/history/N...
.

CONCLUSIONS

After elucidating the various mechanisms of action of the C. d. terrificus venom, it may be stated that this venom is a potential candidate for the development of new antineoplastic therapies that are efficient against different tumor lines and act on different cellular targets.

Considering the selective cytotoxicity of venom components for tumor cells to the detriment of healthy cells, the development of innovative therapies against cancer, based on the bioactive compounds of the rattlesnake, may present greater benefits compared to current therapeutic protocols, such as chemotherapy and radiotherapy, which are known to cause alterations in the normal cells of cancer patients.

The therapeutic use of compounds from Crotalus durissus terrificus snake venom also represents an alternative for the treatment of tumors resistant to drugs currently available on the market.

Therefore, one can conclude that the improvement of studies of the different fractions of ophidian venom is of great pharmacological interest, with potential for immense impact on the future of therapeutic medicine.

REFERENCES

  • 1
    Uetz P, Hošek J. The Reptile Database. [S.I.]: Peter Uetz; 2020 [updated 2021 may 22; cited 2020 october 27]. Available from: Available from: http://www.reptile-database.org
    » http://www.reptile-database.org
  • 2
    Fraga R, Lima AP, Prudente ALC, Magnusson WE. Guide to the snakes of the Manaus region - Central Amazonia, 1st ed. Manaus: Editora Inpa, 2013. 303 p.
  • 3
    Mader DR. Reptile Medicine and Surgery, 2nd ed. [S. I.]: Elsevier; 2005. 1264 p.
  • 4
    Santos IGC, Fortes-Dias CL, Dos-Santos MC. Pharmacological applications of Brazilian snake venoms with emphasis in Crotalus durissus terrificus and Crotalus durissus ruruima Sci Amazon. 2017;6(1):42-53.
  • 5
    Marques OAV, Medeiros CR. Our Incredible Serpents: Characterization, Biology, Accidents and Conservation. 1st ed. Cotia: Ponto A; 2018. 80 p.
  • 6
    Frare BT, Resende YKS, Dornelas BC, Jorge MT, Ricarte VAS, Alves LM, et al. Clinical, Laboratory, and Therapeutic Aspects of Crotalus durissus (South American Rattlesnake) Victims: A Literature Review. Biomed Res Int. 2019; Article ID 1345923:1-7. Available from: https://doi.org/10.1155/2019/1345923
    » https://doi.org/10.1155/2019/1345923
  • 7
    Tasima LJ, Serino-Silva C, Hatakeyama DM, Nishiduka ES, Tashima AK, Sant’Anna SS, et al. Crotamine in Crotalus durissus: distribution according to subspecies and geographic origin, in captivity or nature. J Venom Anim Toxins Incl Trop Dis. 2020;26:e20190053. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2019-0053
    » https://doi.org/10.1590/1678-9199-jvatitd-2019-0053
  • 8
    Rangel-Santos A, Dos-Santos EC, Lopes-Ferreira M, Lima C, Cardoso DF, Mota I. A comparative study of biological activities of crotoxin and CB fraction of venoms from Crotalus durissus terrificus, Crotalus durissus cascavella and Crotalus durissus collilineatus Toxicon. 2004;43(7):801-10. Available from: https://doi.org/10.1016/j.toxicon.2004.03.011.
    » https://doi.org/10.1016/j.toxicon.2004.03.011
  • 9
    Wüster W, Ferguson JE, Quijada-Mascareñas JA, Pook CE, Salomão MDG, Thorpe RS. Tracing an invasion: Landbridges, refugia, and the phylogeography of the Neotropical rattlesnake (Serpentes: Viperidae: Crotalus durissus). Mol Ecol. 2005;14(4):1095-80. Available from: https://doi.org/10.1111/j.1365-294X.2005.02471.x.
    » https://doi.org/10.1111/j.1365-294X.2005.02471.x
  • 10
    Fry BG, Sunagar K, Casewell NR, Kochva E, Roelants K, Scheib H, et al. The origin and evolution of the toxicofera reptile venom system. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press; 2015. p. 1-31.
  • 11
    WHO - World Health Organization. Report of the Tenth Meeting of the WHO Strategic and Technical Advisory Group for Neglected Tropical Diseases. Geneva: WHO; 2017 [cited 2020 Oct 27]. Available from: Available from: http://www.who.int/neglected_diseases/NTD_STAG_report_2017.pdf?ua=1
    » http://www.who.int/neglected_diseases/NTD_STAG_report_2017.pdf?ua=1
  • 12
    Grego KF, Vieira SEM, Vidueiros JP, Serapicos EO, Barbarini CC, Silveira GPM, et al. Maintenance of venomous snakes in captivity for venom production at Butantan Institute from 1908 to the present: a scoping history. J Venom Anim Toxins Incl Trop Dis . 2021;27:e20200068. Available from: https://dx.doi.org/10.1590/1678-9199-jvatitd-2020-0068
    » https://doi.org/10.1590/1678-9199-jvatitd-2020-0068
  • 13
    Chippaux JP. Snakebite envenomation turns again into a neglected tropical disease. J Venom Anim Toxins Incl Trop Dis . 2017;8;23:38. Available from: https://doi.org/10.1186/s40409-017-0127-6
    » https://doi.org/10.1186/s40409-017-0127-6
  • 14
    El-Aziz TMA, Soares AG, Stockand JD. Snake Venoms in Drug Discovery: Valuable Therapeutic Tools for Life Saving. Toxins. 2019;11(10):1-25. Available from: https://doi.org/10.3390/toxins11100564
    » https://doi.org/10.3390/toxins11100564
  • 15
    Almeida JR, Resende LM, Watanabe RK, Carregari VC, Huancahuire-Vega S, Caldeira CAS, et al. Snake Venom Peptides and Low Mass Proteins: Molecular Tools and Therapeutic Agents. Curr Med Chem. 2017;24(30):3254-82. Available from: https://doi.org/10.2174/0929867323666161028155611
    » https://doi.org/10.2174/0929867323666161028155611
  • 16
    Calderon LA, Sobrinho JC, Zaqueo KD, Moura AA, Grabner NA, Mazzi MV, et al. Antitumoral activity of snake venom proteins: New trends in cancer therapy. BioMed Res Int. 2014;2014:203639. Available from: https://doi.org/10.1155/2014/203639
    » https://doi.org/10.1155/2014/203639
  • 17
    Gomes A, Bhattacharjee P, Mishra R, Biswas AK, Dasgupta SC, Giri B. Anticancer potential of animal venoms and toxins. Indian J Exp Biol. 2010;48(2):93-103.
  • 18
    Kerkis I, Silva F, Pereira A, Kerkis A, Rádis-Baptista G. Biological versatility of crotamine--a cationic peptide from the venom of a South American rattlesnake. Expert Opin Investig Drugs. 2010;19(12):1515-25. Available from: https://doi.org/10.1517/13543784.2010.534457
    » https://doi.org/10.1517/13543784.2010.534457
  • 19
    Soares MA, Pujatti PB, Fortes-Dias CI, Antonelli L, Santos RG. Crotalus durissus terrificus venom as a source of antitumoral agents. J Venom Anim Toxins Incl Trop Dis . 2010;16(3):480-92. Available from: https://doi.org/10.1590/S1678-91992010000300015
    » https://doi.org/10.1590/S1678-91992010000300015
  • 20
    Vonk FJ, Jackson K, Doley R, Madaras F, Mirtschin PJ, Vidal N. Snake venom: From fieldwork to the clinic: Recent insights into snake biology, together with new technology allowing high-throughput screening of venom, bring new hope for drug discovery. Bioessays. 2011;33(4):269-79. Available from: http://dx.doi.org/10.1002/bies.201000117
    » https://doi.org/10.1002/bies.201000117
  • 21
    Jain D, Kumar S. Snake venom: a potent anticancer agent. Asian Pac J Cancer Prev. 2012;13(10):4855-60. Available from: http://dx.doi.org/10.7314/APJCP.2012.13.10.4855
    » https://doi.org/10.7314/APJCP.2012.13.10.4855
  • 22
    Kollipara PS, Won DH, Hwang CJ, Jung YY, Yoon HS, Park MH, et al. Enhanced Anti-Cancer Effect of Snake Venom Activated NK Cells on Lung Cancer Cells by Inactivation of NF-κB. Biomol Ther. 2014;22(2):106-13. Available from: http://dx.doi.org/10.4062/biomolther.2013.103
    » https://doi.org/10.4062/biomolther.2013.103
  • 23
    Melani RD, Araujo GDT, Carvalho PC, Goto L, Nogueira FCS, Junqueira M, et al. Seeing beyond the tip of the icebeg: A deep analysis of the venome of the Brazilian Rattlesnake, Crotalus durissus terrificus EuPA Open Proteom. 2015;8:144-56. Available from: https://doi.org/10.1016/j.euprot.2015.05.006
    » https://doi.org/10.1016/j.euprot.2015.05.006
  • 24
    Boldrini-França J, Cologna CT, Pucca MB, Bordon KCF, Amorim FG, Anjolette FAP, et al. Minor snake venom proteins: Structure, function and potential applications. Biochim Biophys Acta Gen Subj. 2017;1861(4):824-38. Available from: http://dx.doi.org/10.1016/j.bbagen.2016.12.022
    » https://doi.org/10.1016/j.bbagen.2016.12.022
  • 25
    Sousa IDL, Barbosa AR, Salvador GHM, Frihling BEF, Santa-Rita PH, Soares AM, et al. Secondary homeostasis studies of crude venom and isolated proteins from the snake Crotalus durissus terrificus. Int J Biol Macromol. 2019;131:127-33. Available from: https://doi.org/10.1016/j.ijbiomac.2019.03.059
    » https://doi.org/10.1016/j.ijbiomac.2019.03.059
  • 26
    Pérez-Peinado C, Defaus S, Andreu D. Hitchhiking with Nature: Snake Venom Peptides to Fight Cancer and Superbugs. Toxins. 2020;12(4):1-23. Available from: https://doi.org/10.3390/toxins12040255
    » https://doi.org/10.3390/toxins12040255
  • 27
    Santos L, Oliveira C, Vasconcelos BM, Vilela D, Melo L, Ambrósio L, et al. Good management practices of venomous snakes in captivity to produce biological venom-based medicines: achieving replicability and contributing to pharmaceutical industry. J Toxicol Environ Health B Crit Rev. 2021;24(1):30-50. Available from: http://dx.doi.org/10.1080/10937404.2020.1855279
    » https://doi.org/10.1080/10937404.2020.1855279
  • 28
    Cunha DB, Silvestrini AVP, Silva ACG, Estevam DMP, Pollettini FL, Navarro JO, et al. Mechanistic insights into functional characteristics of native crotamine. Toxicon. 2018;146:1-12. Available from: https://doi.org/10.1016/j.toxicon.2018.03.007
    » https://doi.org/10.1016/j.toxicon.2018.03.007
  • 29
    Vyas VK, Brahmbhatt K, Bhatt H, Parmar U. Therapeutic potential of snake venom in cancer therapy: current perspectives. Asian Pac J Trop Biomed. 2013;3(2):156-62. Available from: http://dx.doi.org/10.1016/S2221-1691(13)60042-8
    » https://doi.org/10.1016/S2221-1691(13)60042-8
  • 30
    Shanbhag VKL. Applications of snake venoms in treatment of cancer. Asian Pac J Trop Biomed . 2015;5(4):275-6. Available from: http://dx.doi.org/10.1016/S2221-1691(15)30344-0
    » https://doi.org/10.1016/S2221-1691(15)30344-0
  • 31
    Devi A. The Protein and Nonprotein Constituents of Snake Venoms. In: Bucherl W, Buckley EE, Deulofeu V, editors. Venomous Animals and their Venoms. 1st ed. New York: Academic Press; 1968. p. 119-65. Available from: https://doi.org/10.1016/B978-1-4832-2949-2.50014-X
    » https://doi.org/10.1016/B978-1-4832-2949-2.50014-X
  • 32
    Sarkar NK, Devi A. Enzymes in Snake Venoms. In: Bucherl W, Buckley EE, Deulofeu V, editors. Venomous Animals and their Venoms. 1st ed. New York: Academic Press ; 1968. p. 167-216. Available from: https://doi.org/10.1016/B978-1-4832-2949-2.50015-1
    » https://doi.org/10.1016/B978-1-4832-2949-2.50015-1
  • 33
    Fusco LS, Neto EB, Francisco AF, Alfonso J, Soares A, Pimenta DC, et al. Fast venomic analysis of Crotalus durissus terrificus from northeastern Argentina. Toxicon X. 2020;7:100047. Available from: https://doi.org/10.1016/j.toxcx.2020.100047
    » https://doi.org/10.1016/j.toxcx.2020.100047
  • 34
    Lourenço Jr A, Creste CFZ, Barros LC, dos Santos LC, Pimenta DC, Barraviera B, et al. Individual venom profiling of Crotalus durissus terrificus specimens from a geographically limited region: Crotamine assessment and captivity evaluation on the biological activities. Toxicon. 2013;69:75-81. Available from: https://doi.org/10.1016/j.toxicon.2013.01.006
    » https://doi.org/10.1016/j.toxicon.2013.01.006
  • 35
    Montoni F, Andreotti DZ, Eichler RAS, Santos WS, Kisaki CY, Arcos SSS, et al. The impact of rattlesnake venom on mice cerebellum proteomics points to synaptic inhibition and tissue damage. J Proteomics. 2020;221:103779. Available from: https://doi.org/10.1016/j.jprot.2020.103779
    » https://doi.org/10.1016/j.jprot.2020.103779
  • 36
    Lima TS, Neves CL, Zambelli VO, Lopes F, Sampaio SC, Cirillo MC. Crotoxin, a rattlesnake toxin, down-modulates functions of bone marrow neutrophils and impairs the Syk-GTPase pathway. Toxicon. 2017;136:44-55. Available from: https://doi.org/10.1016/j.toxicon.2017.07.002
    » https://doi.org/10.1016/j.toxicon.2017.07.002
  • 37
    Tasima LJ, Hatakeyama DM, Serino-Silva C, Rodrigues C, de Lima E, Sant'Anna SS, et al. Comparative proteomic profiling and functional characterization of venom pooled from captive Crotalus durissus terrificus specimens and the Brazilian crotalic reference venom. Toxicon. 2020;185:26-35. Available from: https://doi.org/10.1016/j.toxicon.2020.07.001
    » https://doi.org/10.1016/j.toxicon.2020.07.001
  • 38
    Corin RE, Viskatis LJ, Vidal JC, Etcheverry MA. Cytotoxicity of crotoxin on murine erythroleukemia cells in vitro. Invest New Drugs. 1993;11(1):11-5. Available from: https://doi.org/10.1007/BF00873905
    » https://doi.org/10.1007/BF00873905
  • 39
    Bordon KCF, Perino MG, Giglio JR, Arantes EC. Isolation, enzymatic characterization and antiedematogenic activity of the first reported rattlesnake hyaluronidase from Crotalus durissus terrificus venom. Biochimie, 2012;94(12):2740-8. Available from: https://doi.org/10.1016/j.biochi.2012.08.014.
    » https://doi.org/10.1016/j.biochi.2012.08.014
  • 40
    Faure G, Bakouh N, Lourdel S, Odolczyk N, Premchandar A, Servel N, et al. Rattlesnake Phospholipase A2 increases CFTR-chloride channel current and corrects F508CFTR dysfunction: impact in Cystic Fibrosis. J Mol Biol. 2016;428(14):2898-915. Available from: http://dx.doi.org/10.1016/j.jmb.2016.05.016
    » https://doi.org/10.1016/j.jmb.2016.05.016
  • 41
    Marcussi S, Santos PRS, Menaldo DL, Silveira LB, Santos-Filho NA, Mazzi MV, et al. Evaluation of the genotoxicity of Crotalus durissus terrificus snake venom and its isolated toxins on human lymphocytes. Mutat Res. 2011;724(1-2):59-63. Available from: https://doi.org/10.1016/j.mrgentox.2011.06.004
    » https://doi.org/10.1016/j.mrgentox.2011.06.004
  • 42
    Muller VDM, Russo RR, Cintra ACO, Sartim MA, Alves-Paiva RM, Figueiredo LTM, et al. Crotoxin and phospholipases A₂ from Crotalus durissus terrificus showed antiviral activity against dengue and yellow fever viruses. Toxicon. 2012;59(4):507-15. Available from: https://doi.org/10.1016/j.toxicon.2011.05.021
    » https://doi.org/10.1016/j.toxicon.2011.05.021
  • 43
    Sartim MA, Menaldo DL, Sampaio SV. Immunotherapeutic potential of crotoxin: anti-inflammatory and immunosuppressive properties. J Venom Anim Toxins Incl Trop Dis . 2018;24:1-13. Available from: https://doi.org/10.1186/s40409-018-0178-3.
    » https://doi.org/10.1186/s40409-018-0178-3
  • 44
    Pimenta LA, Almeida MES, Bretones ML, Cirillo MC, Curi R, Sampaio SC. Crotoxin promotes macrophage reprogramming towards an antiangiogenic phenotype. Sci Rep. 2019;9(1):1-15. Available from: https://doi.org/10.1038/s41598-019-40903-0
    » https://doi.org/10.1038/s41598-019-40903-0
  • 45
    Silva-Júnior LN, Abreu LS, Rodrigues CFB, Galizio NC, Aguiar WS, Serino-Silva C, et al. Geographic variation of individual venom profile of Crotalus durissus snakes. J Venom Anim Toxins Incl Trop Dis . 2020;26:e20200016. Available from: https://doi.org/10.1590/1678-9199-jvatitd-2020-0016
    » https://doi.org/10.1590/1678-9199-jvatitd-2020-0016
  • 46
    Sampaio SC, Brigatte P, Sousa-e-Silva MCC, dos-Santos EC, Rangel-Santos AC, Curi R, et al. Contribution of crotoxin for the inhibitory effect of Crotalus durissus terrificus snake venom on macrophage function. Toxicon. 2003;41(7):899-907. Available from: https://doi.org/10.1016/S0041-0101(03)00069-2.
    » https://doi.org/10.1016/S0041-0101(03)00069-2
  • 47
    Yang C, Yang Y, Qin Z, Gu Z, Reid P, Liang Z. Autophagy is involved in cytotoxic effects of crotoxin in human breast cancer cell line MCF-7 cells. Acta Pharmacol Sin. 2007;28(4):540-8. Available from: http://dx.doi.org/10.1111/j.1745-7254.2007.00530.x
    » https://doi.org/10.1111/j.1745-7254.2007.00530.x
  • 48
    Sampaio SC, Hyslop S, Fontes MRM, Prado-Francechi J, Zambelli VO, Magro AJ, et al. Crotoxin: Novel activities for a classic β-neurotoxin. Toxicon. 2010;55(6):1045-60. Available from: http://dx.doi.org/10.1016/j.toxicon.2010.01.011
    » https://doi.org/10.1016/j.toxicon.2010.01.011
  • 49
    Ye B, Xie Y, Qin ZH, Wu JC, Han R, He JK. Anti-tumor activity of CrTX in human lung adenocarcinoma cell line A549. Acta Pharmacol Sinica . 2011;32(11):1397-401. Available from: https://doi.org/10.1038/aps.2011.116
    » https://doi.org/10.1038/aps.2011.116
  • 50
    Wang J, Xie Y, Wu J, Han R, Reid PF, Qin Z, et al. Crotoxin enhances the antitumor activity of gefinitib (Iressa) in SK-MES-1 human lung squamous carcinoma cells. Oncol Rep. 2012;27(5):1341-7. Available from: https://doi.org/10.3892/or.2012.1677
    » https://doi.org/10.3892/or.2012.1677
  • 51
    Brigatte P, Faiad OJ, Nocelli RCF, Landgraf RG, Palma MS, Cury Y, et al. Walker 256 Tumor Growth Suppression by Crotoxin Involves Formyl Peptide Receptors and Lipoxin A₄. Mediators Inflamm. 2016;2016:2457532. Available from: http://dx.doi.org/10.1155/2016/2457532
    » https://doi.org/10.1155/2016/2457532
  • 52
    Cavalcante WLG, Matos JBN, Timoteo MA, Fontes MRM, Gallacci M, de Sa PC. Neuromuscular paralysis by the basic phospholipase A2 subunit crotoxin from Crotalus durissus terrificus snake venom needs its acid chaperone to con-currently inhibit acetylcholine release and produce muscle blockage. Toxicol Appl Pharmacol. 2017;334:8-17. Available from: https://doi.org/10.1016/j.taap.2017.08.021
    » https://doi.org/10.1016/j.taap.2017.08.021
  • 53
    Muller SP, Silva V, Silvestrini A, Macedo LH, Caetano GF, Reis RM, et al. Crotoxin from Crotalus durissus terrificus venom: In vitro cytotoxic activity of a heterodimeric phospholipase A2 on human cancer-derived cell lines. Toxicon. 2018;156:13-22. Available from: https://doi.org/10.1016/j.toxicon.2018.10.306
    » https://doi.org/10.1016/j.toxicon.2018.10.306
  • 54
    Andrade CM, Rey FM, Cintra A, Sampaio SV, Torqueti MR. Effects of crotoxin, a neurotoxin from Crotalus durissus terrificus snake venom, on human endothelial cells. Int J Biol Macromol . 2019;134:613-21. Available from: https://doi.org/10.1016/j.ijbiomac.2019.05.019
    » https://doi.org/10.1016/j.ijbiomac.2019.05.019
  • 55
    He J, Wu X, Wang Y, Han R, Qin Z, Xie Y. Growth inhibitory effects and molecular mechanisms of crotoxin treatment in esophageal Eca-109 cells and transplanted tumors in nude mice. Acta pharmacol Sin. 2013;34(2):295-300. Available from: https://doi.org/10.1038/aps.2012.156
    » https://doi.org/10.1038/aps.2012.156
  • 56
    Donato NJ, Martin CA, Perez M, Newman RA, Vidal JC, Etcheverry M. Regulation of epidermal growth factor receptor activity by crotoxin, a snake venom phospholipase A2 toxin. A novel growth inhibitory mechanism. Biochem pharmacol. 1996;51(11):1535-43. Available from: https://doi.org/10.1016/0006-2952(96)00097-4
    » https://doi.org/10.1016/0006-2952(96)00097-4
  • 57
    Han R, Liang H, Qin ZH, Liu CY. Crotoxin induces apoptosis and autophagy in human lung carcinoma cells in vitro via activation of the p38MAPK signaling pathway. Acta Pharmacol Sin . 2014;35(10):1323-32. Available from: https://doi.org/10.1038/aps.2014.62
    » https://doi.org/10.1038/aps.2014.62
  • 58
    Cotrim CA, Oliveira SCB, Filho EBSD, Fonseca FV, Jr LB, Antunes E, et al. Quercetin as an inhibitor of snake venom secretory phospholipase A2. Chem Biol Interact. 2011;189(1-2):9-16. Available from: https://doi.org/10.1016/j.cbi.2010.10.016
    » https://doi.org/10.1016/j.cbi.2010.10.016
  • 59
    Sunagar K, Jackson TNW, Reeks T, Fry BG. Group I phospholipase A2 enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 327-34.
  • 60
    Sunagar K, Tsai IH, Lomonte B, Jackson TNW, Fry BG. Group II phospholipase A2 enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 335-40.
  • 61
    Herbst RS. Review of epidermal growth factor receptor biology. Int J Radiat Oncol Biol Phys. 2004;59(2):2-26. Available from: http://dx.doi.org/10.1016/j.ijrobp.2003.11.041
    » https://doi.org/10.1016/j.ijrobp.2003.11.041
  • 62
    Sharma SV, Bell DW, Settleman J, Haber DA. Epidermal growth factor receptor mutations in lung cancer. Nat Rev Cancer. 2007;7:169-81. Available from: https://doi.org/10.1038/nrc2088
    » https://doi.org/10.1038/nrc2088
  • 63
    Ferreira RS, de Barros LC, Abbade LPF, Barraviera SRCS, Silvares MRC, de Pontes LG, et al. Heterologous fibrin sealant derived from snake venom: from bench to bedside - an overview. J Venom Anim Toxins Incl Trop Dis . 2017;23:21. Available from: http://dx.doi.org/10.1186/s40409-017-0109-8
    » https://doi.org/http://dx.doi.org/10.1186/s40409-017-0109-8
  • 64
    Abbade LPF, Barraviera SRCS, Silvares MRC, Lima ABBCO, Haddad GR, Gatti MAN, et al. Treatment of Chronic Venous Ulcers with Heterologous Fibrin Sealant: A Phase I/II Clinical Trial. Front Immunol. 2021;12:627541. Available from: http://dx.doi.org/10.3389/fimmu.2021.627541
    » https://doi.org/10.3389/fimmu.2021.627541
  • 65
    Abbade LPF, Ferreira Jr RS, dos Santos LD, Barraviera B. Chronic venous ulcers: a review on treatment with fibrin sealant and prognostic advances using proteomic strategies. J Venom Anim Toxins Incl Trop Dis . 2020;26:e20190101. Available from: http://dx.doi.org/10.1590/1678-9199-jvatitd-2019-0101
    » https://doi.org/10.1590/1678-9199-jvatitd-2019-0101
  • 66
    Francischetti IMB, Ghazaleh FA, Reis RAM, Carlini CR, Guimarães JA. Convulxin induces platelet activation by a tyrosine-kinase-dependent pathway and stimulates tyrosine phosphorylation of platelet proteins, including PLCγ2, independently of integrin αIIBβ3 Arch biochem biophys. 1998;353(2):239-50.
  • 67
    Arlinghaus FT, Fry BG, Sunagar K, Jackson TNW, Eble JA, Reeks T, et al. Lectin proteins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 299-311.
  • 68
    Kerkis I, Hayashi MAF, da Silva ARBP, Pereira A, Júnior PLS, Zaharenko AJ, et al. State of the art in the studies on crotamine, a cell penetrating peptide from South American rattlesnake. Biomed Res Int . 2014;2014:675985. Available from: https://doi.org/10.1155/2014/675985
    » https://doi.org/10.1155/2014/675985
  • 69
    Rádis-Baptista G, Kerkis I. Crotamine, a small basic polypeptide myotoxin from rattlesnake venom with cell-penetrating properties. Curr Pharm Des. 2011; 17(38):4351-61.
  • 70
    Falcao CB, Rádis-Baptista G. Crotamine and crotalicidin, membrane active peptides from Crotalus durissus terrificus rattlesnake venom, and their structurally-minimized fragments for applications in medicine and biotechnology. Peptides. 2020;126:170234. Available from: https://doi.org/10.1016/j.peptides.2019.170234.
    » https://doi.org/10.1016/j.peptides.2019.170234
  • 71
    Silva ARBP, Fry BG, Sunagar K, Scheib H, Jackson TNW, Rádis-Baptista G, Zaharenko AJ, Jr PLS, Pereira A, Oguiura N, Hayashi MAF, Kerkis A, Yamane T, Kerkis I. Beta-defensins. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 228-38.
  • 72
    Campeiro JD, Marinovic MP, Carapeto FC, Mas CD, Monte GG, Porta LC, et al. Oral treatment with a rattlesnake native polypeptide crotamine efficiently inhibits the tumor growth with no potential toxicity for the host animal and with suggestive positive effects on animal metabolic profile. Amino Acids. 2018;50(2):267-78. Available from: http://dx.doi.org/10.1007/s00726-017-2513-3
    » https://doi.org/10.1007/s00726-017-2513-3
  • 73
    Vu TTT, Jeong B, Yu J, Koo B, Jo S, Robinson RC, et al. Soluble prokaryotic expression and purification of crotamine using an N-terminal maltose-binding protein tag. Toxicon. 2014;92:157-65. Available from: http://dx.doi.org/10.1016/j.toxicon.2014.10.017
    » https://doi.org/10.1016/j.toxicon.2014.10.017
  • 74
    Dal Mas C, Pinheiro DA, Campeiro JD, Mattei B, Oliveira V, Oliveira EB, et al. Biophysical and biological properties of small linear peptides derived from crotamine, a cationic antimicrobial/antitumoral toxin with cell penetrating and cargo delivery abilities. Biochim Biophys Acta Biomembr. 2017;1859(12):2340-9. Available from: https://doi.org/10.1016/j.bbamem.2017.09.006
    » https://doi.org/10.1016/j.bbamem.2017.09.006
  • 75
    Yamane ES, Bizerra FC, Oliveira EB, Moreira JT, Rajabi M, Nunes GL, et al. Unraveling the antifungal activity of a South American rattlesnake toxin crotamine. Biochimie. 2013;95(2):231-40. Available from: https://doi.org/10.1016/j.biochi.2012.09.019
    » https://doi.org/10.1016/j.biochi.2012.09.019
  • 76
    Nascimento FD, Sancey L, Pereira A, Rome C, Oliveira V, Oliveira EB, et al. The natural cell-penetrating peptide crotamine targets tumor tissue in vivo and triggers a lethal calcium-dependent pathway in cultured cells. Mol Pharm. 2012;9(2):211-21. Available from: https://doi.org/10.1021/mp2000605
    » https://doi.org/10.1021/mp2000605
  • 77
    Cura JE, Blanzaco DP, Brisson C, Cura MA, Cabrol R, Larrateguy L, et al. Phase I and pharmacokinetics study of crotoxin (cytotoxic PLA(2), NSC-624244) in patients with advanced cancer. Clin Cancer Res. 2002;8(4):1033-41. PMID: 11948110.
    » https://doi.org/11948110
  • 78
    Anazetti MC, Melo PS. Apoptosis Cell Death: biochemistry and molecular aspects. Metrocamp Pesquisa. 2007;1(1):37-58.
  • 79
    Tan NH, Fry BG, Sunagar K, Jackson TNW, Reeks T, Fung SY. L-Amino acid oxidase enzymes. In: Fry BG. Venomous reptiles & their toxins: Evolution, pathophysiology & biodiscovery. New York: Oxford University Press ; 2015. p. 291-8
  • 80
    Teixeira TL, Silva VAO, Cunha DB, Polettini FL, Thomaz CD, Pianca AA, et al. Isolation, characterization and screening of the in vitro cytotoxic activity of a novel L-amino acid oxidase (LAAOcdt) from Crotalus durissus terrificus venom on human cancer cell lines. Toxicon. 2016;119:203-17. Available from: https://doi.org/10.1016/j.toxicon.2016.06.009
    » https://doi.org/10.1016/j.toxicon.2016.06.009
  • 81
    Fett-Conte AC, Salles ABCF. The importance of the p53 gene in human carcinogenesis. Rev Bras Hematol Hemoter. 2002;24(2):85-9. Available from: https://doi.org/10.1590/S1516-84842002000200004
    » https://doi.org/10.1590/S1516-84842002000200004
  • 82
    Markland JR, Francis S, Swenson S. Snake venom metalloproteinases. Toxicon. 2013;62:3-18. Available from: https://doi.org/10.1016/j.toxicon.2012.09.004
    » https://doi.org/10.1016/j.toxicon.2012.09.004
  • 83
    Galán JA, Sánchez EE, Rodríguez-Acosta A, Soto JG, Bashir S, McLane MA, et al. Inhibition of lung tumor colonization and cell migration with the disintegrin crotatroxin 2 isolated from the venom of Crotalus atrox Toxicon. 2008;51(7):1186-96. Available from: https://doi.org/10.1016/j.toxicon.2008.02.004
    » https://doi.org/10.1016/j.toxicon.2008.02.004
  • 84
    Pennington MW, Czerwinski A, Norton RS. Peptide therapeutics from venom: Current status and potential. Bioorg Med Chem. 2018;26(10):2738-58. Available from: https://doi.org/10.1016/j.bmc.2017.09.029
    » https://doi.org/10.1016/j.bmc.2017.09.029
  • 85
    Medioni J, Brizard M, Elaidi R, Reid PF, Benlhassan K, Bray D. Innovative design for a phase 1 trial with intra-patient dose escalation: The Crotoxin study. Contemp Clin Trials Commun. 2017;7:186-8. Available from: http://dx.doi.org/10.1016/j.conctc.2017.07.008
    » https://doi.org/10.1016/j.conctc.2017.07.008
  • 86
    ClinicalTrials.gov [Internet]. Gil-Delgado MA (MD): National Library of Medicine (US). 2011 Nov 28. Identifier NCT01481532, Open Label Clinical Trial of Intravenous Crotoxin; 2018 Jan 31 [cited 2022 Sep 21]. Available from: Available from: https://clinicaltrials.gov/ct2/history/NCT01481532?V_5=View#StudyPageTop
    » https://clinicaltrials.gov/ct2/history/NCT01481532?V_5=View#StudyPageTop

Publication Dates

  • Publication in this collection
    16 Dec 2022
  • Date of issue
    2022

History

  • Received
    18 Aug 2022
  • Accepted
    04 Nov 2022
Sociedade Brasileira de Medicina Tropical - SBMT Caixa Postal 118, 38001-970 Uberaba MG Brazil, Tel.: +55 34 3318-5255 / +55 34 3318-5636/ +55 34 3318-5287, http://rsbmt.org.br/ - Uberaba - MG - Brazil
E-mail: rsbmt@uftm.edu.br