SciELO - Scientific Electronic Library Online

 
vol.51 issue2The magic numberEvaluation of reduced amino acids diets added with protected protease on productive performance in 25-100 kg barrows author indexsubject indexarticles search
Home Pagealphabetic serial listing  

Services on Demand

Journal

Article

Indicators

Related links

  • On index processCited by Google
  • Have no similar articlesSimilars in SciELO
  • On index processSimilars in Google

Share


Austral journal of veterinary sciences

Print version ISSN 0719-8000On-line version ISSN 0719-8132

Austral j. vet. sci. vol.51 no.2 Valdivia May 2019

http://dx.doi.org/10.4067/S0719-81322019000200045 

REVIEW ARTICLE

Electrochemotherapy in the treatment of neoplasms in dogs and cats

Marcelo M.M. Rangel1 

Jean C.S. Luz1 

Krishna D. Oliveira1 

Javier Ojeda2 

Jennifer O. Freytag1 

Daniela O. Suzuki3 

1 Veterinary Oncology, Vet Cancer, São Paulo, Brazil.

2 Veterinary Clinical Science Institute, Universidad Austral de Chile, Chile.

3 Institute of Biomedical Engineering, Federal University of Santa Ca tarina, Florianópolis, Brazil.

Abstract:

Electrochemotherapy (ECT) is a technique that combines chemotherapy with local application of specific electric pulses with the aim of increasing the permeability of the plasma membrane in a reversible way, improving the influx of chemotherapeutic drugs into the cytoplasm and potentiating their cytotoxic effects. This technique has broadened the range of possible treatments for oncological patients, either on its own or as adjuvant to surgical procedures. It is especially useful in tumors located in regions with only a small surgical safety margin, such as the limb extremities, skull, oral cavity, neck and perianal region, among others. ECT makes it feasible to perform procedures more conservatively, or even to perform otherwise infeasible procedures, by expanding the margins without removing healthy tissues. The objective of this paper is to provide a brief bibliographic review of the principles, applications and future possibilities of electrochemotherapy, helping to disseminate pertinent information about this relatively new technique for the treatment of cancer.

Keywords: bleomycin; electroporation; veterinary oncology; electrochemotherapy

Mechanism of electroporation and electrochemotherapy

Electroporation, or electropermeabilisation , consists of the exposure of cells to specific electrical pulses (1000 V/cm, 100 microseconds, 8 pulses at a 5 kHz frequency), which transiently increase the permeability of the plasma membrane, allowing access to the cytoplasm for molecules to which the membrane is normally impermeable or only slightly permeable (Mir et al 1996, Teissie et al 2005, Mir 2006). Under physiological conditions, the plasma membrane is subject to the so-called membrane resting potential, which remains balanced by pumps and ion channels, keeping the electrical potential of the membrane stable. Exposure of the cells to an external electric field produces an induced transmembrane potential difference, altering its resting potential and causing rearrangement of the molecules in the lipid bilayer, resulting in the formation of pores (Almers et al 2010, Kotnik et al 2010). Electroporation depends on the specific electrical parameters; the amplitude and duration of the electric field to which the cells are exposed are key points of the process (Figure 1). At low amplitude and duration, there is no detectable effect on the plasma membrane in terms of its permeability, while at moderate amplitude and duration, electropermeabilisation occurs. In the pore model, the pores close after the end of exposure, causing the cells to remain viable. Excessive amplitudes and/or durations cause nonthermal irreversible electroper- meabilisation , possibly due to nonclosure or late closure of the pores, resulting in a form of cell death called mitotic catastrophe. Mitotic catastrophe is cell death preceded by multinucleation that occurs during the metaphase (Kroemer et al 2009). Irreversible thermal electroporation can also occur when even higher amplitudes are applied, causing thermal damages to the cells (Cemazar et al 2015). An ultrastruc tural study showed some changes in the membranes, such as defects in the dynamic assembly of lipids and proteins after exposure to the electric field used in electroporation, i.e. proteins formed membrane agglomerates and hydro philic pores. These defects in dynamic assembly would be responsible for the entry of molecules during electroporation (Spugnini et al 2007c).

Figure 1 Electroporation and thermal effects caused by exposure of cells to electric fields. Graphics representing reversible and irreversible electroporation and thermal damage caused to cells due to the electric field strength ([V/cm]) and exposure duration ([s]) in seconds. Source: Cezamar, et al., 2015 (adapted). 

Electrochemotherapy

Electrochemotherapy (ECT) is a technique that com bines chemotherapy with the application of such specific electrical pulses, allowing the entry of nonpermeating antineoplastic drugs (or those with low permeant po tential) into cells (Mir 2006). The waveforms used in electrochemotherapy are square-wave mono- or biphasic pulses. The most common electric parameters used in electrochemotherapy are 8 pulses, 100 ps length, 1-5 kHz frequency and 1000 V/cm (for needle parallel electrodes) or 1300 V/cm (plate electrodes) of electric field intensi ty1 (Spugnini et al 2007a, Pavlin et al 2009, Tozon et al 2016, Maglietti et al 2016, Pierini et al 2016, Suzuki et al 2018, Lowe et al 2017). For this technique, bleomycin and cisplatin are the main drugs used (Sersa et al 2008, Escoffre and Rols 2012). Bleomycin is widely used in electrochemotherapy due to its high intrinsic cytotoxic potential and cell selectivity caused by the mechanism of mitotic cell death. Once inside the cell, bleomycin acts as an endonuclease, with the potential to promote single- and double-stranded ruptures in DNA, predominantly affecting cells in the process of proliferation (Orlowski et al 1988, MIR et al 1988, Mir 2006).

Although bleomycin can enter permeabilised cells using plasma membrane proteins, its capacity is highly restricted by the plasma membrane, which explains its limited action in these cells. This is because only some molecules are effectively internalised and reach the DNA (Mir et al 1996). Electropermeabilisation is a way of overcoming such barriers; in vitro studies report that electropermeabilisation potentiates the cytotoxic effect of the drugs by up to thousands of times (Poddevin 1991, Mir et al 1996, Gehl 2003, Orlowski et al 2016). ECT increases the absorption of antineoplastic drugs, which will remain internalised in the cell after membrane permeability is restored, maximizing their cytotoxicity. Another property of ECT is that its action is restricted to tissues exposed to the electrical pulses, reducing the risk of chemotherapeutic side effects (Spugnini and Porrello 2003, Gehl 2003, Miklavcic et al 2014, Tafuto et al 2015). Bleomycin is a drug that induces two mechanisms of cell death, depending on the number of molecules internalised after electropermeabilisation. At low concentrations of bleomycin, the cell dies after three doubling times (called mitotic death); at high concentrations, cell death occurs in a manner analogous to apoptosis (called mimetic apoptosis) (Tounekti et al 1993).

In order to reach the electroporation threshold, several aspects must be considered, such as physical factors (cell size and shape), biological features (cytoskeleton structure) and adequate electrical parameters. Hypothetically, however, it is possible to perform electroporation independently of the type of cell or tissue (Rols and Teissie 1992, Teissie et al 2005, Cemazar et al 2015, Sersa et al 2015).

Two factors must be taken into consideration for the efficiency of ECT. First, the chemotherapeutic drug can be applied either intratumorally, in which case immediate electropermeabilisation must be performed, or intra venously, with an interval of approximately 8 minutes until electropermeabilisation. The waiting time for the intravenous route allows the antineoplastic drug to reach its pharmacological peak in the tumor, remaining for ap proximately 20 to 25 minutes at an effective concentration according to the principles of the technique. The second factor is that the entire tumor, including any infiltration into adjacent tissue, must be exposed to a sufficient electric field to promote reversible electropermeabilisation, this being obtained by correct choice and placement of elec trodes and application of electrical pulses with suitable parameters (Marty et al 2006, Domenge et al 1996). In the case of tumors with larger proportions due to abnormal vascularization, hindering proper distribution of the drug, both intravenous and intratumoral administration can be considered (Maglietti et al 2016).

Due to the stochastic nature of the process, access to the tumor through electrode placement and efficient distribution of the electric field could be more important factors than the chemosensitivity of the neoplasia to the drug (Gehl 2003, Mir 2006, Cemazar et al 2015).

In addition to increased membrane permeability, other mechanisms are described in relation to ECT, such as the effect of vascular lock and involvement of the immune response. According to Sersa et al (2008), electrochem otherapy promotes a process of vascular dysregulation throughout two distinct mechanisms. One of them is that the electrical pulses cause transient cytoskeleton damage and swelling of the vascular endothelium, which leads to the death of some tumor and vascular cells. Another is the endothelial cytotoxicity promoted by ECT. Both mecha nisms reduce local blood flow, decreasing the oxygenation of the tumor, leading to hypoxia and subsequent necrosis of tumor cells (Spugnini et al 2006). Additionally, some studies have used intravital microscopy to confirm vaso constriction induced by ECT in normal and tumoral blood vessels. Vascular lock (or vascular sequestration), induced transiently by electroporation alone and for a prolonged time by electrochemotherapy, keeps the antineoplastic accumulated and prevents it from rapidly exiting the tumor due to reduced blood flow (Jarm et al 2010).

Involvement of the immune system after electrochem otherapy is important for eliminating tumoral cells. Due to the heterogenicity of tumor cells (affecting the orientation, size and distribution of chemotherapy in the tumor), not all cells are eliminated by electrochemotherapy because some have not been effectively electroporated or inter nalized sufficient amounts of the chemotherapeutic drug (Miklavcic et al 2014, Sersa et al 1997). As with other physical methods, such as radiotherapy, when the fraction of remaining cells is low enough, they can be eliminated by the immune system. A similar situation was observed in immunodeficient organisms, which had a significantly lower cure rate than immunocompetent individuals after electrochemotherapy treatment (Sersa et al 1997). This might occur due to exposure of immunogenic molecules, such as calreticulin, and the release of neoplastic antigens from the cells destroyed by the ECT, which can activate immune cells against the tumor (Calvet et al 2014, Gerlini et al 2013, Gerlini et al 2012, Miklavcic et al 2012).

Clinical results of electrochemotherapy use

Many studies have reported favorable results in different cancer types traditionally known for limited or no treatment option or for poor response to standard therapy (Table 1). Additionally, ECT could be an option for unresectable tumors or those in areas with anatomical limitations that make them amenable to this treatment. For ECT evaluation, many oncologists use the criteria for solid tumors proposed by Veterinary comparative oncology group (VCOG) (Nguyen et al 2013): Compete response (CR) is defined as disappearance of all target lesions and pathologic LNs measuring < 10 mm along the short axis. Partial response (PR) consists of at least 30% reduction in the sum of the diameters of the target lesions. Progressive disease (PD) is defined as either the appearance of one or more new lesions or at least a 20% increase in the sum of the diameters of the lesions, taking as a reference the minimal sum on examination. Additionally, the sum must show an increase of 5 mm. For stable disease (SD), the evaluation must include tumors with less than 30% reduction (PR) or a 20% increase (PD) in the sum of the diameters of the target tumors, compared to the smallest sum of diameters during monitoring. Additionally, many studies include an overall response (CR+PR) to evaluate the study group.

Table 1 Comparative results of different tumors using electrochemotherapy. 

In canines, favorable results are observed in skin tumors, especially mast cell and perianal tumors (Figure 2). In the case of mast cell tumors, using the histological grade established by Patnaik et al (1984), two studies adminis tering intratumoral cisplatin achieved complete remission in 62.6% of grade I and III tumors (25 cases) and in 78% (37 cases), including 7 grade I, 24 grade II and 6 grade III cases, with an average of 1,218 days until recurrence (Kodre et al 2009, Spugnini et al 2011). Another study using endovenous bleomycin in mast cell tumors reported complete objective remission when analysing 80 dogs; histological grade details were not included (Tozon et al 2016). However, ECT showed the greatest effectiveness in tumors < 2 cm. Lowe et al (2017) used a similar treatment, evaluating four groups and concluding that 51 dogs had 100% objective response and a recurrence time of 1,500 days. For perianal tumors, the objective response rate using intratumoral bleomycin or cisplatin was 92% when evaluated at 34 months (Tozon et al 2005) and 94% when evaluated at 14 months, with complete remission in 84.2% (Tozon et al 2010).

Figure 2 Anal hepatoid gland carcinoma in a dog treated with one session of electrochemotherapy (endovenous bleomycin 20 mg/m2). Before ECT (A), 2 months after ECT (B). 

In cats, ECT has emerged as an effective treatment for squamous cell carcinomas (SCC), especially when the tumor invasion suggests aggressive treatments (Figure 3). Several studies have reported that ECT can reach 100% objective response (73% complete remission + 27% partial response), with a median survival time of 452 days, for all stages of SCC considered together (Pierini et al 2016). Previous studies in SCC found an objective response rate of 87.5% (17 masses), with a recurrence rate of 22% observed in 2 to 8 years (Tozon et al 2014). Spugnini et al (2015) determined an 81% objective response rate in 21 cats with SCC. This study used a control group and compared the results with the ECT group. The median recurrence time was 30.5 month for the ECT group and 3.9 months for the control group.

Figure 3 Nasal squamous cell carcinoma in a cat treated with one session of electrochemotherapy (endovenous bleomycin 20 mg/m2). Before ECT (A), 2 months after ECT (B). 

In soft tissue sarcoma (STS) treatment using intra tumoral bleomycin and cisplatin, a sample of 22 dogs had an objective response rate of 95% (Spugnini et al 2007a). On the other hand, two studies of feline STS in a total of 103 cats had overall responses rates of 70% and 54%, with recurrence times of 666 days and 570 days, respectively (Spugnini et al 2011g, Spugnini et al 2007b). In both studies, tumors with smaller size had better responses. The evaluation of ECT in tumors with poor response to surgery or chemotherapy is very positive. Twelve dogs with melanoma treated with ECT using bleomycin IV reached an OR of 83.6% with a complete remission rate of 41.4%2. Maglietti et al (2017) used a single needle electrode for eleven dogs with intranasal tumors, achieving complete response in 27% and partial response in 67%. In this case, the survival time of the ECT group was significantly higher than that of the control group, with mean survival times of 16.86 months and 5.3 months, respectively. On the other hand, dogs with tumors that were chemotherapy resistant to standard protocols had been treated with ECT. In this case, three dogs with transmissible venereal tumors with a resistance to vincristine were treated with intratumoral bleomycin, achieving complete remission (Spugnini et al 2007). Similar results were reached in two dogs and four cats with cutaneous lymphoma (Spugnini et al 2008). Finally, Lowe (2016) treated different spontaneous tumors and achieved objective response in 66% of 176 patients.

Several points can be listed that support the applicability of the ECT, making it an exceptional tool in the treatment of skin and subcutaneous solid tumors. For example:

High remission rate: complete remissions were obtained in 58.4% of tumors, independent of histological origin, and partial responses in 24.7% of nodules treated after a single session, according to the meta-analysis provided by Mali et al (2013) involving 1894 tumors;

It not only treats the tumor but also acts on the adjacent margins, thus eliminating possible infiltrated tumor cells (Miklavcic et al 2012);

It presents an increase in the response rate when reap plied at weekly intervals, on tumors that did not have complete remission with single application (Campana et al 2009);

It stimulates the immune response against possible remaining cells after ECT (Miklavcic et al 2012);

It is effective in areas previously treated by means of surgery or radiotherapy, and even to tumors that are refractory to chemotherapy (Miklavcic et al 2012);

It does not cause immediate or delayed toxic effects to patients (Miklavcic et al 2012);

It has a good cost-benefit relation regarding the tech nology and chemotherapy used, not requiring large investments (Marty et al 2006).

The technique has been developed to enable the use of ECT in tumors and internal cavities (Miklavcic et al 2012), and to imaging exams such as positron emission tomography, ultrasonography and magnetic resonance imaging, making it possible to locate such tumors and even position the electrodes in internal organs (Miklavcic et al 2010, Pavliha et al 2013). The exact localization of the tumor and the use of special needle electrodes allow the application of ECT in nonsuperficial tumors such as metastases of melanomas, sarcomas, bone tumors and even brain tumors (Figure 2), or its use in the surgical treatment of internal organs or percutaneous treatment of the limbs. In these cases, the retraction of the electrodes could cause hemorrhage; however, the effect of vascular blocking would minimise this situation (Miklavcic et al 2010, Jarm et al 2010, Cemazar et al 2015). Techniques to avoid interferences on cardiac electrical activity are also being developed. The planning of the electric field is critical to the effectiveness of ECT and must be correlated with the site of application due to the heterogeneity of tissue conductivities, which may result in an irregular distribution of the electric field, and care must be taken not to affect adjacent organs (Domenge et al 1996, Deodhar et al 2011, Mali et al 2008, Suzuki et al 2018). Electrochemotherapy is under development. Currently, electroporation devices are being improved, and special types of electrodes for use in specific areas are being created as well as syn chronisation techniques timing electrical pulses to the electrocardiogram for regions close to the heart. The use of guided laparoscopy is also being developed in addition to techniques for real-time monitoring of electroporation and distribution of the electric field, which allow improved control of the electrode positioning and an effective treat ment of tumors even in areas of difficult access (Kranjc et al 2011, Mahmood et al 2011, Miklavcic et al 2012, Cemazar et al 2015).

Although electrochemotherapy has become a well-es tablished treatment for malignancies of skin and nonskin origin, standard evaluations and higher-quality clinical data are needed. Therefore, a standardisation of terminology and reporting criteria is necessary to facilitate effective communication among researchers and appropriate comparison between different treatment technologies (Campana et al 2016).

Conclusion

ECT is a technique that increases the range of possible treatments for cancer patients, making it possible to perform more conservative procedures and even surgical procedures that had previously been technically unfeasible. The use of electrochemotherapy along with other therapies has shown encouraging results in dogs and cats with different types of cancer. Further research on this technique prom ises to contribute major advancements in the treatment of cancer.

References:

Aimers W, Edwards R, Hille B, Jan L, Nicoll R, et al. 2010. Transporte de Membrana de Pequenas Moléculas e as Propriedades Elétricas das Membranas. In: Alberts B (ed). Biologia Molecular da Célula. 5a ed. Artmed: Porto Alegre, Brazil, Pp 669-672. [ Links ]

Calvet CY, Famin D, André FM, Mir LM. 2014. Electrochemotherapy with bleomycin induces hallmarks of immunogenic cell death in murine colon cancer cells. Oncoimmunology 3, e28131. [ Links ]

Campana L, Clover A, Valpione S, Quaglino P, Gehl J, et al. 2016. Recommendations for improving the quality of reporting clinical electrochemotherapy studies based on qualitative systematic review. Radiol Oncol 50, 1-13. [ Links ]

Cemazar M, Kotnik T, Sersa G, Miklavcic D. 2015. Electroporation for electrochemotherapy and gene therapy. In: Electromagnetic fields in biology and medicine. 1st ed. CRC Press: Boca Ratón, USA, Pp 395-413. [ Links ]

Cemazar M, Ambrozic Avjustin J, Pavlin D, Sersa G, Poli A, et al. 2017. Efficacy and safety of electrochemotherapy combined with peritu- moral IL-12 gene electrotransfer of canine mast cell tumours. Vet Comp Oncol 15, 641-654. doi.org/10.1111/vco.12208. [ Links ]

Deodhar A, Dickfeld T, Single GW, Hamilton WC, Thornton RH, et al. 2011. Irreversible electroporation near the heart: ventricular arrhythmias can be prevented with ECG synchronization. Am J Roentgenol 196, 330-335. [ Links ]

Domenge C, Orlowski S, Luboinski B, De Baere T, Schwaab G, et al. 1996. Antitumor electrochemotherapy. New advances in the clinical protocol. Cancer 77, 956-963. doi.org/10.1002/ (SICI)1097-0142(19960301)77:5<956::AID-CNCR23>3.0.CO;2-1. [ Links ]

Escoffre JM, Rols MP. 2012. Electrochemotherapy: progress and pros pects. Curr Pharm Design 18, 3406-3415. [ Links ]

Gehl J. 2003. Electroporation: theory and methods, prospectives for drug delivery, gene therapy and research. Acta Physiol Scand 177, 437-447. [ Links ]

Gerlini G, Gennaro PD, Borgognoni L. 2012. Enhancing anti-melanoma immunity by electrochemotherapy and in vivo dendritic-cell acti vation. Oncoimmunology 1, 1655-1657. [ Links ]

Gerlini G, Sestini S, Di Gennaro P, Urso C, Pimpinelli N, et al. 2013. Dendritic cells recruitment in melanoma metastasis treated by electrochemotherapy. Clin Exp Metastasis 30, 37-45. [ Links ]

Jarm T, Cemazar M, Miklavcic D, Sersa G. 2010. Antivascular effects of electrochemotherapy: implications in treatment of bleeding metas-tases. Expert Rev Anticanc 10, 729-746. doi.org/10.1586/era.10.43. [ Links ]

Kodre V, Cemazar M, Pecar J, Sersa G, Cor A, et al. 2009. Electrochemotherapy compared to surgery for treatment of canine mast cell tumours. In vivo 23, 55-62. [ Links ]

Kotnik T, Pucihar G, Miklavcic D. 2010. Induced transmembrane voltage and its correlation with electroporation-mediated molecular transport. J Membr Biol 236, 3-13. [ Links ]

Kranjc M, Bajd F, Sersa G, Miklavcic D. 2011. Magnetic resonance electrical impedance tomography for monitoring electric field dis tribution during tissue electroporation. IEEE Trans Med Imaging 30, 1771-1778. doi 10.1109/TMI.2011.2147328. [ Links ]

Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Almeri ES, et al. 2009. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Ceath Differ 16, 3-11. [ Links ]

Li S, Zhang X, Xia X. 2002. Regression of tumor growth and induction of long-term antitumor memory by interleukin 12 electro-gene therapy. J Natl Cancer Inst 94,762-768. [ Links ]

Lowe RJ. 2016. Benefits and side-effects of electrochemotherapy in veterinary patients. Proceedings of the 1st World Congress on Electroporation and Pulsed Electric Fields in Biology, Medicine and Food & Environmental Technologies , Springer: Singapore, Pp 437-440. [ Links ]

Lowe R, Gavazza A, Impellizeri J, Soden DM, Lubas G. 2017. The treatment of canine mast cell tumours with electrochemotherapy with or without surgical excision. Vet Comp Oncol 15, 775-784. [ Links ]

Maglietti F, Tellado M, Olaiz N, Mishinski SD, Marshall GR. 2016. Combined local and systemic bleomycin administration in electrochemotherapy to reduce the number of treatment sessions. Radiol Oncol 50, 58-63. [ Links ]

Maglietti F, Tellado M, Olaiz N, Michinski S, Marshall G. 2017. Minimally invasive electrochemotherapy procedure for treating nasal duct tumors in dogs using a single needle electrode. Radiol Oncol 51, 422-430. [ Links ]

Mahmood F, Hansen RH, Agerholm-Larsen B, Jensen KS, Iversen HK, et al. 2011. Diffusion-weighted MRI for verification of electropo ration-based treatments. J Membr Biol 240, 131-138. [ Links ]

Mali B, Jarm T, Corovic S, Snezna Paulin-Kosir M. 2008. The effect of electroporation pulses on functioning of the heart. Med Biol Eng Comput 46, 745-757. [ Links ]

Mali B, Jarm T, Snoj M, Sersa G, Miklavcic D. 2013. Antitumor effectiveness of electrochemotherapy: A systematic review and meta-analysis. Eur J Surg Oncol 39, 4-16. [ Links ]

Markelc B, Sersa G, Cemazar M. 2013. Differential mechanisms associated with vascular disrupting action of electrochemotherapy: intravital microscopy on the level of single normal and tumor blood vessels. PloS One 8, e59557. [ Links ]

Marty M, Sersa G, Garbay JR, Gehl J, Collins CG, et al. 2006. Electrochemotherapy - an easy, highly effective and safe treatment of cutaneous and subcutaneous metastases: results of ESOPE (European Standard Operating Procedures of Electrochemotherapy) study. Eur J Cancer Suppl 4, 3-13. [ Links ]

Miklavcic D, Mali B, Kos B, Heller R, Sersa G. 2014. Electrochemotherapy: from the drawing board into medical practice. Biomed Eng Online 13, 29-35. [ Links ]

Miklavcic D, Snoj M, Zupanic A, Kos B, Cemazar M, et al. 2010. Towards treatment planning and treatment of deep-seated solid tumors by electrochemotherapy. Biomed Eng Online 9, 10. doi. org/10.1186/1475-925X-9-10. [ Links ]

Miklavcic D, Sersa G, Brecelj E, Gehl J, Soden D, et al. 2012. Electrochemotherapy: Technological advancements for efficient electroporation-based treatment of internal tumors. Med Biol Eng Comput 50, 1213-1225. [ Links ]

Mir LM. 2006. Bases and rationale of the electrochemotherapy. Eur J Cancer Suppl 4, 38-44. [ Links ]

Mir LM, Banoun H, Paoletti C. 1988. Introduction of definite amounts of nonpermeant molecules into living cells after electropermeabilisation: direct access to the cytosol. Exp Cell Res 175, 15-25. [ Links ]

Mir LM, Tounekti O, Orlowski S. 1996. Bleomycin: revival of an old drug. Gen Pharmacol 27, 745-748. [ Links ]

Orlowski S, Belehradek JJ, Paoletti C, Mir LM. 1988. Transient electropermeabilisation of cells in culture. Increase of the cytotoxicity of anticancer drugs. Biochem Pharmacol 37, 4727-4733. [ Links ]

Pavliha D, Music M M, Sersa G, Miklavcic D. 2013. Electroporation-based treatment planning for deep-seated tumors based on automatic liver segmentation of MRI images. PloS One 8, e69068. doi.org/10.1371/journal.pone.0069068. [ Links ]

Pavlin D, Cemazar M, Kamensek U, Tozon N, Pogacnik A, et al. 2009. Local and systemic antitumor effect of intratumoral and peritumoral IL-12 electrogene therapy on murine sarcoma. Cancer Biol Ther 8, 2114-2122. [ Links ]

Patnaik A, Ehler W, MacEwen E. 1984. Canine cutaneous mast cell tumor: morphologic grading and survival time in 83 dogs. Vet Pathol 21, 469-474. [ Links ]

Pierini A, Lowe R, Granziera V, Marchetti V, Lubas G. 2016. Electrochemotherapy with intravenous bleomycin for treatment of Feline Squamous Cell Carcinoma: Experience in 12 cats. J Vet Intern Med 30, 14-15. [ Links ]

Poddevin B, Orlowski S, Beleradek J, Mir LM. 1991. Very high cyto toxicity of bleomycin introduced into the cytosol of cells in culture. Biochem Pharmacol 42(S), 67-75. [ Links ]

Rols MP, Teissie J. 1992. Experimental evidence for the involvement of the cytoskeleton in mammalian cell electropermeabilisation. Biochim Biophys Acta 1111, 45-50. [ Links ]

Sersa G, Jarm T, Kotnik T, Coer A, Podkrajsek M, et al. 2008. Vascular disrupting action of electroporation and electrochemotherapy with bleomycin in murine sarcoma. Brit J Cancer 98, 388-398. doi 10.1038/sj.bjc.6604168. [ Links ]

Sersa G, Miklavcic D, Cemazar M, Rudlof Z, Pucihar G, et al. 2008. Electrochemotherapy in treatment of tumours. Eur J Surg Oncol 34, 232-240. [ Links ]

Sersa G, Miklavcic D, Cemazar M, Belehradek J, Jarm T, et al. 1997. Electrochemotherapy with CDDP on LPB sarcoma: comparison of the anti-tumor effectiveness in immunocompotent and immunodeficient mice. Bioelectrochem Bioenerg 43, 279-283. doi.org/10.1016/S0302-4598(96)05194-X. [ Links ]

Sersa G, Teissie J, Cemazar M, Signori E, Kamensek U, et al. 2015. Electrochemotherapy of tumors as in situ vaccination boosted by immunogene electrotransfer. Cancer Immunol Immun 64, 1315-1327. doi.org/10.1007/s00262-015-1724-2. [ Links ]

Spugnini E, Vincenzi B, Citro G, Santini D, Dotsinsky I, et al. 2007a. Adjuvant electrochemotherapy for the treatment of incompletely excised spontaneous canine sarcomas. In vivo 21, 819-822. [ Links ]

Spugnini EP, Baldi A, Vincenzi B, Bongiorni F, Bellelli C, et al. 2007b. Intraoperative versus postoperative electrochemotherapy in high grade soft tissue sarcomas: a preliminary study in a spontaneous feline model. Cancer Chemotherar Pharm 59, 375-381. doi.org/10.1007/s00280-006-0281-y. [ Links ]

Spugnini EP, Arancia G, Porrello A, Colone M, Formisano G, et al. 2007c. Ultrastructural modifications of cell membranes induced by “electroporation” on melanoma xenografts. Microsc Res Tech 70, 1041-1050. doi.org/10.1002/jemt.20504. [ Links ]

Spugnini E, Renaud M, Buglioni S, Carocci F, Dragonetti E, et al. 2011. Electrochemotherapy with cisplatin enhances local control after surgical ablation of fibrosarcoma in cats: an approach to improve the therapeutic index of highly toxic chemotherapy drugs. J Transí Med 9, 152. doi.org/10.1186/1479-5876-9-152. [ Links ]

Spugnini E, Vincenzi B, Citro G, Dotsinsky I, Mudrov T, et al. 2011. Evaluation of Cisplatin as an electrochemotherapy agent for the treatment of incompletely excised mast cell tumors in dogs. J Vet Int Med 25, 407-411. doi.org/10.1111/j.1939-1676.2011.0678.x. [ Links ]

Spugnin E, Citro G, Mellone P, Dotsinsky I, Mudrov N, et al. 2007. Electrochemotherapy for localized lymphoma: a preliminary study in companion animals. J Exp Clin Cancer Res, 26, 343-346. [ Links ]

Spugnini E, Dotsinsky I, Mudrov N, Citro G, D’Avino A, et al. 2008. Biphasic pulses enhance bleomycin efficacy in a spontaneous canine genital tumor model of chemoresistance: Sticker sarcoma. J Ex Clin Can Res 27, 58. doi.org/10.1186/1756-9966-27-58. [ Links ]

Spugnini E, Porrello A. 2003. Potentiation of chemotherapy in companion animals with spontaneous large neoplasms by application of biphasic electric pulses. J Ex Clin Can Res 22, 571-580. [ Links ]

Spugnini E, Pizzuto M, Filipponi M, et al. 2015. Electroporation enhances bleomycin efficacy in cats with periocular carcinoma and advanced squamous cell carcinoma of the head. J Vet Int Med 29, 1368-1375. [ Links ]

Suzuki D, Anselmo J, Oliveira K, Freytag J, Rangel MMM, et al. 2015. Numerical model of dog mast cell tumor treated by electrochem otherapy. Artif Organs 39, 192-197. doi.org/10.1111/aor.12333. [ Links ]

Suzuki D, Marques C, Rangel M. 2016. Conductive gel increases the small tumor treatment with electrochemotherapy using needle electrodes. Artif Organs 40, 705-711. [ Links ]

Suzuki D, Berkenbrock J, Frederico MJS, Silva FRMB, Rangel MMM. 2018. Oral mucosa model for electrochemotherapy treatment of dog mouth cancer: Ex vivo, in silico and in vivo experiments. Artif Organs 42, 297-304. doi.org/10.1111/aor.13003. [ Links ]

Tafuto S, Arx CV, De Divitiis C, Maura CT, Palaia R, et al. 2015. Electrochemotherapy as a new approach on pancreatic cancer and on liver metastases. Int J Surg 21, S78-S82. doi.org/10.1016/j.ijsu.2015.04.095. [ Links ]

Teissie J, Golzio M, Rols MP. 2005. Mechanisms of cell membrane electropermeabili-zation: a mini-review of our present (lack of?) knowledge. Bioc Biop Acta 1724, 270-280. [ Links ]

Tounekti O, Pron G, Belehradek J, Mir LM. 1993. Bleomycin, an apop tosis-mimetic drug that induces two types of cell death depending on the number of molecules internalized. Cancer Res 53, 5462-5469. [ Links ]

Tozon N, Kodre V, Sersa G, Cemazar M. 2005. Effective treatment of perianal tumors in dogs with electrochemotherapy. Anticancer Res 25, 839-845. [ Links ]

Tozon N, Kodre V, Juntes P, Sersa G. 2010. Electrochemotherapy is highly effective for the treatment of canine perianal hepatoid adenoma and epithelioma. Acta Veterinaria 60, 285-302. [ Links ]

Tozon N, Pavlin D, Sersa G, Dolinsek T, Cemazar M. 2014. Electrochemotherapy with intravenous bleomycin injection: an observational study in superficial squamous cell carcinoma in cats. J Feline Med Surg 16, 291-299. [ Links ]

Tozon N, Tratar U, Znidar K, Sersa G, Teissie J, et al. 2016. Operating Procedures of the Electrochemotherapy for Treatment of Tumor in Dogs and Cats. Journal of Visualized Experiments 116, e54760. [ Links ]

1Tellado M, Maglietti F, Olaiz N, Michinski S, Marshall G. 2014. Electroquimioterapia como herramienta terapéutica en melanoma oral en caninos. Available at: http://vetoncologia.com/wp-content/uploads/2014/09/melanoma-oral-canino.pdf.

2Electroquimioterapia como herramienta terapéutica en melanoma oral en caninos. http://www.vetoncologia.com/wp-content/uploads/2014/09/melanoma-oral-canino.pdf. Retrieved:04 August 2018.

Received: January 21, 2018; Accepted: May 03, 2018

*Corresponding author: MMM Rangel; contato@vetcancer.com.br.

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License