SciELO - Scientific Electronic Library Online

 
vol.9 número2Oxiclozanida en bovinos lecheros del valle de Cajamarca, como una alternativa en el control de Calicophoron microbothrioides índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

Compartir


Journal of the Selva Andina Animal Science

versión impresa ISSN 2311-3766versión On-line ISSN 2311-2581

J.Selva Andina Anim. Sci. vol.9 no.2 La Paz  2022  Epub 01-Oct-2022

https://doi.org/10.36610/j.jsaas.2022.090200097 

Artículo de Revisión

Bacteriophage cocktail as a substitute for antimicrobials in companion animal dermatology

Yhann Pool Angelo Vallenas-Sánchez1  * 
http://orcid.org/0000-0003-1262-5959

María Fernanda Bautista-Valles2 
http://orcid.org/0000-0001-8684-210X

Fabiana Llaque-Chávarri2 
http://orcid.org/0000-0002-1542-0097

Martin Enrique Mendoza-Coello1 
http://orcid.org/0000-0002-9001-4689

1Antenor Orrego Private University. School of Veterinary Medicine and Animal Husbandry. Semillero de Investigación de Producción Animal Sostenible. Av. América Sur 3145, Urb. Monserrate. Trujillo, Peru.

2Antenor Orrego Private University. School of Veterinary Medicine and Animal Husbandry. Semillero de Investigación de Sanidad Animales de Compañía. Av. América Sur 3145, Urb. Monserrate. Trujillo, Peru.


Resumen

El presente estudio se enfoca en el uso de cocteles de fagos como sustituto de antibióticos en dermatología de animales de compañía. Para este propósito, se realizó una búsqueda sistemática en la base de datos de Scopus, con el criterio de búsqueda: “veterinary” and “bacteriophage” and “dermatology” en título de artículo, resumen y palabras clave durante el periodo 2010-2021. Siete estudios in vitro y un estudio in vivo en animales de compañía, por lo cual se añadieron aquellos realizados en animales de laboratorio. En esta revisión se discute y proyecta la utilización de cócteles de fagos líticos no transductores como terapéuticos de piodermas, asimismo, se revisa la resistencia a fagos y las estrategias para superarla, la comparación con los antibióticos, el uso de cócteles en otras especies animales, así como, la utilización de fagos individuales y cócteles en dermatología veterinaria, y los fagos autóctonos como estrategia cuando las colecciones de fagos de estudios previos no tienen los efectos deseados. Se concluye que los cócteles de autofagos líticos no transductores son una alternativa contra la resistencia antimicrobiana en dermatología de animales de compañía. Finalmente, se recomienda comparar el uso de estos cócteles con otros sustitutos de antibióticos y evaluar su posible sinergismo para reducir bacterias patógenas en piel.

Palabras clave: Antibióticos; enfermedades de piel; terapia de fagos; resistencia antimicrobiana; Staphylococcus; Pseudomonas; veterinaria

Abstract

The present study focuses on the use of phage cocktails as a substitute for antibiotics in companion animal dermatology. For this purpose, a systematic search was carried out in the Scopus database, with the search criteria: "veterinary" and "bacteriophage" and "dermatology" in article title, abstract and keywords during the period 2010-2021. Seven in vitro studies and one in vivo study in companion animals, for which those carried out in laboratory animals were added. In this review, the use of non-transducing lytic phage cocktails as therapeutics for pyodermas is discussed and projected, as well as the resistance to phages and the strategies to overcome it, the comparison with antibiotics, the use of cocktails in other animal species, as well as the use of individual phages and cocktails in veterinary dermatology, and autochthonous phages as a strategy when phage collections from previous studies do not have the desired effects. It is concluded that non-transducing lytic autophage cocktails are an alternative against antimicrobial resistance in companion animal dermatology. Finally, it is recommended to compare the use of these cocktails with other antibiotic substitutes and evaluate their possible synergism to reduce pathogenic bacteria on the skin.

Keywords: Antibiotics; skin diseases; phage therapy; antimicrobial resistance; Staphylococcus; Pseudomonas; veterinary

Introduction

In numerous instances, bacterial skin infections are the main reason for consultation in the veterinary practice of small animals1,2. These are mostly caused by Staphylococcus pseudintermedius, which is part of the skin microbiota of dogs1, along with S. aureus3,4.

Broad-spectrum systemic and local antibiotics are frequently used to eliminate them5,6, while they can be supplied by different routes, topical is the most used one, through various presentations such as shampoos, creams, and gels7,8. However, its inappropriate use has caused bacteria to develop antimicrobial resistance in companion animals9-12. Therefore, it has become a concerning public health problem since staphylococci, which are part of the human and animal microbiota, can act as a reservoir of resistance genes13.

At the same time, many of these skin conditions can be considered secondary and commonly related to intestinal pathologies14.

For this reason, in dermatology, oral probiotics15-17, essential oils18-20 and, sodium hypochlorite21 have been postulated as alternative treatments. However, bacteriophages, which are the natural predators of bacteria, are emerging as an excellent option, due to their lytic properties, even in multidrug-resistant bacteria. Likewise, its ability to multiply at the site of infection22 and its high specificity stand out as potential advantages, so it does not affect beneficial bacteria23. Despite these advantages, it is known that these viruses can transmit virulence and antimicrobial resistance genes through transduction24,25. Hence, the phages to be used as therapy must be genetically characterized.

On the other hand, autophages are an ongoing trend, especially when commercial products do not have the desired effect26. These types of bacteriophages are often isolated from environments in which the target bacteria are found, with the purpose of ensuring that this specific microorganism is part of their spectrum.

Figure 1a Publications of primary scientific articles on bacteriophages in veterinary dermatology. b Publications of scientific articles on bacteriophages in dermatology that used phages cocktails. Data collected from Scopus database (Selection criteria: Article title, abstract, and keywords: “veterinary” “bacteriophage” and “dermatology” during the period 2010-2021) 

In the last decade, many scientific articles regarding the use of bacteriophages for bacterial control in veterinary medicine have been published, nonetheless, there are only a few works in the dermatology field (Figure 1a). Similarly, there are few studies made on phage cocktails (Figure 1b).

The present work focuses on the use of non-transducing lytic bacteriophage cocktails as substitutes for antimicrobials to combat skin diseases of bacterial origin, the routes of application in dermatology, their comparison with antimicrobials and the use of autochthonous phages.

Materials and methods

A search was conducted in Scopus database from January 2010 to December 2021. The selection criteria were that the article title, abstract, and keywords had to contain the following terms: "veterinary", "bacteriophage", "dermatology" and " cocktail". The data obtained were classified according to animal species and the administration route studied. For the development of this article, studies that used phages for the detection of pathogenic bacteria, reduction of bacterial load in the carcass, and disinfection of facilities, in the same way, basic investigations (genotypic and phenotypic characterization) were excluded.

Development

Bacteriophages. Also known as phages, these are viruses that can infect and lyse bacteria27. Phages were reported28 and isolated for the first time29 at the beginning of the 20th century, giving rise to phage therapy (PT) four years after their discovery. Despite that, these viruses were displaced by antibiotics because the latter had a broader spectrum and affordable price30. At the present time, due to antimicrobial resistance and new discoveries in PT, phages have regained popularity and are being used to combat bacterial diseases, as the first report of PT in companion animals in 200631 exhibits. These viruses are abundant in nature32 and notorious for being very specific. Even though its specificity may occur at the strain level33-35, phages that infect more than one bacterial genus have been reported36.

Due to their infective cycle, they can be classified as virulent or lytic (PhL) and temperate (PhT). The virulent ones prevent bacterial multiplication, in contrast, PhT allows it when there is a low bacterial population37,38. Likewise, PhT are involved in the transmission of virulence and antimicrobial resistance genes27,39,40.

Lytic infection begins with the recognition of phage receptors on the bacterial surface, such as antigens41, pili42, glucan43, polysaccharides44, proteins45, and other structures, inducing viral adsorption, subsequently, the phage implants its genetic material (RNA or DNA) in bacteria and destroys the genetic material of the host bacteria by enzymatic action, proceeds to assemble and replicates. Finally, the new phages produce holins and endolysins, the former are carrier proteins that allow endolysins (enzymes) to cross the membrane to their site of action, degrading the peptidoglycan30 and thus forming pores, causing depolarization that produces bacterial lysis, thereby releasing new phages46. However, the lytic cycle can fail by destroying the bacterial genetic material and assembling phages with fragments of it, a phenomenon called transduction. Transduction can be generalized or specialized, the first mentioned, due to errors in the assembly of the new phages, produces viruses with exclusively bacterial and viral genetic material24,25, the latter is caused when the bacterial and viral genome is mixed, resulting in phages with both genomes24. If the bacterium has resistance and virulence genes, these will be transmitted in the next cycle25, therefore, non-transducing PhL should be used.

Phage resistance. Phages and bacteria have co-evolved, with bacteria developing strategies to evade and outcompete phages. Several cases of bacterial phage-resistance have been reported in veterinary medicine33,47-49, studying its different mechanisms such as loss of phage-receptors, modification of phage-receptors, CRISPR-Cas system, abortive system, and production of polysaccharide matrix50.

Some bacterial strategies focus on avoiding phage adsorption by modifying phage receptors, losing them, and producing polysaccharides. In response, phages can change their tail fibers to find newly altered receptors51 and produce depolymerases52. Likewise, bacteria can attack the genetic material of the phage using the CRISPR-Cas system, yet, some phages prevent the degradation of their genetic material by using a protein coat53. Finally, when the previous strategies are not enough to avoid viral infection, the bacteria resort to the abortive system54.

On the other hand, in cases of resistance to individual phages, phage cocktails49,55 and quorum quenching56-58. Can be used. In spite of that, phage resistance to phage cocktails has also been reported33,47,48, in these situations, quorum quenching is most likely the best alternative, but the composition of the phage cocktail could be changed as well.

While it is true that phage resistance is a problem for PT, phage-resistant bacteria have been reported to reduce their ability to grow and absorb nutrients33. Furthermore, phage-resistant bacteria were reported to exhibit sensitivity to antibiotics to which they had previously shown resistance and lower virulence59,60.

Bacteriophages versus antibiotics. Although topical antibiotics are commonly used on localized and superficial wounds61, they can generate an imbalance in the skin microbiota due to their broad spectrum62. In contrast, the specificity of bacteriophages allows other bacteria outside of their range to remain unaffected, ensuring that the beneficial microbiota proliferates without problem23,63.

Moreover, topical drugs can be diluted or inactivated by enzymes or other inflammatory mediators64. Unlike phages that, due to their continuous multiplication, penetrate tissues in the presence of active bacteria, which is particularly useful in the treatment of infections in tissues with less blood supply65.

It should be noted that antimicrobial resistance (AMR) is the main factor that has driven the search for other therapeutic alternatives. Systemic antibiotics tend to generate greater resistance than topical ones66,67, which are prescribed more frequently in canine dermatopathies68,69.

However, this has normalized empiric treatments70,71, carried out without the pertinent microbiological sensitivity tests, which can lead to an increase in AMR.

This resistance, in turn, has been partially increased by poor practices in the daily veterinary clinic, highlighting its preventive use in cases such as vaccinations, sterilizations, among others72. On the contrary, it has been reported that the use of bacteriophages as a preventive measure produces better results. In challenged mice with E. coli CVCC193, those who were inoculated with phages 24 h previously, showed a survival rate of 80-100 %, compared to those who were administered 3 h later (40-50%)73.

In general, chronic infections are difficult to treat successfully because of AMR, increasing the duration of treatment and putting the patient's life at risk74. Enteral antimicrobials are needed in higher concentrations to reach the skin due to the poor irrigation that these tissues have, contributing to the presentation of side effects in pets exposed to these drugs. The adverse effects presented by undergoing treatment on animals are varied and can affect their quality of life. Gastrointestinal problems, and in rarer cases, hemolytic anemias, and acute kidney damage75.

The use of viruses (phages) in pets can be ethically controversial because animal welfare could be compromised76,77, hence few studies have used phages as the sole treatment for bacterial infection in companion animals since it is not a common practice in Veterinary Medicine of small species. Still, a study indicated a positive perception of this alternative therapy, both from veterinarians and owners, so this could be an indicator of its future mass use12.

Despite this, bacteriophage-based therapies have been performed in canines with chronic external otitis, which received previous antimicrobial treatment, with a positive resolution after treatment31,78, hence, its use to treat persistent conditions can be considered a viable alternative without serious side effects.

At the same time, economically, medical costs increase in patients with resistant bacterial infections, while PT is believed to be less expensive than antibiotic therapy if a specialized center is available79.

Regarding administration, bacteriophages multiply logarithmically over antibiotics, hence they would need fewer applications80, thus reducing the treatment period.

Similar to antibiotics, PT can also be affected by bacterial resistance81,82, despite that, resistance to phages can be anticipated and used as part of a therapeutic strategy83. Among the strategies, the decrease in the virulence of phage-resistant strains60,84,85, and varied attenuation according to whether the therapy is performed with single phages, or cocktails, the latter presents better results86.

From a practical perspective, for phages to be widely used in the treatment of bacterial infections, they will have to be effective in combination with antibiotics87. It has been pointed out that phages can reduce the minimum inhibitory concentration (MIC) of drug-resistant bacterial strains, although this arises from the class of antibiotics and the concentration of bacteriophage-antibiotics that are used together88. Thus, phages could positively influence the sensitivity of multidrug-resistant bacteria89.

However, some authors have justified that PhL may be capable of horizontally transmitting AMR genes to other bacteria through generalized transduction90, which would be considered counterproductive for the use of PhL transducers therapeutically. In opposition to these findings, it was stated that antimicrobial resistance genes are rarely encoded in phages since this process rarely occurs in the phage lytic cycle91. At present, the role of phages in the transduction of AMR genes continues to generate debate, and more studies are needed in this regard.

Regarding the comparison of phages and antibiotics in vivo, one study showed that phages (1x109 PFU/animal) had a similar effect to vancomycin (15 mg/kg) and a better effect than clindamycin (20 mg/kg) in reducing skin lesions in laboratory mice with S. aureus ATCC 25923 (6x109 CFU)92.

Another study of a similar nature was made in groups of mice inoculated with P. aeruginosa and treated with phage ZCPA1 (1x109 PFU/mL) in single doses (reduction of 4 log10 of the total bacterial count) and multiple (>4 log10), they showed a 100 % resolution of the wounds and optimal regeneration of the skin, while the group treated with topical gentamicin (2 log10) presented expansion and enlargement of the affected area, which led to purulent wounds that did not heal93. Ultimately, the use of phages, either alone or with antibiotics, will reveal superior results than traditional antibiotic therapy.

Bacteriophage cocktails in veterinary medicine. As mentioned above, phages are very specific, which limits the spectrum of individual phages. For this reason, individual phages are combined to broaden the spectrum, this mixture is known as a phage cocktail, it can be simple or mixed26, the former can infect bacteria of the same genus and the latter several bacterial genera. Phage cocktails were extensively studied in production animals to combat pathogenic bacteria in different animal species, with excellent results (Table 1). Different routes of administration, oral and immersion, were tested as alternatives in veterinary dermatology. The first can be used to maintain intestinal health and indirectly protect the skin; phage titers can be reduced by changes in the pH of the gastrointestinal tract if they are not protected94-96, while the second can be used to directly treat skin lesions.

On the other hand, veterinary cocktails were used in liquid (water) or solid (food) media, with better results in liquid media (Table 1, 2, and 3).

Bacteriophages in companion animal dermatology. There are few studies on the use of phages in dermatology, either in veterinary or human medicine, however, they suggest phages could be useful to treat pyoderma4,97-100. This skin disease could be caused by a wide variety of microorganisms, such as S. aureus, susceptible to phage ΦSA012, when applied intravenously or intraperitoneally in a mouse suffering from mastitis caused by said bacteria97.

In addition, another report carried out in vivo in mice, indicated the efficacy of ΦDMSA-2 bacteriophage against methicillin-resistant S. aureus (MRSA). It was applied topically on a wound generated by infected surgical excision; in a period of 12 to 16 days, a complete re-epithelialization of the lesion and eradication of the infection was achieved100, indicating that phages are effective for infections caused by S. aureus.

On the other hand, phage VB_SauS_SH-St 15644 caused the lysis of 32 % of MRSA strains in vitro and was able to reduce the progress of the infection in vivo when applied subcutaneously in mice98. The low percentage of lytic activity could be due to the specificity of the phage, hence cocktails could be useful to avoid this problem. Similarly, topical applications of SaGU1 phage to mice were effective in preventing the aggravation of S. aureus infection, reducing the presence of bacteria99.

In addition, phages were useful in reducing the defense mechanisms of bacteria, such as phage phiIPLA-RODI, together with lytic protein CHAPSH3b, were able to reduce the formation of S. aureus biofilm, a reduction in viable bacteria was also observed after its application101.

Table 1 Effect of Phage cocktail usage in veterinary 

Bacteria Host Phage Phage Family Phage Dose Provenance of phage Route of administration Animal Result References
C. pertringens CPAS-7, CPAS-12, CPAS-15, CPAS-16, CPTA-37, CPLV-42 Siphoviridae 2.5x109 UFP/ Animal Poultry farms Buffer SM Chickens Reduction of mortality from 66.67 a 18.00 % 104
Water Reduction of mortality from 66.67 a 3.33 %
Food Reduction of mortality from 66.67 a 5.33 %
Salmonella gallinarum ST4, L13, SG3 Siphoviridae 1.0x108 UFP/ kg Sewage Food Chickens Reduction of mortality from 40.00 a 25.00 %. 105
Salmonella typhimurium ATCC 14028 SEP-1, SGP-1, STP-1, SS3eP-1, SalTP-2, SChP-1, SAP-1, SAP-2 - 5.0x109 UFP/ Animal Sewage and stool Food Piglets Reduction of Salmonella in stool 106
E. coli APEC TM1, TM2, TM3, TM4 Siphoviridae 1x1010 UFP/animal Sewage I.V. Japanese quail Reduction of mortality from 46.60 a 13.30 % 107
Aeromonas hydrophila 50AhydR13PP, 60AhydR15PP. Myoviridae 1x105 UFP/mL - Inmersion European anguilla Reduction of mortality from 60.00 a 20.00 % 108
25AhydR2PP Podoviridae
Pseudomonas fluorescens 22PfluR64PP, 67PfluR64PP, 71PfluR64PP, 98PfluR60PP. Podoviridae

I.P.: intraperitoneal, I.V.: intravenosa.

However, in vitro they verified the efficiency of phages to eliminate MRSP, and control the biofilm present, the phages used belonged to the families Myoviridae y Siphoviridae, of these vB_SpsS-SN8, vB_SpsS- SN10, vB_SpsS-SN11, vB_SpsS-SN13, phiSA012, ph 0044 and ph 0045 showed lytic activity. On the other hand, pSp-J and pSp-S prevented the formation of biofilm a dose low and the degraded it a higher dose34. However, these MRSPs were in vitro, and more studies are required to determine its effectiveness in vivo.

Pseudomonas aeruginosa is another bacteria frequently related with pyoderma, especially in canine otitis102. A case was reported31 of a patient of the Saint Bernard breed who suffered from P. aeruginosa, and when treated with a phage, presented an improvement without secondary effects 9 months after of the application of the phage, and the presence of the bacteria was no longer observe. Likewise, the phages ΦS12-1 Y ΦR18, of the families Myoviridae and Podoviridae, respectively, were found to have activity in vitro lytic against various strains of P. aeruginosa isolated from the skin of canines103.

Since P. aeruginosa is on the WHO priority list of multiresistant bacteria109, these studies are extremely important as it is an alternative to combat bacterial resistance.

Lastly, FAL has also been used against Klebsiella pneumoniae in vivo, the phage ZCKP8, of the Siphoviridae family, was applied by topical treatment on infected open wounds in mice. It was possible to close the injury by 99 % after 17 days, compared to the group control, in which the lesion was closed by 79.76 %, showing the re-epithelialization in those treated with phages110.

This suggests that there is great potential for the use of phages within the clinical medicine of small animals. In addition, its use has several advantages, like the facility to obtain them, because they have various provenances (Table 2 y 3).

Likewise, there also exists a variety of pathways of application (Table 2), facilitating its use according to the area to be treated. Knowing that 36 % of owners they prefer the topical route and 1% the parenteral routes111, the topical route can be used through creams or baths, facilitating the application for homeowners.

However, it may be difficult to control the viral dose and many times pets could lick themselves, interfering with treatment, so it would be appropriate to recommend the use of Elizabethan collar. The intradermal and subdermal pathways would be adequate for veterinarians, also, it would allow to have to control of the applied dose more accurately and protect the phages from external factors such as licks, UV rays, etc.

Cocktails in veterinary dermatology of small animals. While it is true that phages are highly specific, which reduces its range of infection, without embargo, phage cocktails can eradicate P. aeruginosa; there even exist reports showing lytic activity against multidrug-resistant bacterial strains (MDR), extensive drug resistant (XDR) and pandrugresistant (PDR)112.

The use of phage cocktail for treatment in dogs diagnosed with otitis by P. aeruginosa, used six phages (BC-BP-01 to BC-BP-06), showed lytic activity, with no apparent side effects, eradicating the disease78.

Regarding other bacteria that cause lesions in the skin, like E. coli, P. aeruginosa and S. aureus, a cocktail was applied by topical route of three different phages for each of these, managing to eradicate the infection in an approximate of 9 to 13 days. In the case of E. coli, 16.70 % of the lesions healed in 9 days, and the remaining in 13 days. Regarding P. aeruginosa, 55.50 % of the lesions were free of bacteria in 9 days, and 45.50 % in 13 days, lastly, in those lesions generated by S. aureus, 60 % healed in 9 days, and the remaining in 13 days after of the application of the phage cocktail113.

Regarding MRSA, the use of a phage cocktail has been reported, with 3 different phages of the family Myoviridae applied topically, achieving to decrease the bacterial load, being equal to or even more efficient than vancomycin114, it should be noted no cases of mortality or side effects were reported in the mice treated with phages.

The use of a phage cocktail is usually more effective compared to an individual phage; in lesions by K. pneumoniae in mice were treated with 5 individual phages, and a cocktail of 5 phages. The cocktail was more efficient to remove the bacterial charge and decreased the healing time of the wound, a difference of the individual phages121.

The use of phage cocktails to treat infections points to the need for phage banks, which collect, characterize, and conserve these viruses. However, to date there are very few establishments122. A worldwide network of such banks would drastically reduce the possibility of a bacterial outbreak difficult to deal with, however, at present, it is still a long process and complicated to assign phages for determined necessities123. Veterinary centers could choose to isolate bacteriophages from the residual water from medicated baths, or from physiological samples (skin and stool) of the patients, to create a phage bank belonging to the clinic with therapeutic purposes.

Table 2 Spectrum of phages used for fight bacteria pathogenic in veterinary dermatology in vitro 

Host bacteria Phage Family of phage Provenance of phage Resultado References
S. pseudintermedius (41 cepas) pSp-J Siphovirus Floor and water from animal parks Lysis plates 34
S. pseudintermedius (47 cepas) pSp-S
S. pseudintermedius E133 S. pseudintermedius E140 vB_SpsS-SN8, vB_SpsS-SN10, vB_SpsS-SN11, vB_SpsS-SN13 Siphoviridae Dog stool Lysis plates 40
S. schleiferi, S. intermedius y S. pseudintermedius PhiSA012 Myoviridae Sewage Lysis plates 115
S. pseudintermedius SP015, SP017, SP197, SP251, SP253. ɸDP001 Siphoviridae Dog saliva Lysis plates 116
S. pseudintermedius SP015, SP017, SP070, SP145, SP188, SP195, SP197, SP251, SP253, SP276. ϕSA039 Myoviridae Sewage Lysis plates
S. pseudintermedius SP015, SP017, SP070, SP197, SP251, SP253, SP276. ϕSA012 Myoviridae Sewage Lysis plates
P. aeruginosa BrSP1 Myoviridae Sewage Lysis plates 117
S. pseudintermedius 625, 2854, CCM 2885, CCM 7315, CCM 7829, CCM 7830, 33, 35, 259, 621. QT1 Siphoviridae Félix d'Hérelle Collection Lysis plates 118
Staphylococcus spp. W15, W17, W33, W31, W36 Myoviridae Sea water Lysis plates 119
P. aeruginosa pPa_SNUABM_DT01 Myoviridae Water samples Lysis plates 120

Autochthonous phages or autophages. From a practical point of view, commercial products and phage collections from universities and various research centers could be used, where the use of products commercials of phages has been reported118. However, its high specificity could limit the effect expected, because it is possible that the bacteria present in patients are not susceptible to these. Facing this scenario, phages can be obtained or isolated from the patient where the pathogenic agent is found, calling this virus autochthonous phage or autophage26,126. In addition, the exogenous phage is also considered autophage, which is applied in an individual so it can later be reisolated127.

Autochthonous phages can be used as a cocktail to reduce the pro- probability of phage resistance and enlarge its spectrum. As described previously, phages can be obtained from the skin and feces (Table 2), and it is considered one of the main sources of autophages in dermatology of small animals.

In dogs, several phages have been reported like T4virus, Jerseyvirus, T5virus, Phix174microvirus, N4virus, T7virus, Bppunalikevirus, Bxz1virus, likewise, bacteriophages belonging to the families Myoviridae, Podoviridae, Siphoviridae and others not identified in the virome fecal of healthy dogs and those with enteropathy128,129.

Table 3 Effect of the utilization of phages against pathogenic bacteria in dermatology in vivo 

Bacteria host Phage Phage dose (UFP/ animal) Phage provenance Via Nnmber of dosis Especie animal Result References
P. aeruginosa BC-BP-01, BC-BP-02, BC-BP-03, BC-BP-04, BC-BP-05, BC-BP-06. 6x105 - Topica 1 Canis familiaris Reduction of P. aeruginosa 78
S. aureus ATCC 25923 F1, F4, F7, F8, F9, F10. 1x109 Nasal and pharyngeal swab and sewage SC 14 Mus musculus Reduction of clinical signs and clinical cure. 92
K. pneumoniae B5055 Kpn5 2x1010 Sewage Topica 1 Mus musculus Reduction of K. pneumoniae 124
S. aureus SA325 JD007 5x108 Chicken stool ID 1 Mus musculus Prevention and reduction of abscesses. 125

UFP: plate forming units. SC: subcutaneous. ID: intradermal.

Similarly, it was reported that the tick harbors a low amount of phages of the families: Myoviridae, Podoviridae, Siphoviridae, Sphae- rolipoviridae and Microviridae, which could be absorbed during the feeding moment or even arise in the same tick130.

Regard in vitro studies, the autophages vB_SpsS-SN8, vB_SpsS- SN10, vB_SpsS-SN11, vB_SpsS-SN13 were isolated from the skin and mucous membranes of a canine patient, autophages with lithic activity against S. pseudintermedius E133 and E14040, similarly, the autophage ɸDP001, found in the saliva of dog had lysed S. pseudointermedius116.

Regarding in vivo study92 a cocktail of autophages was used (F1, F4, F7, F8, F9 Y F10), which were obtained from nasal and pharyngeal swabs, and from sewage waters with lytic character against S. aureus in mice applied via subcutaneous. Similarly, autochthonous phages have been used in bovines, such as the phage SAvB14 that was isolated from the secretion of the gland mammary of cows with mastitis, with high activity lithic against S. aureus var. Bovis131.

The autophages have the advantage of being able to isolate them directly from the affected environment and prepare for its application in the future, being more specific and effective than a cocktail commercial126, emphasizing that the autophages will be more selective and more efficient due to the effect they have in the infection zone, allowing us to classify autophages as an alternative therapy.

Conclusion

Bacteriophages are an excellent substitute for anti biotics, since they are more specific and do not lyse beneficial bacteria, and have lithic activity against bacteria resistant to antimicrobials. There are bacteria resistant to phages, being unfavorable for phage therapy, however it has been reported many times that by acquiring this resistance, its virulence is reduced, and they become more sensitive to antibiotics that they used to be resistant to. Also, if the bacteria keeps its virulence and resistance to antimicrobials, phage cocktails or quorum quenching can be used. Regarding the routes of application, topical and parenteral are the optimal way for treating pyodermas in company animals.

In the present study, we emphasize that FaL should be used instead of tempered, to ensure bacterial lysis even when the bacteria are in low density. Similarly, these FaL must not possess resistance and virulent genes, so that the objective bacteria don’t acquire said genes through transduction. Likewise, the appropriate presentation is in the form of a cocktail, since it increases the lytic spectrum and decreases the risk of phage resistance. In short, autochthonous phages can be used when commercial phage cocktails of commercial phages or from previous studies do not have the effect desired. Thus, it can be concluded that the cocktails of lithic autophages without transduction are against antimicrobial resistance in small animal dermatology. Finally, it is recommended to compare the use of these cocktails with other antibiotic substitutes and assess their potential synergism to reduce bacteria pathogenic in the skin.

REFERENCES

1. García-Fonticoba R, Ferrer L, Francino O, Cuscó A. The microbiota of the surface, dermis, and subcutaneous tissue of dog skin. Anim Microbiome 2020;2(1):34. DOI: https://doi.org/10.1186/s42523-020-00050-8Links ]

2. Park Y, Oh J, Park S, Sum S, Song W, Chae J, et al. Antimicrobial resistance and novel mutations detected in the gyrA and parC genes of Pseudomonas aeruginosa strains isolated from companion dogs. BMC Vet Res 2020;16:111. DOI: https://doi.org/10.1186/s12917-020-02328-0Links ]

3. Papic B, Golob M, Zdovc I, Kusar D, Avbersek J. Genomic insights into the emergence and spread of methicillin-resistant Staphylococcus pseudintermedius in veterinary clinics. Vet Microbiol 2021;258:109119. DOI: https://doi.org/10.1016/j.vetmic.2021.109119Links ]

4. Ruiz-Ripa L, Simón C, Ceballos S, Ortega C, Zarazaga M, Torres C, et al. S. pseudintermedius and S. aureus lineages with transmission ability circulate as causative agents of infections in pets for years. BMC Vet Res 2021;17:42. DOI: https://doi.org/10.1186/s12917-020-02726-4Links ]

5. Guardabassi L, Schwarz S, Lloyd DH. Pet animals as reservoirs of antimicrobial-resistant bacteria. J Antimicrob Chemother 2004;54(2):321-32. DOI: https://doi.org/10.1093/jac/dkh332Links ]

6. Hughes LA, Williams N, Clegg P, Callaby R, Nuttall T, Coyne K, et al. Cross-sectional survey of antimicrobial prescribing patterns in UK small animal veterinary practice. Prev Vet Med 2012;104(3):309-16. DOI: https://doi.org/10.1016/j.prevetmed.2011.12.003Links ]

7. Loeffler A, Lloyd D. What has changed in canine pyoderma? A narrative review. Vet J 2018;235:73-82. DOI: https://doi.org/10.1016/j.tvjl.2018.04.002Links ]

8. Ramos SJ, Woodward M, Hoppers SM, Liu CC, Pucheu-Haston CM, Mitchell MS. Residual antibacterial activity of canine hair treated with five mousse products against Staphylococcus pseudintermedius in vitro. Vet Dermatol 2019;30(3):183-e57. DOI: https://doi.org/10.1111/vde.12737Links ]

9. Palomino-Farfán JA, Alvarez L, Siuce J, Calle S. Antimicrobial resistance in coagulase-positive staphylococci (CoPS) isolated from dogs with external otitis. Rev Inv Vet Perú 2020;31(1):e17558. DOI: https://doi.org/10.15381/rivep.v31i1.17558Links ]

10. Lee GY, Yang SJ. Comparative assessment of genotypic and phenotypic correlates of Staphylococcus pseudintermedius strains isolated from dogs with otitis externa and healthy dogs. Comp Immunol Microbiol Infect Dis 2020;70:101376. DOI: https://doi.org/10.1016/j.cimid.2019.101376Links ]

11. Ortiz-Díez G, López R, Sánchez-Díaz AM, Turrientes MC, Baquero MR, Luque R, et al. Epidemiology of the colonization and acquisition of methicillin-resistant staphylococci and vancomycin-resistant enterococci in dogs hospitalized in a clinic veterinary hospital in Spain. Comp Immunol Microbiol Infect Dis 2020;72:101501. DOI: https://doi.org/10.1016/j.cimid.2020.101501Links ]

12. Rhys-Davies L, Ogden J. Vets' and pet owners' views about antibiotics for companion animals and the use of phages as an alternative. Front Vet Sci 2020;7:513770. DOI: https://doi.org/10.3389/fvets.2020.513770Links ]

13. Rossi CC, Andrade-Oliveira AL, Giambiagi-deMarval M. CRISPR tracking reveals global spreading of antimicrobial resistance genes by Staphylococcus of canine origin. Vet Microbiol 2019;232:65-9. DOI: https://doi.org/10.1016/j.vetmic.2019.04.009Links ]

14. Craig JM. Atopic dermatitis and the intestinal microbiota in humans and dogs. Vet Med Sci 2016;2(2):95-105. DOI: https://doi.org/10.1002/vms3.24Links ]

15. Kim H, Rather IA, Kim H, Kim S, Kim T, Jang J, et al. A double-blind, placebo controlled-trial of a probiotic strain Lactobacillus sakei Probio-65 for the prevention of canine atopic dermatitis. J Microbiol Biotechnol 2015;25(11):1966-9. DOI: https://doi.org/10.4014/jmb.1506.06065Links ]

16. Osumi T, Shimada T, Sakaguchi M, Tsujimoto H. A double-blind, placebo-controlled evaluation of orally administered heat-killed Enterococcus faecalis FK-23 preparation in atopic dogs. Vet Dermatol 2019;30(2):127-e36. DOI: https://doi.org/10.1111/vde.12725Links ]

17. Lee KI, Yun T, Ham J, Lee WK, Kang JH, Yang MP, et al. Clinical trial of oral administration of Bifidobacterium longum in dogs with atopic dermatitis. Korean J Vet Res 2020;60(1):19-24. DOI: https://doi.org/10.14405/kjvr.2020.60.1.19Links ]

18. Neves RCSM, Makino H, Cruz TPPS, Silveira MM, Sousa VRF, Dutra V, et al. In vitro and in vivo efficacy of tea tree essential oil for bacterial and yeast ear infections in dogs. Pesqui Vet Bras 2018;38(8):1597-607. DOI: https://doi.org/10.1590/1678-5150-pvb-5055Links ]

19. Sim JXF, Khazandi M, Chan WY, Trott DJ, Deo P. Antimicrobial activity of thyme oil, oregano oil, thymol and carvacrol against sensitive and resistant microbial isolates from dogs with otitis externa. Vet Dermatol 2019;30(6):524-e159. DOI: https://doi.org/10.1111/vde.12794Links ]

20. Sim JXF, Khazandi M, Pi H, Venter H, Trott DJ, Deo P. Antimicrobial effects of cinnamon essential oil and cinnamaldehyde combined with EDTA against canine otitis externa pathogens. J Appl Microbiol 2019;127(1):99-108. DOI: https://doi.org/10.1111/jam.14298Links ]

21. Banovic F, Olivry T, Bäumer W, Paps J, Stahl J, Rogers A, et al. Diluted sodium hypochlorite (bleach) in dogs: Antiseptic efficacy, local tolerability and in vitro effect on skin barrier function and inflammation. Vet Dermatol 2018;29(1):6-e5. DOI: https://doi.org/10.1111/vde.12487Links ]

22. Squires RA. Bacteriophage therapy for management of bacterial infections in veterinary practice: What was once old is new again. N Z Vet J 2018;66(5):229-35. DOI: https://doi.org/10.1080/00480169.2018.1491348Links ]

23. Upadhaya SD, Ahn JM, Cho JH, Kim JY, Kang DK, Kim SW, et al. Bacteriophage cocktail supplementation improves growth performance, gut microbiome and production traits in broiler chickens. J Anim Sci Biotechnol 2021;12(1):49. DOI: https://doi.org/10.1186/s40104-021-00570-6Links ]

24. Chiang YN, Penadés JR, Chen J. Genetic transduction by phages and chromosomal islands: The new and noncanonical. PLoS Pathog 2019;15(8):e1007878. DOI: https://doi.org/10.1371/journal.ppat.1007878Links ]

25. Fillol-Salom A, Alsaadi A, Sousa JAM, Zhong L, Foster KR, Rocha EPC, et al. Bacteriophages benefit from generalized transduction. PLoS Pathog 2019;15(7):e1007888. DOI: https://doi.org/10.1371/journal.ppat.1007888Links ]

26. Honorio-Javes CE, Vallenas-Sánchez Y, Bazán Pérez JT. Coctel de bacteriófagos como sustituto de promotores de crecimiento tipo antibiótico en avicultura. Sci Agropecu 2021;12(4):499-508. DOI: https://doi.org/10.17268/sci.agropecu.2021.054Links ]

27. Calero-Cáceres W, Ye M, Balcázar JL. Bacteriophages as environmental reservoirs of antibiotic resistance. Trends Microbiol 2019;27(7):570-7. DOI: https://doi.org/10.1016/j.tim.2019.02.008Links ]

28. Twort FW. An investigation on the nature of ultra-microscopic viruses. Lancet 1915;186(4814): 1241-3. DOI: https://doi.org/10.1016/S0140-6736(01)20383-3Links ]

29. D'Herelle F. On an invisible microbe antagonistic toward dysenteric bacilli: brief note by Mr. F. D'Herelle, presented by Mr. Roux. 1917. Res Microbiol 2007;158(7):553-4. DOI: https://doi.org/10.1016/j.resmic.2007.07.005Links ]

30. Murray E, Draper LA, Ross RP, Hill C. The Advantages and challenges of using endolysins in a clinical setting. Viruses 2021;13(4):680. DOI: https://doi.org/10.3390/v13040680Links ]

31. Marza JA, Soothill JS, Boydell P, Collyns TA. Multiplication of therapeutically administered bacteriophages in Pseudomonas aeruginosa infected patients. Burns 2006;32(5):644-6. DOI: https://doi.org/10.1016/j.burns.2006.02.012Links ]

32. Welsh JE, Steenhuis P, de Moraes KR, van der Meer J, Thieltges DW, Brussaard CPD. Marine virus predation by non-host organisms. Sci Rep 2020;10(1):5221. DOI: https://doi.org/10.1038/s41598-020-61691-yLinks ]

33. Christiansen RH, Madsen L, Dalsgaard I, Castillo D, Kalatzis PG, Middelboe M. Effect of bacteriophages on the growth of Flavobacterium psychrophilum and development of phage-resistant strains. Microb Ecol 2016;71(4):845-59. DOI: https://doi.org/10.1007/s00248-016-0737-5Links ]

34. Kim SG, Giri SS, Yun S, Kim SW, Han SJ, Kwon J, et al. Two novel bacteriophages control multidrug- and methicillin-resistant Staphylococcus pseudintermedius biofilm. Front Med (Lausanne) 2021;8:524059. DOI: https://doi.org/10.3389/fmed.2021.524059Links ]

35. Kuzminska-Bajor M, Sliwka P, Ugorski M, Korzeniowski P, Skaradzinska A, Kuczkowski M, et al. Genomic and functional characterization of five novel Salmonella-targeting bacteriophages. Virol J 2021;18(1):183. DOI: https://doi.org/10.1186/s12985-021-01655-4Links ]

36. Mateus L, Costa L, Silva YJ, Pereira C, Cunha A, Almeida A. Efficiency of phage cocktails in the inactivation of Vibrio in aquaculture. Aquaculture 2014;424-425:167-73. DOI: https://doi.org/10.1016/j.aquaculture.2014.01.001Links ]

37. Knowles B, Silveira CB, Bailey BA, Barott K, Cantu VA, Cobián-Güemes AG, et al. Lytic to temperate switching of viral communities. Nature 2016;531(7595):466-70. DOI: https://doi.org/10.1038/nature17193Links ]

38. Erez Z, Steinberger-Levy I, Shamir M, Doron S, Stokar-Avihail A, Peleg Y, et al. Communication between viruses guides lysis-lysogeny decisions. Nature 2017;541(7638):488-93. DOI: https://doi.org/10.1038/nature21049Links ]

39. Colomer-Lluch M, Jofre J, Muniesa M. Antibiotic resistance genes in the bacteriophage DNA fraction of environmental samples. PLoS One 2011;6(3):e17549. DOI: https://doi.org/10.1371/journal.pone.0017549Links ]

40. Moodley A, Kot W, Nälgård S, Jakociune D, Neve H, Hansen LH, et al. Isolation and characterization of bacteriophages active against methicillin-resistant Staphylococcus pseudintermedius. Res Vet Sci 2019;122:81-5. DOI: https://doi.org/10.1016/j.rvsc.2018.11.008Links ]

41. Gambino M, Nørgaard Sørensen A, Ahern S, Smyrlis G, Gencay YE, Hendrix H, et al. Phage S144, a new polyvalent phage infecting Salmonella spp. And Cronobacter sakazakii. Int J Mol Sci 2020;21(15):5196. DOI: https://doi.org/10.3390/ijms21155196Links ]

42. McCutcheon JG, Peters DL, Dennis JJ. Identification and characterization of type IV pili as the cellular receptor of broad host range Stenotrophomonas maltophilia bacteriophages DLP1 and DLP2. Viruses 2018;10(6):338. DOI: https://doi.org/10.3390/v10060338Links ]

43. Szymczak P, Filipe SR, Covas G, Vogensen FK, Neves AR, Janzen T. Cell wall glycans mediate recognition of the dairy bacterium Streptococcus thermophilus by bacteriophages. Appl Environ Microbiol 2018;84(23):e01847-18. DOI: https://doi.org/10.1128/AEM.01847-18Links ]

44. Ha E, Chun J, Kim M, Ryu S. Capsular polysaccharide is a receptor of a Clostridium perfringens bacteriophage CPS1. Viruses 2019;11(11):1002. DOI: https://doi.org/10.3390/v11111002Links ]

45. Li P, Lin H, Mi Z, Xing S, Tong Y, Wang J. Screening of polyvalent phage-resistant Escherichia coli strains based on phage receptor analysis. Front Microbiol 2019;10:850. DOI: https://doi.org/10.3389/fmicb.2019.00850Links ]

46. Santos SB, Costa AR, Carvalho C, Nóbrega FL, Azeredo J. Exploiting bacteriophage proteomes: The hidden biotechnological potential. Trends Biotechnol 2018;36(9):966-84. DOI: https://doi.org/10.1016/j.tibtech.2018.04.006Links ]

47. Carvalho CM, Gannon BW, Halfhide DE, Santos SB, Hayes CM, Roe JM, et al. The in vivo efficacy of two administration routes of a phage cocktail to reduce numbers of Campylobacter coli and Campylobacter jejuni in chickens. BMC Microbiol 2010;10:232. DOI: https://doi.org/10.1186/1471-2180-10-232Links ]

48. Fischer S, Kittler S, Klein G, Glünder G. Impact of a single phage and a phage cocktail application in broilers on reduction of Campylobacter jejuni and development of resistance. PLoS One 2013;8(10):e78543. DOI: https://doi.org/10.1371/journal.pone.0078543Links ]

49. Richards PJ, Connerton PL, Connerton IF. Phage biocontrol of Campylobacter jejuni in chickens does not produce collateral effects on the gut microbiota. Front Microbiol 2019;10:476. DOI: https://doi.org/10.3389/fmicb.2019.00476Links ]

50. Brockhurst MA, Koskella B, Zhang QG. Bacteria-phage antagonistic coevolution and the implications for phage therapy. In: Harper DR, Abedon ST, Burrowes BH, McConville ML, editors. Bacteriophages. Switzerland: Springer Cham; 2020. p. 1-21. DOI: https://doi.org/10.1007/978-3-319-40598-8_7-1Links ]

51. Munsch-Alatossava P, Alatossava T. The extracellular phage-host interactions involved in the bacteriophage LL-H infection of Lactobacillus delbrueckii ssp. lactis ATCC 15808. Front Microbiol 2013;4:408. DOI: https://doi.org/10.3389/fmicb.2013.00408Links ]

52. Wang C, Li P, Niu W, Yuan X, Liu H, Huang Y, et al. Protective and therapeutic application of the depolymerase derived from a novel KN1 genotype of Klebsiella pneumoniae bacteriophage in mice. Res Microbiol 2019;170(3):156-64. DOI: https://doi.org/10.1016/j.resmic.2019.01.003Links ]

53. Malone LM, Warring SL, Jackson SA, Warnecke C, Gardner PP, Gumy LF, et al. A jumbo phage that forms a nucleus-like structure evades CRISPR-Cas DNA targeting but is vulnerable to type III RNA-based immunity. Nat Microbiol 2020;5(1):48-55. DOI: https://doi.org/10.1038/s41564-019-0612-5Links ]

54. Castillo D, Rørbo N, Jørgensen J, Lange J, Tan D, Kalatzis PG, et al. Phage defense mechanisms and their genomic and phenotypic implications in the fish pathogen Vibrio anguillarum. FEMS Microbiol Ecol 2019;95(3):fiz004. DOI: https://doi.org/10.1093/femsec/fiz004Links ]

55. Quiroz-Guzmán E, Peña-Rodriguez A, Vázquez-Juárez R, Barajas-Sandoval DR, Balcázar, JL, Martínez-Díaz SF. Bacteriophage cocktails as an environmentally-friendly approach to prevent Vibrio parahaemolyticus and Vibrio harveyi infections in brine shrimp (Artemia franciscana) production. Aquaculture 2018;492:273-9. DOI: https://doi.org/10.1016/j.aquaculture.2018.04.025Links ]

56. Hoque MM, Naser IB, Bari SM, Zhu J, Mekalanos JJ, Faruque SM. Quorum regulated resistance of Vibrio cholerae against environmental bacteriophages. Sci Rep 2016;6:37956. DOI: https://doi.org/10.1038/srep37956Links ]

57. Mion S, Plener L, Rémy B, Daudé D, Chabrière É. Lactonase SsoPox modulates CRISPR-Cas expression in gram-negative proteobacteria using AHL-based quorum sensing systems. Res Microbiol 2019;170(6-7):296-9. DOI: https://doi.org/10.1016/j.resmic.2019.06.004Links ]

58. Mion S, Rémy B, Plener L, Brégeon F, Chabrière E, Daudé D. Quorum quenching lactonase strengthens bacteriophage and antibiotic arsenal against Pseudomonas aeruginosa clinical isolates. Front Microbiol 2019;10:2049. DOI: https://doi.org/10.3389/fmicb.2019.02049Links ]

59. Fong K, Mu K, Rheault JG, Levesque RC, Kitts DD, Delaquis P, et al. Bacteriophage-insensitive mutants of antimicrobial-resistant Salmonella enterica are altered in their tetracycline resistance and virulence in caco-2 intestinal cells. Int J Mol Sci 2020;21(5):1883. DOI: https://doi.org/10.3390/ijms21051883Links ]

60. Kunttu HMT, Runtuvuori-Salmela A, Sundell K, Wiklund T, Middelboe M, Landor L, et al. Bacteriophage resistance affects Flavobacterium columnare virulence partly via mutations in genes related to gliding motility and the Type IX secretion system. Appl Environ Microbiol 2021;87(16):e0081221. DOI: https://doi.org/10.1128/AEM.00812-21Links ]

61. Frosini SM, Loeffler A. Treating canine pyoderma with topical antibacterial therapy. In Pract 2020;42(6):323-30. DOI: https://doi.org/10.1136/inp.m 2591 [ Links ]

62. Hoffman PS. Antibacterial discovery: 21st century challenges. Antibiotics (Basel). 2020;9(5):213. DOI: https://doi.org/10.3390/antibiotics9050213Links ]

63. Federici S, Nobs SP, Elinav E. Phages and their potential to modulate the microbiome and immunity. Cell Mol Immunol 2021;18(4):889-904. DOI: https://doi.org/10.1038/s41423-020-00532-4Links ]

64. Nainu F, Permana AD, Djide NJN, Anjani QK, Utami RN, Rumata NR, et al. Pharmaceutical approaches on antimicrobial resistance: prospects and challenges. Antibiotics (Basel) 2021;10(8): 981. DOI: https://doi.org/10.3390/antibiotics10080981Links ]

65. Kwiatek M, Parasion S, Nakonieczna A. Therapeutic bacteriophages as a rescue treatment for drug-resistant infections - an in vivo studies overview. J Appl Microbiol 2020;128(4):985-1002. DOI: https://doi.org/10.1111/jam.14535Links ]

66. Beco L, Guaguère E, Lorente Méndez C, Noli C, Nuttall T, Vroom M. Suggested guidelines for using systemic antimicrobials in bacterial skin infections (1): diagnosis based on clinical presentation, cytology and culture. Vet Rec 2013;172(3):72-8.DOI: https://doi.org/10.1136/vr.101069Links ]

67. Hillier A, Lloyd DH, Weese JS, Blondeau JM, Boothe D, Breitschwerdt E, et al. Guidelines for the diagnosis and antimicrobial therapy of canine superficial bacterial folliculitis (Antimicrobial Guidelines Working Group of the International Society for Companion Animal Infectious Diseases). Vet Dermatol 2014;25(3):163-e43. DOI: https://doi.org/10.1111/vde.12118Links ]

68. Summers JF, Hendricks A, Brodbelt DC. Prescribing practices of primary-care veterinary practitioners in dogs diagnosed with bacterial pyoderma. BMC Vet Res 2014;10:240. DOI: https://doi.org/10.1186/s12917-014-0240-5Links ]

69. Goggs R, Menard JM, Altier C, Cummings KJ, Jacob ME, Lalonde-Paul DF, et al. Patterns of antimicrobial drug use in veterinary primary care and specialty practice: A 6-year multi-institution study. J Vet Intern Med 2021;35(3):1496-508. DOI: https://doi.org/10.1111/jvim.16136Links ]

70. Gómez-Poveda B, Moreno MA. Antimicrobial prescriptions for dogs in the capital of Spain. Front Vet Sci 2018;5:309. DOI: https://doi.org/10.3389/fvets.2018.00309Links ]

71. Alcantara GLC, Pinello KC, Severo M, Niza-Ribeiro J. Antimicrobial resistance in companion animals - Veterinarians' attitudes and prescription drivers in Portugal. Comp Immunol Microbiol Infect Dis 2021;76:101640. DOI: https://doi.org/10.1016/j.cimid.2021.101640Links ]

72. Singleton DA, Pinchbeck GL, Radford AD, Arsevska E, Dawson S, Jones PH, et al. Factors associated with prescription of antimicrobial drugs for dogs and cats, United Kingdom, 2014-2016. Emerg Infect Dis 2020;26(8):1778-91. DOI: https://doi.org/10.3201/eid2608.191786Links ]

73. Wu J, Zeng H, Qian X, Li Y, Xue F, Ren J, et al. Pre-treatment with phages achieved greater protection of mice against infection with Shiga toxin-producing Escherichia coli than post-treatment. Res Vet Sci 2022;150:72-8. DOI: https://doi.org/10.1016/j.rvsc.2022.03.022Links ]

74. Abedon ST. Use of phage therapy to treat long-standing, persistent, or chronic bacterial infections. Adv Drug Deliv Rev 2019;145:18-39. DOI: https://doi.org/10.1016/j.addr.2018.06.018Links ]

75. De Lucia M, Bardagi M, Fabbri E, Ferreira D, Ferrer L, Scarampella F, et al. Rifampicin treatment of canine pyoderma due to multidrug-resistant meticillin-resistant staphylococci: a retrospective study of 32 cases. Vet Dermatol 2017;28(2):171-e36. DOI: https://doi.org/10.1111/vde.12404Links ]

76. Consejo General de Veterinarios. Código deontológico para el ejercicio de la profesión veterinaria [Internet]. Madrid: Consejo General de Veterinarios; 2019 [citado 22 de mayo de 2022]. 36 p. Recuperado a partir de: https://colegioveterinarios.net/wp-content/uploads/2018/10/BORRADOR-C%C3%93DIGO-DEONTOL%C3%93GICO-PARA-EL-EJERCICIO-DE-LA-PROFESI%C3%93N-VETERINARIA-Versi%C3%B3n-Septiembre-de-2018.pdf [ Links ]

77. Petetta F, Ciccocioppo R. Public perception of laboratory animal testing: Historical, philosophical, and ethical view. Addict Biol 2021;26(6):e12991. DOI. https://doi.org/10.1111/adb.12991Links ]

78. Hawkins C, Harper D, Burch D, Anggård E, Soothill J. Topical treatment of Pseudomonas aeruginosa otitis of dogs with a bacteriophage mixture: a before/after clinical trial. Vet Microbiol 2010;146(3-4):309-13. DOI: https://doi.org/10.1016/j.vetmic.2010.05.014Links ]

79. Loganathan A, Manohar P, Eniyan K, VinodKumar CS, Leptihn S, Nachimuthu R. Phage therapy as a revolutionary medicine against Gram-positive bacterial infections. Beni Suef Univ J Basic Appl Sci 2021;10(1):49. DOI. https://doi.org/10.1186/s43088-021-00141-8Links ]

80. Principi N, Silvestri E, Esposito S. Advantages and limitations of bacteriophages for the treatment of bacterial infections. Front Pharmacol 2019;10: 513. DOI: https://doi.org/10.3389/fphar.2019.00513Links ]

81. Oechslin F. Resistance development to bacteriophages occurring during bacteriophage therapy. Viruses 2018;10(7):351. DOI: https://doi.org/10.3390/v10070351Links ]

82. Ngiam L, Schembri MA, Weynberg K, Guo J. Bacteriophage isolated from non-target bacteria demonstrates broad host range infectivity against multidrug-resistant bacteria. Environ Microbiol 2021;23(9):5569-86. DOI: https://doi.org/10.1111/1462-2920.15714Links ]

83. Shen Y, Loessner MJ. Beyond antibacterials - exploring bacteriophages as antivirulence agents. Curr Opin Biotechnol 2021;68:166-73. DOI: https://doi.org/10.1016/j.copbio.2020.11.004Links ]

84. Castillo D, Christiansen RH, Dalsgaard I, Madsen L, Middelboe M. Bacteriophage resistance mechanisms in the fish pathogen Flavobacterium psychrophilum: Linking genomic mutations to changes in bacterial virulence factors. Appl Environ Microbiol 2015;81(3):1157-67. DOI: https://doi.org/10.1128/AEM.03699-14Links ]

85. Henrici De Angelis L, Poerio N, Di Pilato V, De Santis F, Antonelli A, Thaller MC, et al. Phage resistance is associated with decreased virulence in KPC-producing Klebsiella pneumoniae of the clonal group 258 Clade II lineage. Microorganisms 2021;9(4):762. DOI: https://doi.org/10.3390/microorganisms9040762Links ]

86. Markwitz P, Olszak T, Gula G, Kowalska M, Arabski M, Drulis-Kawa Z. Emerging phage resistance in Pseudomonas aeruginosa PAO1 Is accompanied by an enhanced heterogeneity and reduced virulence. Viruses 2021;13(7):1332. DOI: https://doi.org/10.3390/v13071332Links ]

87. Berryhill BA, Huseby DL, McCall IC, Hughes D, Levin BR. Evaluating the potential efficacy and limitations of a phage for joint antibiotic and phage therapy of Staphylococcus aureus infections. Proc Natl Acad Sci USA 2021;118(10):e2008007118. DOI: https://doi.org/10.1073/pnas.2008007118Links ]

88. Gu Liu C, Green SI, Min L, Clark JR, Salazar KC, Terwilliger AL, et al. Phage-antibiotic synergy is driven by a unique combination of antibacterial mechanism of action and stoichiometry. mBio 2020;11(4):e01462-20. DOI: https://doi.org/10.1128/mBio.01462-20Links ]

89. Segall AM, Roach DR, Strathdee SA. Stronger together? Perspectives on phage-antibiotic synergy in clinical applications of phage therapy. Curr Opin Microbiol 2019;51:46-50. DOI: https://doi.org/10.1016/j.mib.2019.03.005Links ]

90. Mašlaňová I, Stříbná S, Doškař J, Pantůček R. Efficient plasmid transduction to Staphylococcus aureus strains insensitive to the lytic action of transducing phage. FEMS Microbiol Lett 2016;363(19):fnw211. DOI: https://doi.org/10.1093/femsle/fnw211Links ]

91. Enault F, Briet A, Bouteille L, Roux S, Sullivan MB, Petit MA. Phages rarely encode antibiotic resistance genes: a cautionary tale for virome analyses. ISME J 2017;11(1):237-47. DOI: https://doi.org/10.1038/ismej.2016.90Links ]

92. Tamariz JH, Lezameta L, Guerra H. Fagoterapia frente a infecciones por Staphylococcus aureus meticilino resistente en ratones. Rev Perú Med Exp Salud Publica 2014;31(1):69-77. DOI: https://doi.org/10.17843/rpmesp.2014.311.10Links ]

93. Rezk N, Abdelsattar AS, Elzoghby D, Agwa MM, Abdelmoteleb M, Aly RG, et al. Bacteriophage as a potential therapy to control antibiotic-resistant Pseudomonas aeruginosa infection through topical application onto a full-thickness wound in a rat model. J Genet Eng Biotechnol 2022;20(1):133. DOI: https://doi.org/10.1186/s43141-022-00409-1Links ]

94. Colom J, Cano-Sarabia M, Otero J, Cortés P, Maspoch D, Llagostera M. Liposome-encapsulated bacteriophages for enhanced oral phage therapy against Salmonella spp. Appl Environ Microbiol 2015;81(14):4841-9. DOI: https://doi.org/10.1128/AEM.00812-15Links ]

95. Kaikabo AA, AbdulKarim SM, Abas F. Evaluation of the efficacy of chitosan nanoparticles loaded FKAZ14 bacteriophage in the biological control of colibacillosis in chickens. Poult Sci 2017;96(2):295-302. DOI: https://doi.org/10.3382/ps/pew255Links ]

96. Tie K, Yuan Y, Yan S, Yu X, Zhang Q, Xu H, et al. Isolation and identification of Salmonella pullorum bacteriophage YSP2 and its use as a therapy for chicken diarrhea. Virus Genes 2018;54(3):446 -56.DOI: https://doi.org/10.1007/s11262-018-1549-0Links ]

97. Iwano H, Inoue Y, Takasago T, Kobayashi H, Furusawa T, Taniguchi K, et al. Bacteriophage FSA012 has a broad host range against Staphylococcus aureus and effective lytic capacity in a mouse mastitis model. Biology (Basel) 2018;7(1) :8. DOI: http://doi.org/10.3390/biology7010008Links ]

98. Ji J, Liu Q, Wang R, Luo T, Guo X, Xu M, et al. Identification of a novel phage targeting methicillin-resistant Staphylococcus aureus in vitro and in vivo. Microb Pathog 2020;149:104317. DOI: https://doi.org/10.1016/j.micpath.2020.104317Links ]

99. Shimamori Y, Mitsunaka S, Yamashita H, Suzuki T, Kitao T, Kubori T, et al. Staphylococcal phage in combination with Staphylococcus epidermidis as a potential treatment for Staphylococcus aureus-associated atopic dermatitis and suppressor of phage-resistant mutants. Viruses 2020;13(1):7. DOI: https://doi.org/10.3390/v13010007Links ]

100. Shetru MN, Karched M, Agsar D. Locally isolated broad host-range bacteriophage kills methicillin-resistant Staphylococcus aureus in an in vivo skin excisional wound model in mice. Microb Pathog 2021;152:104744. DOI: https://doi.org/10.1016/j.micpath.2021.104744Links ]

101. Duarte AC, Fernández L, De Maesschalck V, Gutiérrez D, Campelo AB, Briers Y. et al. Synergistic action of phage phiIPLA-RODI and lytic protein CHAPSH3b: a combination strategy to target Staphylococcus aureus biofilms. NPJ Biofilms Microbiomes 2021;7:39. DOI: https://doi.org/10.1038/s41522-021-00208-5Links ]

102. Son Y, Bae S. In vitro efficacy of N-acetylcysteine in combination with antimicrobial agents against Pseudomonas aeruginosa in canine otitis externa. Korean J Vet Res 2021;61(2):e16. DOI: https://doi.org/10.14405/kjvr.2021.61.e16Links ]

103. Furusawa T, Iwano H, Higuchi H, Yokota H, Usui M, Iwasaki T, et al. Bacteriophage can lyse antibiotic-resistant Pseudomonas aeruginosa isolated from canine diseases. J Vet Med Sci 2016;78(6):1035-8.DOI: https://doi.org/10.1292%2Fjvms.15-0310Links ]

104. Miller RW, Skinner EJ, Sulakvelidze A, Mathis GF, Hofacre CL. Bacteriophage therapy for control of necrotic enteritis of broiler chickens experimentally infected with Clostridium perfringens. Avian Dis 2010;54(1):33-40. DOI: https://doi.org/10.1637/8953-060509-Reg.1Links ]

105. Hong SS, Jeong J, Lee J, Kim S, Min W, Myung H. Therapeutic effects of bacteriophages against Salmonella gallinarum infection in chickens. J Microbiol Biotechnol 2013;23(10):1478-83. DOI: https://doi.org/10.4014/jmb.1304.04067Links ]

106. Seo BJ, Song ET, Lee K, Kim JW, Jeong CG, Moon SH, et al. Evaluation of the broad-spectrum lytic capability of bacteriophage cocktails against various Salmonella serovars and their effects on weaned pigs infected with Salmonella typhimurium. J Vet Med Sci 2018;80(6):851-60. DOI: https://doi.org/10.1292/jvms.17-0501Links ]

107. Naghizadeh M, Karimi Torshizi MA, Rahimi S, Dalgaard TS. Synergistic effect of phage therapy using a cocktail rather than a single phage in the control of severe colibacillosis in quails. Poult Sci 2019;98(2):653-63.DOI: https://doi.org/10.3382/ps/pey414Links ]

108. Schulz P, Robak S, Dastych J, Siwicki AK. Influence of bacteriophages cocktail on European eel (Anguilla anguilla) immunity and survival after experimental challenge. Fish Shellfish Immunol 2019;84:28-37. DOI: https://doi.org/10.1016/j.fsi.2018.09.056Links ]

109. Tacconelli E, Carrara E, Savoldi A, Harbarth S, Mendelson M, Monnet DL, Theuretzbacher U, Magrini N, et al. WHO Pathogens Priority List Working Group. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis 2018;18(3):318-27. DOI: https://doi.org/10.1016/S1473-3099(17)30753-3Links ]

110. Fayez MS, Hakim TA, Agwa MM, Abdelmoteleb M, Aly RG, Montaser NN, et al. Topically applied bacteriophage to control multi-drug resistant Klebsiella pneumoniae infected wound in a rat model. Antibiotics (Basel) 2021;10(9):1048. DOI: https://doi.org/10.3390/antibiotics10091048Links ]

111. Astaiza Martínez JM, Benavides Melo CJ, Muñoz García GK, Mora Muñoz MF, Cháves Velasquez CA. Principales hábitos de medicación por los propietarios de caninos que acuden a consulta veterinaria en Pasto, Nariño, Colombia. Rev Colomb Cienc Quim Farm 2016;45(1):92-108.DOI: https://doi.org/10.15446/rcciquifa.v45n1.58019Links ]

112. Shokri D, Soleimani-Delfan A, Fatemi SM. Assessment of phage cocktails with extended host range activity against antibiotic resistant strains of Pseudomonas aeruginosa. Comp Clin Pathol 2017;26(2):417-22.DOI: https://doi.org/10.1007/s00580-016-2394-yLinks ]

113. Gupta P, Singh HS, Shukla VK, Nath G, Bhartiya SK. Bacteriophage therapy of chronic nonhealing wound: clinical study. Int J Low Extrem Wounds 2019;18(2):171-5. DOI: https://doi.org/10.1177/1534734619835115Links ]

114. Kifelew LG, Warner MS, Morales S, Vaughan L, Woodman R, Fitridge R, et al. Efficacy of phage cocktail AB-SA01 therapy in diabetic mouse wound infections caused by multidrug-resistant Staphylococcus aureus. BMC Microbiol 2020;20(1):204. DOI: https://doi.org/10.1186/s12866-020-01891-8Links ]

115. Nakamura T, Kitana J, Fujiki J, Takase M, Iyori K, Simoike K, et al. Lytic activity of polyvalent staphylococcal bacteriophage PhiSA012 and its endolysin Lys-PhiSA012 against antibiotic-resistant staphylococcal clinical isolates from canine skin infection sites. Front Med (Lausanne) 2020;7:234. DOI: https://doi.org/10.3389/fmed.2020.00234Links ]

116. Azam AH, Kadoi K, Miyanaga K, Usui M, Tamura Y, Cui L, et al. Analysis host-recognition mechanism of staphylococcal kayvirus ?SA039 reveals a novel strategy that protects Staphylococcus aureus against infection by Staphylococcus pseudintermedius Siphoviridae phages. Appl Microbiol Biotechnol 2019;103(16):6809-23. DOI: https://doi.org/10.1007/s00253-019-09940-7Links ]

117. de Melo ACC, da Mata Gomes A, Melo FL, Ardisson-Araújo DMP, de Vargas APC, Ely VL, et al. Characterization of a bacteriophage with broad host range against strains of Pseudomonas aeruginosa isolated from domestic animals. BMC Microbiol 2019;19(1):134. DOI: https://doi.org/10.1186/s12866-019-1481-zLinks ]

118. Zeman M, Bárdy P, Vrbovská V, Roudnický P, Zdráhal Z, Růžičková V, et al. New genus Fibralongavirus in Siphoviridae phages of Staphylococcus pseudintermedius. Viruses 2019;11(12): 1143. DOI: https://doi.org/10.3390/v11121143Links ]

119. Urban-Chmiel R, Balicki I, Swiader K, Nowaczek A, Pyzik E, Stepien-Pysniak D, et al. The in vitro efficacy of eye drops containing a bacteriophage solution specific for Staphylococcus spp. isolated from dogs with bacterial conjunctivitis. Ir Vet J 2020;73(1):21. DOI: https://doi.org/10.1186/s13620-020-00175-xLinks ]

120. Kwon J, Kim SW, Kim SG, Kang JW, Jung WJ, Lee SB, et al. The Characterization of a novel phage, pPa_SNUABM_DT01, infecting Pseudomonas aeruginosa. Microorganisms 2021;9(10): 2040. DOI: https://doi.org/10.3390/microorganisms9102040Links ]

121. Chadha P, Katare OP, Chhibber S. In vivo efficacy of single phage versus phage cocktail in resolving burn wound infection in BALB/c mice. Microb Pathog 2016;99:68-77. DOI: https://doi.org/10.1016/j.micpath.2016.08.001Links ]

122. Anand T, Virmani N, Bera B, Vaid R, Kumar A, Tripathi B. Applications of personalised phage therapy highlighting the importance of bacteriophage banks against emerging antimicrobial resistance. Def Life Sci J 2020;5(4):305-14. DOI: https://doi.org/10.14429/dlsj.5.15760Links ]

123. Yerushalmy O, Khalifa L, Gold N, Rakov C, Alkalay-Oren S, Adler K, et al. The Israeli Phage Bank (IPB). Antibiotics 2020;9(5):269. DOI: https://doi.org/10.3390/antibiotics9050269Links ]

124. Kumari S, Harjai K, Chhibber S. Bacteriophage versus antimicrobial agents for the treatment of murine burn wound infection caused by Klebsiella pneumoniae B5055. J Med Microbiol 2011;60(Pt 2):205-10. DOI: https://doi.org/10.1099/jmm.0.018580-0Links ]

125. Ding B, Li Q, Guo M, Dong K, Zhang Y, Guo X, et al. Prevention of dermal abscess formation caused by Staphylococcus aureus using phage JD007 in nude mice. Front Microbiol 2018;9:1553. DOI: https://doi.org/10.3389/fmicb.2018.01553Links ]

126. Sevilla-Navarro S, Marín C, Cortés V, García C, Vega S, Catalá-Gregori P. Autophage as a control measure for Salmonella in laying hens. Poult Sci 2018;97(12):4367-73. DOI: https://doi.org/10.3382/ps/pey294Links ]

127. Rohde C, Resch G, Pirnay JP, Blasdel BG, Debarbieux L, Gelman D, et al. Expert opinion on three phage therapy related topics: bacterial phage resistance, phage training and prophages in bacterial production strains. Viruses 2018;10(4):178. DOI: https://doi.org/10.3390/v10040178Links ]

128. Moreno PS, Wagner J, Mansfield CS, Stevens M, Gilkerson JR, Kirkwood CD. Characterisation of the canine faecal virome in healthy dogs and dogs with acute diarrhoea using shotgun metagenomics. PLoS One 2017;12(6):e0178433. DOI: https://doi.org/10.1371/journal.pone.0178433Links ]

129. Moreno PS, Wagner J, Kirkwood CD, Gilkerson JR, Mansfield CS. Characterization of the fecal virome in dogs with chronic enteropathy. Vet Microbiol 2018;221:38-43. DOI: https://doi.org/10.1016/j.vetmic.2018.05.020Links ]

130. Wang S, Zhao T, Yu X, Lin Z, Hua X, Cui L. Characterization of tick viromes collected from dogs in China. Biosafety and Health 2020;2(2):79-88. DOI: https://doi.org/10.1016/j.bsheal.2020.03.002Links ]

131. Horiuk Y, Horiuk V, Kukhtyn M, Tsvihun A, Kernychnyi S. Characterization of lytic activity of phage SAvB14 on Staphylococcus aureus variant bovis. J Adv Vet Anim Res 2020;7(3):509-13. DOI: https://doi.org/10.5455/javar.2020.g447Links ]

Source of financing The authors declare that they received no specific funding for this article.

Conflicts of interest There is no conflict of interest in this research.

Acknowledgments The authors would like to thank the Universidad Privada Antenor Orrego de Trujillo.

Ethical considerations The authors declare that the writing of the article is developed by carefully using previous studies in the literature and acknowledge them through the respective cited authors and sources.

Authors' contribution to the articleVallenas-Sánchez Yhann Pool Angelo, contributed to the conception and design of study, acquisition, and analysis of data, discussion of results, writing of the manuscript, approval of the final version of the manuscript. Bautista-Valles Maria Fernanda, Llaque-Chávarri Fabiana, Mendoza-Coello Martin Enrique, contributed to the acquisition and analysis of data, discussion of the results, drafting of the manuscript, approval of the version final of the manuscript.

Article ID: 113/JSAAS/2022

Editor's Note: Journal of the Selva Andina Animal Science (JSAAS). All statements expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, editors and reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Received: May 01, 2022; Revised: August 01, 2022; Accepted: September 01, 2022

*Contact address: Antenor Orrego Private University. School of Veterinary Medicine and Animal Husbandry. Semillero de Investigación de Producción Animal Sostenible. Av. América Sur 3145, Urb. Monserrate. Trujillo, Peru. Yhann Pool Angelo Vallenas-Sánchez E-mail address: yvallenass1@upao.edu.pe

Creative Commons License Este es un articulo publicado en acceso abierto bajo una licencia Creative Commons