Next Article in Journal
Effect of Prior Antibiotic Use on Culture Results in People with Diabetes and Foot Osteomyelitis
Next Article in Special Issue
Isolation, Characterization of Pyraclostrobin Derived from Soil Actinomycete Streptomyces sp. HSN-01 and Its Antimicrobial and Anticancer Activity
Previous Article in Journal
Swine Colibacillosis: Global Epidemiologic and Antimicrobial Scenario
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Alternatives to Antimicrobial Treatment in Bovine Mastitis Therapy: A Review

by
Dragana Tomanić
1,
Marko Samardžija
2 and
Zorana Kovačević
1,*
1
Department of Veterinary Medicine, Faculty of Agriculture, University of Novi Sad, Trg Dositeja Obradovica 8, 21000 Novi Sad, Serbia
2
Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia
*
Author to whom correspondence should be addressed.
Antibiotics 2023, 12(4), 683; https://doi.org/10.3390/antibiotics12040683
Submission received: 10 March 2023 / Revised: 25 March 2023 / Accepted: 28 March 2023 / Published: 30 March 2023
(This article belongs to the Special Issue Antimicrobial Activity of Secondary Metabolites Produced in Nature)

Abstract

:
Despite preventive and therapeutic measures, mastitis continues to be the most prevalent health problem in dairy herds. Considering the risks associated with antibiotic therapy, such as compromised effectiveness due to the emergence of resistant bacteria, food safety issues, and environmental impact, an increasing number of scientific studies have referred to the new therapeutic procedures that could serve as alternatives to conventional therapy. Therefore, the aim of this review was to provide insight into the currently available literature data in the investigation of non-antibiotic alternative approaches. In general, a vast number of in vitro and in vivo available data offer the comprehension of novel, effective, and safe agents with the potential to reduce the current use of antibiotics and increase animal productivity and environmental protection. Constant progress in this field could overcome treatment difficulties associated with bovine mastitis and considerable global pressure being applied on reducing antimicrobial therapy in animals.

1. Introduction

Mastitis is one of the health disorders that is common in dairy herds, affecting production, animal health, welfare, and the economy of the industry worldwide [1,2]. The high incidence of mastitis is directly related to increased milk production [3], with an estimation that this disease affects between 15 and 20% of the dairy cow population each year [4]. Being known as a multifactorial disease, its incidence depends on pathogens, udder defence mechanisms, and the presence of environmental factors [5,6]. It is defined as inflammation of the mammary gland caused by many different bacteria strains [7], as well as fungi such as Candida spp. [8] and algae such as Prototheca [9]. Literature data show that more than 140 microorganisms are associated with the aetiology of mastitis, indicating that the most common mastitis-associated bacteria include Staphylococcus aureus, Streptococcus agalactiae, Escherichia coli, and Streptococcus uberis [10], making the treatment approach complex.
According to the type of clinical manifestation, bovine mastitis occurs in subclinical and clinical forms [11], ranging from mild, moderate, to severe cases [12]. Of the two forms of mastitis, clinical mastitis manifests with visible changes of the milk as well as clinical signs of infection and inflammation, identified by visual investigation. On the other hand, subclinical mastitis (SCM) is difficult to diagnose as the cow appears healthy, while the udder and milk show no visible changes [11]. However, an increased number of somatic cells and the presence of the causative agent are used as the means of detecting SCM. Reported as a more prevalent form [3], SCM is associated with higher losses compared to clinical mastitis [13].
The main treatment of bovine mastitis still relies on antibiotic administration [14]. However, its efficacy is decreasing because of growing drug resistance in bacteria, being considered as a leading global health problem [15]. We are currently facing a rapid global spread of resistance, with the estimation that antimicrobial resistance (AMR) is responsible for over 30,000 deaths per year in the EU [16] and 700,000 deaths per year globally, with a projection of causing millions of deaths [6,17]. In the EU alone, it is estimated that AMR costs EUR 1.5 billion annually in healthcare and productivity losses [18]. Hence, we need to raise awareness of including a multisectoral One Health approach in tackling complex problems such as AMR in order to attain optimal health for people, animals, and the environment [19]. Moreover, antibiotic residues in food and the environment have affected consumers’ concern, thus emphasizing the importance of the reduction of antibiotic use for treating bacterial infections in animals [11,20]. In addition, long-term antibiotic treatment may be required, with a significant economic impact in terms of lost milk production and the cost of the antibiotic [21,22].
Considering the facts mentioned above, the treatment of mastitis is recognized as one of the greatest challenges in recent times. Thus, this work aimed to provide a review of the literature data regarding the possible solutions recognized as alternative treatment approaches to existing conventional bovine mastitis therapy. The availability of vast amount of scientific data may support future research in the study, development, and production of new, effective pharmaceutical formulations with higher efficiency against resistant pathogens.

2. Conventional Strategies of Control and Treatment of Bovine Mastitis

2.1. Biosecurity

The prevention of infectious diseases is important for animal health, welfare, and production efficiency [23,24,25]. Although this risk can never be totally avoided, it can at least be minimized by the implementation of preventive and control strategies based on improved therapeutic protolocols, hygiene, biosecurity strategies, etc. [5,26]. The National Mastitis Council developed a “5-Point Plan” mastitis control program where numerous approaches for decreasing mastitis incidence are summarized [5,27].
When it comes to prevention, biosecurity measures minimize the risk of new pathogens entering a farm, their transmission within the farm [28,29], and antibiotic usage [25]. Researchers are trying to develop and implement biosecurity tools. Among others, the BIOCHECK CATTLE® protocol for dairy cattle has been developed and used for the biosecurity assessment [30] of farms in Belgium, Kyrgyzstan, North Macedonia, Portugal, and Romania, with a wide variation of biosecurity levels across the farms [31]. Even though the type of biosecurity measures might differ to a large degree between the farming systems [30,32], using this tool, implementation of biosecurity on cattle farms can be assessed in a standardized and reproducible manner [30].
Although the focus on biosecurity as a preventive tool in livestock herds has increased [33], studies demonstrated that most cattle farmers hardly implement all adequate biosecurity measures [34,35,36]. However, protective measures, especially when performed properly, are not always practical and cost time and money, which is important for stakeholders [37,38]. Therefore, the problem of mastitis still requires a wide application of antibiotics for prevention and treatment of mastitis. Probably, further education of farmers on proper implementation of biosecurity measures could improve preventive strategies of bovine mastitis. This way of mastitis management could decrease antibiotic use in prevention and treatment of this disease.

2.2. Antibiotic Therapy

Antibiotics are used in intensive livestock production for therapeutic and prophylactic purposes [39], with an estimation that more than 50% of all antimicrobials are used in veterinary medicine globally [15,40]. Van Boeckel et al. [41] projected that antimicrobial consumption will rise by 67% by 2030 in food-producing animals, while the expected rise according to Tiseo et al. [42] is reported to be 11.5%. Bovine mastitis is the most common indication for the use of antibiotics in dairy production [43]. The administration of antibiotics can be performed by local application of intramammary preparations, as well as the systemic application of antibiotics [22], while the intramammary route is the most common route of drug administration for bovine mastitis [44]. The most frequently used antibiotics in mastitis therapy are penicillins, sulfonamides, ampicillin, cloxacillin, and aminoglycosides [45,46]. In addition to antibiotic therapy, symptomatic and supportive therapy are of great importance, alleviating local inflammation of the mammary gland, enabling a better effect of antibiotics due to greater perfusion of the mammary gland as well as faster recovery and regeneration of milk production [22,47].
However, antibiotic therapy has led to numerous concerns regarding public health. Namely, antibiotic use in animals leads to the possibility of transmission of resistant microorganisms through the food chain [48,49], as well as through ecosystems [50]. Besides posing threat to public health, the residues of antibiotics in milk can be a problem in the dairy industry, affecting the technology of production of fermented milk products [40]. Additional consequences of inadequate antibiotic use include partly compromised efficacy, reduced food safety, and changes in cow’s milk [40,51]. Moreover, the financial profitability of the antibiotic should be taken into account [21]. During intramammary treatment with antibiotics in lactation, milk has to be discarded and can only be used again after the withdrawal period has expired, which is quite expensive [48,52]. Some of frequently applied antibiotics are considered critically important for human healthcare, representing an additional global concern related to the protection of public health [15]. Moreover, the discovery and development rate of new antibiotic agents is exceed by the current rate of resistance development [53], which is leading to the search for alternatives together with consumers’ demand for antibiotic-free products [54].

2.3. Vaccination

Among preventive strategies applied to minimize bovine mastitis, vaccination is taking significant place. Vaccines are common and effective tools for the control of infective diseases [55], but only few vaccines are fully effective in veterinary medicine [56]. The success of vaccination depends on the vaccine formulation, administration route, vaccination quality, and timely vaccination coverage [57,58].
Immunoprophylaxis enables a modern approach to solving mastitis, providing reduction of antibiotics in therapy, thus affecting antibiotic treatment limitations [59]. Mastitis vaccines are gaining more interest as tools in combating AMR, even though scientific papers in the field of vaccination against the mastitis-associated pathogens point to limited success in obtaining significant results [37]. Additionally, limited success is caused by limited range of protection, since a wide range of strains and their different mechanisms of pathogenesis can be present within an individual cow [21].
Most vaccines are designed to target S. aureus, S. uberis, S. agalactiae, and E. coli [22,49], while S. aureus mastitis has been particularly difficult to control as the most important etiological agent of bovine mastitis, causing over 50% of the reported cases of mastitis [60]. Various approaches to the vaccine developed against mastitis caused by S. aureus have been described in a few reports [37,59,61]. Collado et al. [62] reported the efficacy of the S. uberis subunit vaccine against bovine mastitis, with significantly reduced clinical signs of mastitis, bacterial count, rectal temperature, and daily milk yield losses.
Evaluation of a commercially available polyvalent vaccine against mastitis, Startvac®, containing inactivated E. coli J5 strain and inactivated S. aureus SP140 strain, demonstrated different results. Piepers et al. [63] reported more potent immune response and elimination of the bacteria from the mammary gland in comparison with nonvaccinated animals, whereas Landin et al. [64] reported no significant differences between vaccinated and unvaccinated groups. Furthermore, Tashakkori et al. [65] pointed out that vaccination with commercially available Startvac® and Mastivac® was not associated with the reduction of clinical mastitis incidence. However, in order to achieve desired results, vaccination on its own will not usually achieve the desired results unless the vaccination programme is part of an integrated control antimicrobial stewardship strategy utilizing a combination of control measures [22,38]. Nevertheless, improvement of national control programs on vaccination will enable implementation of this strategy as a part of an antimicrobial stewardship program.

3. Alternative Strategies—Potential Solutions

Given the importance of bovine mastitis as a public health problem, many in vivo and in vitro studies of alternative treatments for mastitis have been conducted. The introduction of new strategies such as nanotherapy; bacteriophage therapy; animal-, plant-, and bacteria-derived antimicrobials; and probiotics, among others, are aiming to replace conventional antibiotic treatment, as well as solving the problem of AMR. The main advantage of the listed non-antibiotic alternatives is the absence of resistance development [10,66]. Potential solutions, which have been promoted through the One Health concept, as well as numerous EU regulations, aim primarily at reducing antibiotic usage by 50% by 2030 and giving preference to alternative approaches before conventional drugs are applied [67].

3.1. Nanotherapy

Nanotechnology is an increasingly growing field of the 21st century with a broad range of applications in industry, engineering, electronics, environment, food, medicine, and consumer products [68,69]. By providing new tools and materials at the nanoscale level that are beneficial for public health [4], nanotechnology has enabled the development of novel treatment options. Interest in their application in veterinary medicine has been rapidly growing, even though scientific data of veterinary nanomedicine are still relatively new and scarce. Recently, nanotechnology has been started to be applied in veterinary medicine in prevention, diagnosis, and therapy. The different types of nanomaterials are also being used for animal breeding, reproduction, and nutrition, and as disinfectants.
Taking into consideration all limitations of antibiotic-based therapy, nanotherapy, by replacing commonly used antibiotics, minimizes the problem of AMR, as well as the problem of drug residues. By this way, nanotherapy has an economic impact, minimizing the amounts of discarded milk and the number of permanently removed cows in dairy herds [70,71,72]. Nanoparticles may serve as potential delivery systems that deliver drugs directly into the target cells [4], enabling the use of very low doses and decreasing the amount of the used drug and withdrawal time in farm animals, which leads to the reduction of cost and side effects [70,71]. Furthermore, these nanoparticles display important biopharmaceutical advantages such as higher intracellular uptake than other conventional forms of drug delivery systems, increasing the accumulation and the retention time of the drug, improving the antibacterial activity, decreasing AMR, and inhibiting the biofilm formation [73]. As an example, nitric-oxide-releasing polymeric particles as a delivery system could be used to combat bovine-mastitis-associated bacteria [74].
According to the literature data, nanoparticles such as silver [72,75], copper nanoparticles [72,75], zinc oxide nanoparticles [76], nanogels [4], and chitosan nanoparticles [77,78] have been reported to show positive results in bovine mastitis management. Such therapeutic techniques are gaining popularity as tools for managing S. aureus mastitis due to the antimicrobial activity, including antibiotic-resistant strains and antibiofim activity [4]. The synergistic effect of silver nanoparticles and antibiotics was also evaluated, and a successful combination was obtained using antibiotics, such as erythromycin, in combination with silver nanoparticles against S. aureus [20,60]. Considering all mentioned above, veterinary nanomedicine as an innovative approach could play a significant role in the improvement of animal health, welfare, and production [79]. Hence, nanotherapy has a huge potential in the control of the bovine mastitis.

3.2. Bacteriophage Therapy

Bacteriophages are defined as viruses that have the ability to infect and replicate inside bacteria, suppressing their proliferation [80,81]. The main advantage of bacteriophages as alternative antibiotics is their specificity. Phages target only the pathogens of interest without affecting the host’s microbiota [82,83]. However, their specificity is also their main limitation [82], due to the fact that a single bacteriophage will only work on a limited number of bacterial strains, meaning that various different phages are needed to treat all possible bacteria associated with infection [84]. Compared to antibiotics, phages multiply in the host cell while the specific host is present, while concentration of antibiotics decreases. Moreover, after solving infection, phages are degraded, while antibiotics can persist in nature for a long time [84]. In addition, unlike antibiotics or vaccines, phages can be easily isolated from the environment, leading to shorter product development time and reduced production costs [85]. Besides being very specific, environmentally friendly, having good biological safety, as well as ability to evolve and to multiply at the site of infections [53,86,87], phages are also praised for their low probability of resistance development [85]. Moreover, bacteriophages also have shown the potential for vaccine developments [22].
Several potential candidates have been identified for bacteriophage therapy against bovine-mastitis-associated S. aureus [81,88,89,90], S. agalactiae [91], E. coli [80,92], S. uberis [93], K. pneumoniae [94], and K. oxytoca [87]. Moreover, the bactericidal effect of phage achieved by affecting bacterial essential cellular processes [85] is making them valuable antibacterial agents with effectiveness against sensitive as well as resistant bacteria [83]. Furthermore, phage therapy will probably not completely replace conventional antibiotics, but it could be employed as an additional treatment option [82], since phages can be used alone, as cocktails, or synergistically with other antimicrobials [95]. Even though bacteriophages can be considered as potential candidates for reducing antibiotic use in livestock production and increasing animal productivity, limitations in their usage in terms of environmental stability require storage and certain special conditions, which limit their usage [66].

3.3. Phytotherapy

Phytotherapy is among the most promising and widely used alternative options in the prevention and treatment of different infections in livestock [96,97]. Plants are widely applied due to therapeutic efficacy, low risk of adverse effects, low manufacturing costs, reduced resistance, and low drug residues in animal products and the environment [11,98,99,100]. The antimicrobial, anti-inflammatory, antioxidant, and immunomodulatory efficacy of medicinal plants have been highlighted in numerous studies over the last few years [101,102,103,104]. Furthermore, the multi-target mode of action of plants might be a useful tool in controlling multi-drug resistance in animal pathogens [97,101].
Biological diversity of plants offers an endless supply of new candidates with potential as therapeutic agents [97]. Almost 50% of the main pharmaceuticals available today are derived from natural resources [101]. Moreover, phytotherapy is often utilized in combination with conventional treatment protocols [101,105]. A review paper by Cheesman et al. [106] provides insight into the possibility and advantages of synergistic therapy, highlighting that medical plant–antibiotic combinations not only enhance the antimicrobial effect but also can act as resistance-modifying agents, in addition to other benefits.
Scientific data have reported the effectiveness of many plant-derived compounds in the treatment of mastitis, where essential oils (EOs) such as aromatic oily liquids obtained from plant materials are employed in phytotherapy for a long period of time [96,107]. Physiological and therapeutic benefits displayed by EOs along with GRAS (generally recognized as safe) status provide their wide range of application in veterinary medicine [108]. Due to antimicrobial and antibiofilm activities [109] EOs are being considered as a subject of interest to the scientific community in the development of novel therapeutic options. The antimicrobial activity of several different EOs on common bovine mastitis pathogens was the subject of research in different in vitro studies [105,110,111,112,113,114,115,116,117,118], while some authors focused on their antibiofilm effect [118,119,120].
While there have been many in vitro studies on the efficacy of EOs relating the potential use in bovine mastitis treatment, only few of them targeted in vivo efficacy. Pharmaceutical formulations based on plant extracts and EOs under the form of a spray, gel, ointment, or infusion have been developed [99,114,121,122,123,124,125,126], and different results of clinical and bacteriological cures were reported. In addition, the commercial product Phyto-Mast® composed of extracts of Thymus vulgaris, Gaultheria procumbens, Glycyrrhiza uralensis, Angelica sinensis, and vitamin E, in the treatment of clinical mastitis in dairy cows, did not result in a resolution of clinical mastitis, nor a bacteriological cure [121]. However, Mullen et al. [122] reported that therapy effect of Cinnatube® and Phyto-Mast® was similar to conventional therapy, without an irritating effect on the udder. Furthermore, Tomanic et al. [126] suggested the possible use of an EO-based pharmaceutical formulation (Phyto-Bomat®) consisting of Thymus vulgaris, Thymus serpyllum, Origanum vulgare, and Satureja montana as a component in mastitis control programs due to the resolution of symptoms post-treatment, as well as the prevention of the development of clinical mastitis in cases with subclinical mastitis.
As mentioned earlier, intramammary applied antibiotics consequently lead to deposition of residues in milk, affecting food safety, as well as economic profitability [127]. However, Kovačević et al. [52] reported minimal milk residues of thymol and carvacrol, two major chemical compounds of Phyto-Bomat® pharmaceutical formulation, which after 24 h decreased to the same level as before application. Moreover, in the study conducted by McPhee et al. [128] thymol residues in milk samples of goats were only detecTable 12 h after treatment with Phyto-Mast®.
The literature data suggest that the limitation of EO application could be due to the instability, biodegradability, and low solubility in certain solutions, pointing out that the development of delivery systems, such as metal nanoparticles, could improve previously mentioned EO shortcomings [4]. Moreover, pharmacoeconomic analysis was applied in veterinary medicine for the first time in order to assess the clinical and economic value of the EO-based alternative (Phyto-Bomat), highlighting economic benefits (savings) for farmers associated with the use of this treatment, with a focus on subclinical mastitis, since it contributes to most of the financial losses [129]. In line with the aforementioned, phytotherapy has a great potential in the management of bovine mastitis, especially EO-based formulations.

3.4. Homeopathy

Although quite controversial, the use of homeopathy in food-producing animals is evidence-based [130,131], even though a small number of scientific studies on the evaluation of the efficacy of homeopathy have been conducted so far. As an alternative treatment method, homeopathy is enjoying increasing popularity, particularly on organic farms, as a way of combating AMR [131]. Homeopathy, which is based on a holistic approach of stimulating the animal’s immune system [132], besides clinical signs and the pathogen agents, considers also behaviour, constitution, and environmental factors [130].
However, references to homeopathy are limited concerning bovine mastitis. While some studies found negative or no effects of homeopathic therapy for mastitis [133,134], Mimoune et al. [135] reported an encouraging clinical success rate of tested homeopathic preparation in the treatment of this disease. Furthermore, in India [136], the testing of homeopathic combination medicine was effective and economical in the management of mastitis in lactating dairy cows. According to Zeise and Fritz [137], homeotherapy in combination with antibiotics could be one of the options for successful control of bovine mastitis. Regardless, homeopathy offers some great advantages—it does not induce an allergic reaction or other side effects, no residue problems, no withdrawal period for the product, and no environmental pollution. In addition, the remedies are relatively cheap [132,136]. On the other hand, this alternative treatment can be associated with disadvantages due to their simplified registration, as most homeopathic formulations are present in the market without pharmacological, toxicological, or clinical assessment [133]. Further research in the field of homeopathy is needed in order to prove the efficiency of this alternative approach.

3.5. Bacteria-Derived Antimicrobials

Antimicrobial peptides such as bacteriocins are ribosomally synthesized peptids produced by bacteria. Being biologically active against Gram-positive and Gram-negative bacteria, they represent an alternative solution to conventional antibiotics [49,138,139]. Compared to antibiotics, bacteriocins have a narrow spectrum of antimicrobial activity, targeting specific pathogenic organisms [140,141] and efficiency against antibiotic-resistant strains. Moreover, they show antimicrobial activity at lower concentrations than antibiotics [142]. Bacteriocins of lactic acid bacteria demonstrated in vitro inhibitory antimicrobial activity against mastitis-associated Gram-positive organisms such as S. uberis and S. agalactiae [49,138,139].
A bacteriocin produced by Lactococcus lactis known as nisin is reported in the literature data as a therapeutic option due to its efficacy against mastitis-associated pathogens and foodborne pathogens [49,139,143]. Nisin, as well as several other bacteriocins, have generally regarded as safe (GRAS) status, being approved for application as food preservatives [142,144]. Currently, nisin is available in some countries for use in the dairy industry for teat disinfection in the form of wipes (Wipe Out®) [49,139]. Furthermore, Bennett et al. [141] demonstrated promising results of bactofencin, nisin, and reuterin in bovine mastitis treatment, being active against multidrug-resistant clinical bovine mastitis isolates, while a study by Hernández-González [144] demonstrated the antimicrobial activity of nisin on biofilm-producing S. aureus cultures. Moreover, promising results of the aureocins A70 and A53 combination against S. aureus-associated bovine mastitis were reported by Coelho et al. [145]. Some bacteriocins can act synergistically with conventional antibiotics, reducing concentrations, undesirable side effects, and the prevalence of resistant strains [144].

3.6. Animal-Derived Antimicrobials

Lactoferrin is a multifunctional glycoprotein found in saliva, tears, bronqueal mucus, colostrum, and milk [21,22], with a broad range of biological activities including antimcrobial, antiinflammatory, immunomodulatory, anticatabolic, and antioxidative effects [146,147]. Several in vitro studies reported an antibacterial effect against some major mastitis-causing pathogens [148,149,150], showing not only bacteriostatic but also bactericidal and antifungal activities [151]. Moreover, it can be effective as a preventive or therapeutic option for bovine mastitis [151].
Milk contains not only lactoferrin but also antibacterial proteins such as lysozyme, immunoglobulins, lactoperoxidase, and β-defensin with a mechanism of action consisting of inactivation of bacteria, prevention of bacterial adherence to mammary tissue, and neutralization of toxins [151]. Lysozyme with a hydrolyzing effect on the essential bacterial cell component peptidoglycan leading to its lysis [49,152] was used successfully in increasing antibiotic efficacy against S. uberis and S. dysgalactiae associated with bovine mastitis [49]. Chaneton et al. [153] suggests that β-lactoglobulin and lactoferrin may complement each other against bacterial infection, due to the different effects on different bacteria, which expend their spectrum of antimicrobial activity [21]. It was reported that lactoferrin, lysozyme, and other peptids as potential non-antibiotic antimicrobial agents for the treatment and prevention of bovine mastitis can be used in combination with antibiotics [147,149,154].
When it comes to other animal-derived treatment options for mastitis, propolis is one of the most widely known and used natural products. As a natural resinous mixture produced by honeybees from substances collected from different plants [155,156], propolis is enriched with bees’ salivary and enzymatic secretions [157]. Furthermore, propolis has a complex chemical composition, with identified compounds such as polyphenols, terpenoids, steroids, and amino acids [155]. In addition, propolis has several biological activities such as antiviral, antibacterial, antifungal, immunostimulatory, antioxidant, imunomodulatory, hepatoprotective, and anti-inflammatory effects [155,158,159], leading to its application in the development of products for use in human and animal health [158].
In the study conducted in Croatia, it is described that propolis could be used in the development of an intramammary pharmaceutical formulation for the prevention and treatment of bovine mastitis during lactation [160,161]. Moreover, this innovative intramammary formulation of a non-alcoholic solution of propolis showed antibacterial, immunostimulatory, antioxidant, and anti-inflammatory activity, with the potential to become an alternative to conventional therapy [160,162]. Furthermore, Hegazi et al. [155] confirmed the efficacy of propolis as an antibacterial agent against bacterial strains isolated from mastitis, where propolis exhibited significant antimicrobial activity against ≈41% of the isolated pathogens, being more effective against Gram-positive bacteria than Gram-negative bacteria [155]. These results are in accordance with the work of Bačić et al. [161], where an in vitro tested propolis formulation also exhibited higher antimicrobial activity against Gram-positive bacteria than Gram-negative bacteria. Among all animal-derived antimicrobials, the clinical efficiency of pharmaceutical formulation based on propolis in the prevention and treatment of bovine mastitis can lead to the reduction of antibiotics usage and AMR in livestock production.

3.7. Other Alternatives

Other alternatives have included the use of probiotics [163,164], zeolites [165,166], cytokines [167,168,169,170], ozone [171,172,173], and aloe vera [174,175]. Zeolites are natural, microporous, three-dimensional crystalline materials with well-defined channels and cavities and various technological applications in veterinary medicine [176,177]. Moreover, a widely used zeolite is known as clinoptilolite, which has been proven as safe for veterinary use [178], acting as an antibacterial, antiviral, antioxidant, anti-diarrheic, and growth-promoting agent [165,166,178,179]. Đuričić et al. [165], studying dietary vibroactivated and micronised clinoptilolite, found out its effect on reducing the incidence of intramammary infections, while Ural [180] reported the efficacy of supplementation with 3% clinoptilolite in dairy cows in terms of milk yield and somatic cell count.
Probiotics are gaining more attention as an interesting alternative for the prevention or treatment of bovine mastitis [181,182]. Pellegrino et al. [183] demonstrated that lactic acid bacteria, as one of the components of the indigenous microbiota of the teat canal, could be used as candidates in mastitis prevention, while Klostermann et al. [184] reported that intramammary administration of a live culture of Lactococcus lactis in some cases may be as effective as antibiotic treatment. Researchers worldwide are trying to develop probiotic-based formulations for the prevention and treatment of mastitis, since they have shown great potential in this field.
In conclusion, much of the reviewed data have shown the opportunity of exploiting novel therapeutic approaches in overcoming the limitations of conventional antibiotic-based therapies. The reviewed alternative options will probably not completely replace conventional antibiotics, but they could be employed as an additional or supportive option in the treatment of infections. When it comes to EO-based pharmaceutical alternatives, Phyto-Mast and Cinnatube have great potential in achieving similar results as conventional therapy. Furthermore, Phyto-Bomat could be an important part of the mastitis control program, as a treatment approach in subclinical mastitis, and a supportive option in clinical mastitis. In addition, phytotherapy as an alternative approach has shown good results against resistant strains and biofilm formation. Moreover, nanotherapy and use of a propolis-based formulation have shown significant results in clinical studies of bovine mastitis therapy. As an important part of antimicrobial stewardship, the use of antimicrobial alternatives together with other strategies such as vaccination and biosecurity measures could help us to enhance public health by decreasing AMR.

Author Contributions

Conceptualization, D.T. and Z.K.; methodology, M.S. investigation, D.T.; writing—original draft preparation, D.T., M.S. and Z.K.; writing—review and editing, D.T., M.S. and Z.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Science Fund of the Republic of Serbia, PROMIS, #GRANT No. 6066966, InfoBomat.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Boboš, S.; Radinović, M.; Vidić, B.; Pajić, M.; Vidić, V.; Galfi, A. Mastitis therapy: Direct and indirect costs. Biotechnol. Anim. Husb. 2013, 29, 269–275. [Google Scholar] [CrossRef] [Green Version]
  2. Ali, T.; Raziq, A.; Wazir, I.; Ullah, R.; Shah, P.; Ali, M.I.; Han, B.; Liu, G. Prevalence of mastitis pathogens and antimicrobial susceptibility of isolates from cattle and buffaloes in Northwest of Pakistan. Front. Vet. Sci. 2021, 8, 1148. [Google Scholar] [CrossRef] [PubMed]
  3. Rogožarski, D.; Dimitrijević, G.; Dobrosavljević, I. Participation of diagnosed mastitits in cows in milk hygiene of Branicevo district in 2002. Arch. Vet. Med. 2011, 4, 65–71. [Google Scholar] [CrossRef]
  4. Neculai-Valeanu, A.S.; Ariton, A.M.; Mădescu, B.M.; Rîmbu, C.M.; Creangă, Ş. Nanomaterials and essential oils as candidates for developing novel treatment options for bovine mastitis. Animals 2021, 11, 1625. [Google Scholar] [CrossRef]
  5. Bradley, A.J. Bovine mastitis: An evolving disease. Vet. J. 2002, 164, 116–128. [Google Scholar] [CrossRef]
  6. Krömker, V.; Leimbach, S. Mastitis treatment—Reduction in antibiotic usage in dairy cows. Reprod. Domest. Anim. 2017, 52, 21–29. [Google Scholar] [CrossRef] [Green Version]
  7. Cvetnić, L.; Samardžija, M.; Habrun, B.; Kompes, G.; Benić, M. Microbiological monitoring of mastitis pathogens in the control of udder health in dairy cows. Slov. Vet. Res. 2016, 53, 131–140. [Google Scholar]
  8. Costa, E.O.d.; Gandra, C.; Pires, M.; Coutinho, S.; Castilho, W.; Teixeira, C. Survey of bovine mycotic mastitis in dairy herds in the State of São Paulo, Brazil. Mycopathologia 1993, 124, 13–17. [Google Scholar] [CrossRef]
  9. Huilca-Ibarra, M.P.; Vasco-Julio, D.; Ledesma, Y.; Guerrero-Freire, S.; Zurita, J.; Castillejo, P.; Barceló Blasco, F.; Yanez, L.; Changoluisa, D.; Echeverría, G. High Prevalence of Prototheca bovis Infection in Dairy Cattle with Chronic Mastitis in Ecuador. Vet. Sci. 2022, 9, 659. [Google Scholar] [CrossRef]
  10. Prusa, B. Alternative solutions to antibiotics in mastitis treatment for dairy cows-a review. Anim. Sci. Pap. Rep. 2020, 38, 117–133. [Google Scholar]
  11. Lopes, T.S.; Fontoura, P.S.; Oliveira, A.; Rizzo, F.A.; Silveira, S.; Streck, A.F. Use of plant extracts and essential oils in the control of bovine mastitis. Res. Vet. Sci. 2020, 131, 186–193. [Google Scholar] [CrossRef] [PubMed]
  12. Pedersen, R.R.; Krömker, V.; Bjarnsholt, T.; Dahl-Pedersen, K.; Buhl, R.; Jørgensen, E. Biofilm research in bovine mastitis. Front. Vet. Sci. 2021, 8, 656810. [Google Scholar] [CrossRef] [PubMed]
  13. Tezera, M.; Aman Ali, E. Prevalence and associated risk factors of Bovine mastitis in dairy cows in and around Assosa town, Benishangul-Gumuz Regional State, Western Ethiopia. Vet. Med. Sci. 2021, 7, 1280–1286. [Google Scholar] [CrossRef] [PubMed]
  14. Pascu, C.; Herman, V.; Iancu, I.; Costinar, L. Etiology of Mastitis and Antimicrobial Resistance in Dairy Cattle Farms in the Western Part of Romania. Antibiotics 2022, 11, 57. [Google Scholar] [CrossRef] [PubMed]
  15. Kovačević, Z.; Samardžija, M.; Horvat, O.; Tomanić, D.; Radinović, M.; Bijelić, K.; Vukomanović, A.G.; Kladar, N. Is There a Relationship between Antimicrobial Use and Antibiotic Resistance of the Most Common Mastitis Pathogens in Dairy Cows? Antibiotics 2022, 12, 3. [Google Scholar] [CrossRef]
  16. Gajdács, M.; Urbán, E.; Stájer, A.; Baráth, Z. Antimicrobial resistance in the context of the sustainable development goals: A brief review. Eur. J. Investig. Health Psychol. Educ. 2021, 11, 71–82. [Google Scholar] [CrossRef]
  17. Mestrovic, T.; Aguilar, G.R.; Swetschinski, L.R.; Ikuta, K.S.; Gray, A.P.; Weaver, N.D.; Han, C.; Wool, E.E.; Hayoon, A.G.; Hay, S.I. The burden of bacterial antimicrobial resistance in the WHO European region in 2019: A cross-country systematic analysis. Lancet Public Health 2022, 7, e897–e913. [Google Scholar] [CrossRef]
  18. European Commission. EU Action on Antimicrobial Resistance. Available online: https://health.ec.europa.eu/antimicrobial-resistance/eu-action-antimicrobial-resistance_en (accessed on 1 March 2023).
  19. Lancet, T. Antimicrobial resistance: Time to repurpose the Global Fund. Lancet 2022, 399, 335. [Google Scholar] [CrossRef]
  20. Rajamanickam, K.; Yang, J.; Chidambaram, S.B.; Sakharkar, M.K. Enhancing Drug Efficacy against Mastitis Pathogens—An In Vitro Pilot Study in Staphylococcus aureus and Staphylococcus epidermidis. Animals 2020, 10, 2117. [Google Scholar] [CrossRef]
  21. Gomes, F.; Henriques, M. Control of bovine mastitis: Old and recent therapeutic approaches. Curr. Microbiol. 2016, 72, 377–382. [Google Scholar] [CrossRef] [Green Version]
  22. Cheng, W.N.; Han, S.G. Bovine mastitis: Risk factors, therapeutic strategies, and alternative treatments—A review. Asian-Australas. J. Anim. Sci. 2020, 33, 1699–1713. [Google Scholar] [CrossRef] [PubMed]
  23. Barkema, H.W.; Green, M.; Bradley, A.J.; Zadoks, R. Invited review: The role of contagious disease in udder health. J. Dairy Sci. 2009, 92, 4717–4729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Alarcón, L.V.; Allepuz, A.; Mateu, E. Biosecurity in pig farms: A review. Porc. Health Manag. 2021, 7, 1–15. [Google Scholar] [CrossRef] [PubMed]
  25. Butucel, E.; Balta, I.; McCleery, D.; Morariu, F.; Pet, I.; Popescu, C.A.; Stef, L.; Corcionivoschi, N. Farm Biosecurity Measures and Interventions with an Impact on Bacterial Biofilms. Agriculture 2022, 12, 1251. [Google Scholar] [CrossRef]
  26. Reshi, A.A.; Husain, I.; Bhat, S.; Rehman, M.U.; Razak, R.; Bilal, S.; Mir, M.R. Bovine mastitis as an evolving disease and its impact on the dairy industry. Int. J. Curr. Res. Rev. 2015, 7, 48. [Google Scholar]
  27. Ruegg, P.L. A 100-Year Review: Mastitis detection, management, and prevention. J. Dairy Sci. 2017, 100, 10381–10397. [Google Scholar] [CrossRef] [Green Version]
  28. Dinsmore, R.P. Biosecurity for mammary diseases in dairy cattle. Vet. Clin. Food Anim. Pract. 2002, 18, 115–131. [Google Scholar] [CrossRef]
  29. Laanen, M.; Maes, D.; Hendriksen, C.; Gelaude, P.; De Vliegher, S.; Rosseel, Y.; Dewulf, J. Pig, cattle and poultry farmers with a known interest in research have comparable perspectives on disease prevention and on-farm biosecurity. Prev. Vet. Med. 2014, 115, 1–9. [Google Scholar] [CrossRef]
  30. Damiaans, B.; Renault, V.; Sarrazin, S.; Berge, A.C.; Pardon, B.; Saegerman, C.; Dewulf, J. A risk-based scoring system to quantify biosecurity in cattle production. Prev. Vet. Med. 2020, 179, 104992. [Google Scholar] [CrossRef]
  31. Rissala, A. Quantitative Assessment of Biosecurity on Finnish Dairy Cattle Farms by Using Biocheck. UGent™ Tool; Eesti Maaülikool: Tartu, Estonia, 2022. [Google Scholar]
  32. Kjosevski, M.; Kondratenko, V.; Angjelovski, B.; Janevski, A.; Simovikj, M.; Djadjovski, I.; Dodovski, A.; Krstevski, K. Biosecurity Assessment and Critical Improvement Points of Dairy Cattle Farms; University of Cyril and Methodius: Skopje, Macedonia, 2022. [Google Scholar]
  33. Oliveira, V.H.; Anneberg, I.; Voss, H.; Sørensen, J.T.; Thomsen, P.T. Attitudes of Danish dairy farmers towards biosecurity. Livest. Sci. 2018, 214, 153–160. [Google Scholar] [CrossRef]
  34. Sarrazin, S.; Cay, A.B.; Laureyns, J.; Dewulf, J. A survey on biosecurity and management practices in selected Belgian cattle farms. Prev. Vet. Med. 2014, 117, 129–139. [Google Scholar] [CrossRef] [PubMed]
  35. Brennan, M.L.; Wright, N.; Wapenaar, W.; Jarratt, S.; Hobson-West, P.; Richens, I.F.; Kaler, J.; Buchanan, H.; Huxley, J.N.; O’Connor, H.M. Exploring attitudes and beliefs towards implementing cattle disease prevention and control measures: A qualitative study with dairy farmers in Great Britain. Animals 2016, 6, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Sayers, R.; Sayers, G.; Mee, J.F.; Good, M.; Bermingham, M.L.; Grant, J.; Dillon, P. Implementing biosecurity measures on dairy farms in Ireland. Vet. J. 2013, 197, 259–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Talbot, B.G.; Lacasse, P. Progress in the development of mastitis vaccines. Livest. Prod. Sci. 2005, 98, 101–113. [Google Scholar] [CrossRef]
  38. Stanković, B.; Hristov, S.; Zlatanović, Z.; Bojkovski, J.; Maksimović, N. Sustainibility ahd Efficiency of Dairy Farms Biosecurity Plans. Agro-Knowl. J. 2016, 16, 437–453. [Google Scholar] [CrossRef] [Green Version]
  39. Landers, T.F.; Cohen, B.; Wittum, T.E.; Larson, E.L. A review of antibiotic use in food animals: Perspective, policy, and potential. Public Health Rep. 2012, 127, 4–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Anđelković, J.; Radonjić, V. Usage of intramammary antimicrobial veterinary medicinal products in the republic of Serbia from 2011 to 2014. Serb. J. Exp. Clin. Res. 2017, 18, 27–31. [Google Scholar] [CrossRef] [Green Version]
  41. Van Boeckel, T.P.; Brower, C.; Gilbert, M.; Grenfell, B.T.; Levin, S.A.; Robinson, T.P.; Teillant, A.; Laxminarayan, R. Global trends in antimicrobial use in food animals. Proc. Natl. Acad. Sci. USA 2015, 112, 5649–5654. [Google Scholar] [CrossRef] [Green Version]
  42. Tiseo, K.; Huber, L.; Gilbert, M.; Robinson, T.P.; Van Boeckel, T.P. Global trends in antimicrobial use in food animals from 2017 to 2030. Antibiotics 2020, 9, 918. [Google Scholar] [CrossRef]
  43. Ruegg, P. Management of mastitis on organic and conventional dairy farms. J. Anim. Sci. 2009, 87, 43–55. [Google Scholar] [CrossRef]
  44. Pyörälä, S. Treatment of mastitis during lactation. Ir. Vet. J. 2009, 62, 1–5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Vidović, J.; Stojanović, D.; Cagnardi, P.; Kladar, N.; Horvat, O.; Ćirković, I.; Bijelić, K.; Stojanac, N.; Kovačević, Z. Farm Animal Veterinarians’ Knowledge and Attitudes toward Antimicrobial Resistance and Antimicrobial Use in the Republic of Serbia. Antibiotics 2022, 11, 64. [Google Scholar] [CrossRef] [PubMed]
  46. Vakanjac, S.; Pavlović, V.; Magaš, V.; Pavlović, M.; Đurić, M.; Maletić, M.; Nedić, S.; Sočo, I. Investigations of efficacy of intramammary applied antimicrobials and glucocorticosteroides in the treatment of subclinical and clinical mastitis in cows. Vet. Glas. 2013, 67, 15–27. [Google Scholar] [CrossRef]
  47. Radinović, M.; Davidov, I.; Kovačević, Z.; Stojanović, D.; Galfi, A.; Erdeljan, M. Osnovni principi terapije mastitisa krava. Ветеринарски Журнал Републике Српске 2019, 19, 105–109. [Google Scholar] [CrossRef]
  48. Abdi, R.D.; Gillespie, B.E.; Ivey, S.; Pighetti, G.M.; Almeida, R.A.; Kerro Dego, O. Antimicrobial resistance of major bacterial pathogens from dairy cows with high somatic cell count and clinical mastitis. Animals 2021, 11, 131. [Google Scholar] [CrossRef]
  49. Kabelitz, T.; Aubry, E.; van Vorst, K.; Amon, T.; Fulde, M. The role of Streptococcus spp. in bovine mastitis. Microorganisms 2021, 9, 1497. [Google Scholar] [CrossRef]
  50. Cagnardi, P.; Grilli, G.; Villa, R.; Di Cesare, F.; Piccirillo, A. Antimicrobials in farm animals: Impact on the environment and consequent antimicrobial resistance dissemination. Int. J. Health Anim. Sci. Food Saf. 2018, 5, 22. [Google Scholar]
  51. Virto, M.; Santamarina-García, G.; Amores, G.; Hernández, I. Antibiotics in dairy production: Where is the Problem? Dairy 2022, 3, 541–564. [Google Scholar] [CrossRef]
  52. Kovačević, Z.; Tomanić, D.; Čabarkapa, I.; Šarić, L.; Stanojević, J.; Bijelić, K.; Galić, I.; Ružić, Z.; Erdeljan, M.; Kladar, N. Chemical Composition, Antimicrobial Activity, and Withdrawal Period of Essential Oil-Based Pharmaceutical Formulation in Bovine Mastitis Treatment. Int. J. Environ. Res. Public Health 2022, 19, 16643. [Google Scholar] [CrossRef]
  53. Liang, S.; Qi, Y.; Yu, H.; Sun, W.; Raza, S.H.A.; Alkhorayef, N.; Alkhalil, S.S.; Salama, E.E.A.; Zhang, L. Bacteriophage Therapy as an Application for Bacterial Infection in China. Antibiotics 2023, 12, 417. [Google Scholar] [CrossRef]
  54. Svircev, A.; Roach, D.; Castle, A. Framing the future with bacteriophages in agriculture. Viruses 2018, 10, 218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Fanelli, A.; Mantegazza, L.; Hendrickx, S.; Capua, I. Thermostable Vaccines in Veterinary Medicine: State of the Art and Opportunities to Be Seized. Vaccines 2022, 10, 245. [Google Scholar] [CrossRef] [PubMed]
  56. Forner, M.; Cañas-Arranz, R.; Defaus, S.; De León, P.; Rodríguez-Pulido, M.; Ganges, L.; Blanco, E.; Sobrino, F.; Andreu, D. Peptide-based vaccines: Foot-and-mouth disease virus, a paradigm in animal health. Vaccines 2021, 9, 477. [Google Scholar] [CrossRef]
  57. Zhylkaidar, A.; Oryntaev, K.; Altenov, A.; Kylpybai, E.; Chayxmet, E. Prevention of Bovine Mastitis through Vaccination. Arch. Razi Inst. 2021, 76, 1381. [Google Scholar] [PubMed]
  58. Milićević, V.; Veljović, L.; Maksimović-Zorić, J.; Žutić, J.; Kureljušić, B.; Radosavljević, V.; Jezdimirović, N.; Radojičić, S. Some adverse events following immunization in veterinary medicine. Arh. Vet. Med. 2018, 11, 67–77. [Google Scholar] [CrossRef]
  59. Vakanjac, S.; Maletić, M.; Pavlović, M.; Obrenović, S.; Bojkovski, J.; Magaš, V. Značaj vakcinacije u prevenciji stafilokoknih mastitisa krava. Vet. Žurnal Repub. Srp. 2017, 17, 170–181. [Google Scholar] [CrossRef] [Green Version]
  60. Giraudo, J.A.; Calzolari, A.; Rampone, H.; Rampone, A.; Giraudo, A.T.; Bogni, C.; Larriestra, A.; Nagel, R. Field trials of a vaccine against bovine mastitis. 1. Evaluation in heifers. J. Dairy Sci. 1997, 80, 845–853. [Google Scholar] [CrossRef]
  61. Pereira, U.; Oliveira, D.; Mesquita, L.; Costa, G.; Pereira, L. Efficacy of Staphylococcus aureus vaccines for bovine mastitis: A systematic review. Vet. Microbiol. 2011, 148, 117–124. [Google Scholar] [CrossRef]
  62. Collado, R.; Montbrau, C.; Sitjà, M.; Prenafeta, A. Study of the efficacy of a Streptococcus uberis mastitis vaccine against an experimental intramammary infection with a heterologous strain in dairy cows. J. Dairy Sci. 2018, 101, 10290–10302. [Google Scholar] [CrossRef] [Green Version]
  63. Piepers, S.; Prenafeta, A.; Verbeke, J.; De Visscher, A.; March, R.; De Vliegher, S. Immune response after an experimental intramammary challenge with killed Staphylococcus aureus in cows and heifers vaccinated and not vaccinated with Startvac, a polyvalent mastitis vaccine. J. Dairy Sci. 2017, 100, 769–782. [Google Scholar] [CrossRef] [Green Version]
  64. Landin, H.; Mörk, M.J.; Larsson, M.; Waller, K.P. Vaccination against Staphylococcus aureus mastitis in two Swedish dairy herds. Acta Vet. Scand. 2015, 57, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Tashakkori, N.; Khoramian, B.; Farhoodi Moghadam, M.; Heidarpour, M.; Mashayekhi, K.; Farzaneh, N. Evaluating the effectiveness of two bovine mastitis vaccines and their influences on oxidant and antioxidant capacities of milk. Trop. Anim. Health Prod. 2020, 52, 1493–1501. [Google Scholar] [CrossRef] [PubMed]
  66. Sharun, K.; Dhama, K.; Tiwari, R.; Gugjoo, M.B.; Iqbal Yatoo, M.; Patel, S.K.; Pathak, M.; Karthik, K.; Khurana, S.K.; Singh, R. Advances in therapeutic and managemental approaches of bovine mastitis: A comprehensive review. Vet. Q. 2021, 41, 107–136. [Google Scholar] [CrossRef] [PubMed]
  67. Weiermayer, P.; Frass, M.; Peinbauer, T.; Ellinger, L.; De Beukelaer, E. Evidence-Based Human Homeopathy and Veterinary Homeopathy. Comment on Bergh et al. A Systematic Review of Complementary and Alternative Veterinary Medicine:“Miscellaneous Therapies”. Animals 2022, 12, 2097. [Google Scholar] [CrossRef] [PubMed]
  68. Jafary, F.; Motamedi, S.; Karimi, I. Veterinary nanomedicine: Pros and cons. Vet. Med. Sci. 2023, 9, 494–506. [Google Scholar] [CrossRef]
  69. Sapino, S.; Chindamo, G.; Chirio, D.; Morel, S.; Peira, E.; Vercelli, C.; Gallarate, M. Nanocarriers in Veterinary Medicine: A Challenge for Improving Osteosarcoma Conventional Treatments. Nanomaterials 2022, 12, 4501. [Google Scholar] [CrossRef]
  70. El-Sayed, A.; Kamel, M. Advanced applications of nanotechnology in veterinary medicine. Environ. Sci. Pollut. Res. 2020, 27, 19073–19086. [Google Scholar] [CrossRef]
  71. Osama, E.; El-Sheikh, S.M.; Khairy, M.H.; Galal, A.A. Nanoparticles and their potential applications in veterinary medicine. J. Adv. Vet. Res. 2020, 10, 268–273. [Google Scholar]
  72. Kalińska, A.; Jaworski, S.; Wierzbicki, M.; Gołębiewski, M. Silver and copper nanoparticles—An alternative in future mastitis treatment and prevention? Int. J. Mol. Sci. 2019, 20, 1672. [Google Scholar] [CrossRef] [Green Version]
  73. Algharib, S.A.; Dawood, A.; Xie, S. Nanoparticles for treatment of bovine Staphylococcus aureus mastitis. Drug Deliv. 2020, 27, 292–308. [Google Scholar] [CrossRef] [Green Version]
  74. Cardozo, V.F.; Lancheros, C.A.; Narciso, A.M.; Valereto, E.C.; Kobayashi, R.K.; Seabra, A.B.; Nakazato, G. Evaluation of antibacterial activity of nitric oxide-releasing polymeric particles against Staphylococcus aureus and Escherichia coli from bovine mastitis. Int. J. Pharm. 2014, 473, 20–29. [Google Scholar] [CrossRef] [PubMed]
  75. Lange, A.; Grzenia, A.; Wierzbicki, M.; Strojny-Cieslak, B.; Kalińska, A.; Gołębiewski, M.; Radzikowski, D.; Sawosz, E.; Jaworski, S. Silver and copper nanoparticles inhibit biofilm formation by mastitis pathogens. Animals 2021, 11, 1884. [Google Scholar] [CrossRef] [PubMed]
  76. Hozyen, H.; Ibrahim, E.; Khairy, E.; El-Dek, S. Enhanced antibacterial activity of capped zinc oxide nanoparticles: A step towards the control of clinical bovine mastitis. Vet. World 2019, 12, 1225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Rivera Aguayo, P.; Bruna Larenas, T.; Alarcón Godoy, C.; Cayupe Rivas, B.; González-Casanova, J.; Rojas-Gómez, D.; Caro Fuentes, N. Antimicrobial and antibiofilm capacity of chitosan nanoparticles against wild type strain of Pseudomonas sp. isolated from milk of cows diagnosed with bovine mastitis. Antibiotics 2020, 9, 551. [Google Scholar] [CrossRef] [PubMed]
  78. Orellano, M.S.; Bohl, L.P.; Breser, M.L.; Isaac, P.; Falcone, R.D.; Porporatto, C. A comparative study of antimicrobial activity of differently-synthesized chitosan nanoparticles against bovine mastitis pathogens. Soft Matter 2021, 17, 694–703. [Google Scholar] [CrossRef]
  79. Alghuthaymi, M.A.; Hassan, A.A.; Kalia, A.; Sayed El Ahl, R.M.; El Hamaky, A.A.; Oleksak, P.; Kuca, K.; Abd-Elsalam, K.A. Antifungal nano-therapy in veterinary medicine: Current status and future prospects. J. Fungi 2021, 7, 494. [Google Scholar] [CrossRef] [PubMed]
  80. Porter, J.; Anderson, J.; Carter, L.; Donjacour, E.; Paros, M. In vitro evaluation of a novel bacteriophage cocktail as a preventative for bovine coliform mastitis. J. Dairy Sci. 2016, 99, 2053–2062. [Google Scholar] [CrossRef]
  81. Titze, I.; Lehnherr, T.; Lehnherr, H.; Krömker, V. Efficacy of bacteriophages against Staphylococcus aureus isolates from bovine mastitis. Pharmaceuticals 2020, 13, 35. [Google Scholar] [CrossRef] [Green Version]
  82. Nilsson, A.S. Phage therapy—Constraints and possibilities. Upsala J. Med. Sci. 2014, 119, 192–198. [Google Scholar] [CrossRef]
  83. Basdew, I.; Laing, M. Mini-Review: Biological control of bovine mastitis using bacteriophage therapy. Sci. Against Microb. Pathog. Commun. Curr. Res. Technol. Adv. 2011, 1, 386–393. [Google Scholar]
  84. Baker, S.J.; Payne, D.J.; Rappuoli, R.; De Gregorio, E. Technologies to address antimicrobial resistance. Proc. Natl. Acad. Sci. USA 2018, 115, 12887–12895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Makumi, A.; Mhone, A.L.; Odaba, J.; Guantai, L.; Svitek, N. Phages for Africa: The potential benefit and challenges of phage therapy for the livestock sector in sub-Saharan Africa. Antibiotics 2021, 10, 1085. [Google Scholar] [CrossRef] [PubMed]
  86. Ferriol-González, C.; Domingo-Calap, P. Phage therapy in livestock and companion animals. Antibiotics 2021, 10, 559. [Google Scholar] [CrossRef] [PubMed]
  87. Amiri Fahliyani, S.; Beheshti-Maal, K.; Ghandehari, F. Novel lytic bacteriophages of Klebsiella oxytoca ABG-IAUF-1 as the potential agents for mastitis phage therapy. FEMS Microbiol. Lett. 2018, 365, fny223. [Google Scholar] [CrossRef] [PubMed]
  88. Iwano, H.; Inoue, Y.; Takasago, T.; Kobayashi, H.; Furusawa, T.; Taniguchi, K.; Fujiki, J.; Yokota, H.; Usui, M.; Tanji, Y. Bacteriophage ΦSA012 has a broad host range against Staphylococcus aureus and effective lytic capacity in a mouse mastitis model. Biology 2018, 7, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Breyne, K.; Honaker, R.W.; Hobbs, Z.; Richter, M.; Żaczek, M.; Spangler, T.; Steenbrugge, J.; Lu, R.; Kinkhabwala, A.; Marchon, B. Efficacy and safety of a bovine-associated Staphylococcus aureus phage cocktail in a murine model of mastitis. Front. Microbiol. 2017, 8, 2348. [Google Scholar] [CrossRef] [PubMed]
  90. Li, L.; Zhang, Z. Isolation and characterization of a virulent bacteriophage SPW specific for Staphylococcus aureus isolated from bovine mastitis of lactating dairy cattle. Mol. Biol. Rep. 2014, 41, 5829–5838. [Google Scholar] [CrossRef]
  91. Shan, Y.; Yang, N.; Teng, D.; Wang, X.; Mao, R.; Hao, Y.; Ma, X.; Fan, H.; Wang, J. Recombinant of the staphylococcal bacteriophage lysin CHAPk and its elimination against Streptococcus agalactiae biofilms. Microorganisms 2020, 8, 216. [Google Scholar] [CrossRef] [Green Version]
  92. Guo, M.; Gao, Y.; Xue, Y.; Liu, Y.; Zeng, X.; Cheng, Y.; Ma, J.; Wang, H.; Sun, J.; Wang, Z. Bacteriophage cocktails protect dairy cows against mastitis caused by drug resistant Escherichia coli infection. Front. Cell. Infect. Microbiol. 2021, 11, 690377. [Google Scholar] [CrossRef]
  93. Vander Elst, N.; Linden, S.B.; Lavigne, R.; Meyer, E.; Briers, Y.; Nelson, D.C. Characterization of the bacteriophage-derived endolysins PlySs2 and PlySs9 with in vitro lytic activity against bovine mastitis Streptococcus uberis. Antibiotics 2020, 9, 621. [Google Scholar] [CrossRef]
  94. Zhao, W.; Shi, Y.; Liu, G.; Yang, J.; Yi, B.; Liu, Y.; Kastelic, J.P.; Han, B.; Gao, J. Bacteriophage has beneficial effects in a murine model of Klebsiella pneumoniae mastitis. J. Dairy Sci. 2021, 104, 3474–3484. [Google Scholar] [CrossRef] [PubMed]
  95. Oechslin, F. Resistance development to bacteriophages occurring during bacteriophage therapy. Viruses 2018, 10, 351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Nehme, R.; Andrés, S.; Pereira, R.B.; Ben Jemaa, M.; Bouhallab, S.; Ceciliani, F.; López, S.; Rahali, F.Z.; Ksouri, R.; Pereira, D.M. Essential oils in livestock: From health to food quality. Antioxidants 2021, 10, 330. [Google Scholar] [CrossRef] [PubMed]
  97. Groot, M.J.; Berendsen, B.J.; Cleton, N.B. The Next Step to Further Decrease Veterinary Antibiotic Applications: Phytogenic Alternatives and Effective Monitoring; the Dutch Approach. Front. Vet. Sci. 2021, 8, 709750. [Google Scholar] [CrossRef]
  98. Guo, W.; Qiu, M.; Pu, Z.; Long, N.; Yang, M.; Ren, K.; Ning, R.; Zhang, S.; Peng, F.; Sun, F. Geraniol-a potential alternative to antibiotics for bovine mastitis treatment without disturbing the host microbial community or causing drug residues and resistance. Front. Cell. Infect. Microbiol. 2023, 13, 131. [Google Scholar] [CrossRef]
  99. Pașca, C.; Mărghitaș, L.A.; Dezmirean, D.S.; Matei, I.A.; Bonta, V.; Pașca, I.; Chirilă, F.; Cîmpean, A.; Fiț, N.I. Efficacy of natural formulations in bovine mastitis pathology: Alternative solution to antibiotic treatment. J. Vet. Res. 2020, 64, 523–529. [Google Scholar] [CrossRef]
  100. Yap, P.S.X.; Lim, S.H.E.; Hu, C.P.; Yiap, B.C. Combination of essential oils and antibiotics reduce antibiotic resistance in plasmid-conferred multidrug resistant bacteria. Phytomedicine 2013, 20, 710–713. [Google Scholar] [CrossRef]
  101. Vaou, N.; Stavropoulou, E.; Voidarou, C.; Tsigalou, C.; Bezirtzoglou, E. Towards advances in medicinal plant antimicrobial activity: A review study on challenges and future perspectives. Microorganisms 2021, 9, 2041. [Google Scholar] [CrossRef]
  102. Amber, R.; Adnan, M.; Tariq, A.; Khan, S.N.; Mussarat, S.; Hashem, A.; Al-Huqail, A.A.; Al-Arjani, A.-B.F.; Abd_Allah, E.F. Antibacterial activity of selected medicinal plants of northwest Pakistan traditionally used against mastitis in livestock. Saudi J. Biol. Sci. 2018, 25, 154–161. [Google Scholar] [CrossRef]
  103. Pașca, C.; Mărghitaș, L.; Dezmirean, D.; Bobiș, O.; Bonta, V.; Chirilă, F.; Matei, I.; Fiț, N. Medicinal plants based products tested on pathogens isolated from mastitis milk. Molecules 2017, 22, 1473. [Google Scholar] [CrossRef] [Green Version]
  104. Vitale, S.; Colanero, S.; Placidi, M.; Di Emidio, G.; Tatone, C.; Amicarelli, F.; D’Alessandro, A.M. Phytochemistry and Biological Activity of Medicinal Plants in Wound Healing: An Overview of Current Research. Molecules 2022, 27, 3566. [Google Scholar] [CrossRef] [PubMed]
  105. Kovačević, Z.; Radinović, M.; Čabarkapa, I.; Kladar, N.; Božin, B. Natural agents against bovine mastitis pathogens. Antibiotics 2021, 10, 205. [Google Scholar] [CrossRef] [PubMed]
  106. Cheesman, M.J.; Ilanko, A.; Blonk, B.; Cock, I.E. Developing new antimicrobial therapies: Are synergistic combinations of plant extracts/compounds with conventional antibiotics the solution? Pharmacogn. Rev. 2017, 11, 57. [Google Scholar] [PubMed] [Green Version]
  107. Ebani, V.V.; Mancianti, F. Use of essential oils in veterinary medicine to combat bacterial and fungal infections. Vet. Sci. 2020, 7, 193. [Google Scholar] [CrossRef]
  108. Tomanić, D.Z.; Stanojević, J.B.; Galić, I.M.; Ružić, Z.N.; Kukurić, T.B.; Tešin, N.B.; Prpa, B.P.; Kovačević, Z.R. Review of trends in essential oils as alternatives to antibiotics in bovine mastitis treatment. Zb. Matice Srp. Za Prir. Nauk. 2022, 142, 47–60. [Google Scholar] [CrossRef]
  109. Rhimi, W.; Mohammed, M.A.; Zarea, A.A.K.; Greco, G.; Tempesta, M.; Otranto, D.; Cafarchia, C. Antifungal, antioxidant and antibiofilm activities of essential oils of Cymbopogon spp. Antibiotics 2022, 11, 829. [Google Scholar] [CrossRef]
  110. Tomanić, D.; Božin, B.; Čabarkapa, I.; Kladar, N.; Radinović, M.; Maletić, M.; Kovačević, Z. Chemical Composition, Antioxidant and Antibacterial Activity of Two Different Essential Oils Against Mastitis Associated Pathogens. Acta Vet. 2022, 72, 45–58. [Google Scholar] [CrossRef]
  111. Barreiros, Y.; de Meneses, A.C.; Alves, J.L.F.; Mumbach, G.D.; Ferreira, F.A.; Machado, R.A.F.; Bolzan, A.; de Araujo, P.H.H. Xanthan gum-based film-forming suspension containing essential oils: Production and in vitro antimicrobial activity evaluation against mastitis-causing microorganisms. LWT 2022, 153, 112470. [Google Scholar] [CrossRef]
  112. Kovačević, Z.; Kladar, N.; Čabarkapa, I.; Radinović, M.; Maletić, M.; Erdeljan, M.; Božin, B. New perspective of Origanum vulgare L. and Satureja montana L. essential oils as bovine mastitis treatment alternatives. Antibiotics 2021, 10, 1460. [Google Scholar] [CrossRef]
  113. Tomanić, D.; Božin, B.; Kladar, N.; Stanojević, J.; Čabarkapa, I.; Stilinović, N.; Apić, J.; Božić, D.D.; Kovačević, Z. Environmental Bovine Mastitis Pathogens: Prevalence, Antimicrobial Susceptibility, and Sensitivity to Thymus vulgaris L., Thymus serpyllum L., and Origanum vulgare L. Essential Oils. Antibiotics 2022, 11, 1077. [Google Scholar] [CrossRef]
  114. Abboud, M.; El Rammouz, R.; Jammal, B.; Sleiman, M. In vitro and in vivo antimicrobial activity of two essential oils Thymus vulgaris and Lavandula angustifolia against bovine Staphylococcus and Streptococcus mastitis pathogen. Middle East J. Agric. Res. 2015, 4, 975–983. [Google Scholar]
  115. Corona-Gómez, L.; Hernández-Andrade, L.; Mendoza-Elvira, S.; Suazo, F.M.; Ricardo-González, D.I.; Quintanar-Guerrero, D. In vitro antimicrobial effect of essential tea tree oil (Melaleuca alternifolia), thymol, and carvacrol on microorganisms isolated from cases of bovine clinical mastitis. Int. J. Vet. Sci. Med. 2022, 10, 72–79. [Google Scholar] [CrossRef] [PubMed]
  116. Fratini, F.; Casella, S.; Leonardi, M.; Pisseri, F.; Ebani, V.V.; Pistelli, L.; Pistelli, L. Antibacterial activity of essential oils, their blends and mixtures of their main constituents against some strains supporting livestock mastitis. Fitoterapia 2014, 96, 1–7. [Google Scholar] [CrossRef]
  117. Zhu, H.; Du, M.; Fox, L.; Zhu, M.-J. Bactericidal effects of Cinnamon cassia oil against bovine mastitis bacterial pathogens. Food Control 2016, 66, 291–299. [Google Scholar] [CrossRef]
  118. Abd El-Aziz, N.K.; Ammar, A.M.; El-Naenaeey, E.-s.Y.; El Damaty, H.M.; Elazazy, A.A.; Hefny, A.A.; Shaker, A.; Eldesoukey, I.E. Antimicrobial and antibiofilm potentials of cinnamon oil and silver nanoparticles against Streptococcus agalactiae isolated from bovine mastitis: New avenues for countering resistance. BMC Vet. Res. 2021, 17, 1–14. [Google Scholar] [CrossRef] [PubMed]
  119. Cerioli, M.F.; Moliva, M.V.; Cariddi, L.N.; Reinoso, E.B. Effect of the essential oil of Minthostachys verticillata (Griseb.) epling and limonene on biofilm production in pathogens causing bovine mastitis. Front. Vet. Sci. 2018, 5, 146. [Google Scholar] [CrossRef] [Green Version]
  120. Budri, P.E.; Silva, N.C.; Bonsaglia, E.C.; Júnior, A.F.; Júnior, J.A.; Doyama, J.T.; Gonçalves, J.L.; Santos, M.V.d.; Fitzgerald-Hughes, D.; Rall, V.L. Effect of essential oils of Syzygium aromaticum and Cinnamomum zeylanicum and their major components on biofilm production in Staphylococcus aureus strains isolated from milk of cows with mastitis. J. Dairy Sci. 2015, 98, 5899–5904. [Google Scholar] [CrossRef]
  121. Pinedo, P.; Karreman, H.; Bothe, H.; Velez, J.; Risco, C. Efficacy of a botanical preparation for the intramammary treatment of clinical mastitis on an organic dairy farm. Can. Vet. J. 2013, 54, 479. [Google Scholar] [CrossRef]
  122. Mullen, K.; Anderson, K.; Washburn, S. Effect of 2 herbal intramammary products on milk quantity and quality compared with conventional and no dry cow therapy. J. Dairy Sci. 2014, 97, 3509–3522. [Google Scholar] [CrossRef] [Green Version]
  123. Hase, P.; Digraskar, S.; Ravikanth, K.; Dandale, M.; Maini, S. Management of subclinical mastitis with mastilep gel and herbal spray (AV/AMS/15). Int. J. Pharm. Pharm. 2013, 4, 64–67. [Google Scholar]
  124. Yang, W.-T.; Ke, C.-Y.; Wu, W.-T.; Lee, R.-P.; Tseng, Y.-H. Effective treatment of bovine mastitis with intramammary infusion of Angelica dahurica and Rheum officinale extracts. Evid.-Based Complement. Altern. Med. 2019, 2019, 7242705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Cho, B.-W.; Cha, C.-N.; Lee, S.-M.; Kim, M.-J.; Park, J.-Y.; Yoo, C.-Y.; Son, S.-E.; Kim, S.; Lee, H.-J. Therapeutic effect of oregano essential oil on subclinical bovine mastitis caused by Staphylococcus aureus and Escherichia coli. Korean J. Vet. Res. 2015, 55, 253–257. [Google Scholar] [CrossRef] [Green Version]
  126. Tomanić, D.; Kladar, N.; Radinović, M.; Stančić, I.; Erdeljan, M.; Stanojević, J.; Galić, I.; Bijelić, K.; Kovačević, Z. Intramammary Ethno-Veterinary Formulation in Bovine Mastitis Treatment for Optimization of Antibiotic Use. Pathogens 2023, 12, 259. [Google Scholar] [CrossRef] [PubMed]
  127. Nisha, A. Antibiotic residues-a global health hazard. Vet. World 2008, 1, 375. [Google Scholar] [CrossRef]
  128. McPhee, C.; Anderson, K.; Yeatts, J.; Mason, S.; Barlow, B.; Baynes, R. Hot topic: Milk and plasma disposition of thymol following intramammary administration of a phytoceutical mastitis treatment. J. Dairy Sci. 2011, 94, 1738–1743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Kovačević, Z.; Mihajlović, J.; Mugoša, S.; Horvat, O.; Tomanić, D.; Kladar, N.; Samardžija, M. Pharmacoeconomic Analysis of the Different Therapeutic Approaches in Control of Bovine Mastitis: Phytotherapy and Antimicrobial Treatment. Antibiotics 2023, 12, 11. [Google Scholar] [CrossRef] [PubMed]
  130. Doehring, C.; Sundrum, A. Efficacy of homeopathy in livestock according to peer-reviewed publications from 1981 to 2014. Vet. Rec. 2016, 179, 628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Keller, D.; Sundrum, A. Comparative effectiveness of individualised homeopathy and antibiotics in the treatment of bovine clinical mastitis: Randomised controlled trial. Vet. Rec. 2018, 182, 407. [Google Scholar] [CrossRef] [Green Version]
  132. Løken, T. Alternative therapy of animals–homeopathy and other alternative methods of therapy. Acta Vet. Scand. 2002, 43, 1–4. [Google Scholar] [CrossRef] [Green Version]
  133. Ebert, F.; Staufenbiel, R.; Simons, J.; Pieper, L. Randomized, blinded, controlled clinical trial shows no benefit of homeopathic mastitis treatment in dairy cows. J. Dairy Sci. 2017, 100, 4857–4867. [Google Scholar] [CrossRef]
  134. Williamson, J.; Lacy-Hulbert, S. Lack of efficacy of homeopathic therapy against post-calving clinical mastitis in dairy herds in the Waikato region of New Zealand. New Zealand Vet. J. 2014, 62, 8–14. [Google Scholar] [CrossRef] [PubMed]
  135. Mimoune, N.; Saidi, R.; Benadjel, O.; Khelef, D.; Kaidi, R. Alternative treatment of bovine mastitis. Vet. Stanica 2021, 52, 639–649. [Google Scholar] [CrossRef]
  136. Varshney, J.; Naresh, R. Comparative efficacy of homeopathic and allopathic systems of medicine in the management of clinical mastitis of Indian dairy cows. Homeopathy 2005, 94, 81–85. [Google Scholar] [CrossRef] [PubMed]
  137. Zeise, J.; Fritz, J. Use and efficacy of homeopathy in prevention and treatment of bovine mastitis. Open Agric. 2019, 4, 203–212. [Google Scholar] [CrossRef]
  138. Vidal Amaral, J.R.; Jucá Ramos, R.T.; Almeida Araújo, F.; Bentes Kato, R.; Figueira Aburjaile, F.; de Castro Soares, S.; Góes-Neto, A.; Matiuzzi da Costa, M.; Azevedo, V.; Brenig, B. Bacteriocin Producing Streptococcus agalactiae Strains Isolated from Bovine Mastitis in Brazil. Microorganisms 2022, 10, 588. [Google Scholar] [CrossRef] [PubMed]
  139. Carson, D.A.; Barkema, H.W.; Naushad, S.; De Buck, J. Bacteriocins of non-aureus staphylococci isolated from bovine milk. Appl. Environ. Microbiol. 2017, 83, e01015–e01017. [Google Scholar] [CrossRef] [Green Version]
  140. Diez-Gonzalez, F. Applications of bacteriocins in livestock. Curr. Issues Intest. Microbiol. 2007, 8, 15. [Google Scholar] [PubMed]
  141. Bennett, S.; Ben Said, L.; Lacasse, P.; Malouin, F.; Fliss, I. Susceptibility to nisin, bactofencin, pediocin and reuterin of multidrug resistant Staphylococcus aureus, Streptococcus dysgalactiae and Streptococcus uberis causing bovine mastitis. Antibiotics 2021, 10, 1418. [Google Scholar] [CrossRef]
  142. Newstead, L.L.; Varjonen, K.; Nuttall, T.; Paterson, G.K. Staphylococcal-produced bacteriocins and antimicrobial peptides: Their potential as alternative treatments for Staphylococcus aureus infections. Antibiotics 2020, 9, 40. [Google Scholar] [CrossRef] [Green Version]
  143. Castelani, L.; Arcaro, J.; Braga, J.; Bosso, A.; Moura, Q.; Esposito, F.; Sauter, I.; Cortez, M.; Lincopan, N. Activity of nisin, lipid bilayer fragments and cationic nisin-lipid nanoparticles against multidrug-resistant Staphylococcus spp. isolated from bovine mastitis. J. Dairy Sci. 2019, 102, 678–683. [Google Scholar] [CrossRef] [Green Version]
  144. Hernández-González, J.C.; Martínez-Tapia, A.; Lazcano-Hernández, G.; García-Pérez, B.E.; Castrejón-Jiménez, N.S. Bacteriocins from lactic acid bacteria. A powerful alternative as antimicrobials, probiotics, and immunomodulators in veterinary medicine. Animals 2021, 11, 979. [Google Scholar] [CrossRef] [PubMed]
  145. Coelho, M.L.V.; dos Santos Nascimento, J.; Fagundes, P.C.; Madureira, D.J.; de Oliveira, S.S.; de Paiva Brito, M.A.V.; de Freire Bastos, M.d.C. Activity of staphylococcal bacteriocins against Staphylococcus aureus and Streptococcus agalactiae involved in bovine mastitis. Res. Microbiol. 2007, 158, 625–630. [Google Scholar] [CrossRef] [PubMed]
  146. Bruni, N.; Capucchio, M.T.; Biasibetti, E.; Pessione, E.; Cirrincione, S.; Giraudo, L.; Corona, A.; Dosio, F. Antimicrobial activity of lactoferrin-related peptides and applications in human and veterinary medicine. Molecules 2016, 21, 752. [Google Scholar] [CrossRef]
  147. Lacasse, P.; Lauzon, K.; Diarra, M.; Petitclerc, D. Utilization of lactoferrin to fight antibiotic-resistant mammary gland pathogens. J. Anim. Sci. 2008, 86, 66–71. [Google Scholar] [CrossRef]
  148. Diarra, M.; Petitclerc, D.; Lacasse, P. Effect of lactoferrin in combination with penicillin on the morphology and the physiology of Staphylococcus aureus isolated from bovine mastitis. J. Dairy Sci. 2002, 85, 1141–1149. [Google Scholar] [CrossRef] [PubMed]
  149. Diarra, M.S.; Petitclerc, D.; Deschênes, E.; Lessard, N.; Grondin, G.; Talbot, B.G.; Lacasse, P. Lactoferrin against Staphylococcus aureus Mastitis: Lactoferrin alone or in combination with penicillin G on bovine polymorphonuclear function and mammary epithelial cells colonisation by Staphylococcus aureus. Vet. Immunol. Immunopathol. 2003, 95, 33–42. [Google Scholar] [CrossRef]
  150. Kutila, T.; Pyörälä, S.; Saloniemi, H.; Kaartinen, L. Antibacterial effect of bovine lactoferrin against udder pathogens. Acta Vet. Scand. 2003, 44, 1–8. [Google Scholar] [CrossRef]
  151. Shimazaki, K.-I.; Kawai, K. Advances in lactoferrin research concerning bovine mastitis. Biochem. Cell Biol. 2017, 95, 69–75. [Google Scholar] [CrossRef] [Green Version]
  152. Sahoo, N.; Kumar, P.; Bhusan, B.; Bhattacharya, T.; Dayal, S.; Sahoo, M. Lysozyme in livestock: A guide to selection for disease resistance: A review. J. Anim. Sci. Adv 2012, 2, 347–360. [Google Scholar]
  153. Chaneton, L.; Sáez, J.P.; Bussmann, L.E. Antimicrobial activity of bovine β-lactoglobulin against mastitis-causing bacteria. J. Dairy Sci. 2011, 94, 138–145. [Google Scholar] [CrossRef] [Green Version]
  154. Petitclerc, D.; Lauzon, K.; Cochu, A.; Ster, C.; Diarra, M.; Lacasse, P. Efficacy of a lactoferrin-penicillin combination to treat β-lactam-resistant Staphylococcus aureus mastitis. J. Dairy Sci. 2007, 90, 2778–2787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Hegazi, A.; Abdou, A.M.; Allah, F. Antimicrobial activity of propolis on the bacterial causes of mastitis. Life Sci. J. 2014, 11, 572–576. [Google Scholar]
  156. Tatli Seven, P.; Seven, I.; Gul Baykalir, B.; Iflazoglu Mutlu, S.; Salem, A.Z. Nanotechnology and nano-propolis in animal production and health: An overview. Ital. J. Anim. Sci. 2018, 17, 921–930. [Google Scholar] [CrossRef] [Green Version]
  157. Masek, T.; Perin, N.; Racané, L.; Cindric, M.; Paljetak, H.C.; Peric, M.; Matijasic, M.; Verbanac, D.; Radic, B.; Suran, J. Chemical Composition, antioxidant and antibacterial activity of different extracts of poplar type propolis. Croat. Chem. Acta 2018, 91, 81–89. [Google Scholar] [CrossRef]
  158. Santos, L.M.; Fonseca, M.S.; Sokolonski, A.R.; Deegan, K.R.; Araújo, R.P.; Umsza-Guez, M.A.; Barbosa, J.D.; Portela, R.D.; Machado, B.A. Propolis: Types, composition, biological activities, and veterinary product patent prospecting. J. Sci. Food Agric. 2020, 100, 1369–1382. [Google Scholar] [CrossRef]
  159. Fiordalisi, S.A.; Honorato, L.A.; Loiko, M.R.; Avancini, C.A.; Veleirinho, M.B.; Machado Filho, L.C.; Kuhnen, S. The effects of Brazilian propolis on etiological agents of mastitis and the viability of bovine mammary gland explants. J. Dairy Sci. 2016, 99, 2308–2318. [Google Scholar] [CrossRef] [Green Version]
  160. Bačić, C.; Macesic, N.; Radin, L.; Aladrovic, J.; Matanovic, K.; Masek, T.; Brozic, D.; Benic, M.; Radic, B.; Bacic, I. SuranJ: Intramammary propolis formulation for subclinical mastitis prevention and treatment in dairy cows. J. Dairy Vet. Anim. Res. 2016, 3, 159. [Google Scholar] [CrossRef] [Green Version]
  161. Bačić, G.; Mačešić, N.; Radin, L.; Aladrović, J.; Matanović, K.; Mašek, T.; Brozić, D.; Benić, M.; Radić, B.; Bačić, I. Intramammary Propolis Formulation for Prevention and Treatment of Mastitis in Dairy Ruminants (RC. 2.2. 08–0003). J. Anim. Res. 2016, 6, 227–229. [Google Scholar] [CrossRef]
  162. Suran, J.; Matanovic, K.; Bozic, F.; Martinec, B.S.; Aladrovic, J.; Bacic, G.; Macesic, N.; Masek, T.; Brozic, D.; Benic, M. Intramammary propolis formulation for prevention and treatment of mastitis in dairy ruminants. Proc. J. Vet. Pharmacol. Ther. 2015, 67. [Google Scholar]
  163. Kober, A.H.; Saha, S.; Islam, M.A.; Rajoka, M.S.R.; Fukuyama, K.; Aso, H.; Villena, J.; Kitazawa, H. Immunomodulatory Effects of Probiotics: A Novel Preventive Approach for the Control of Bovine Mastitis. Microorganisms 2022, 10, 2255. [Google Scholar] [CrossRef]
  164. Espeche, M.C.; Pellegrino, M.; Frola, I.; Larriestra, A.; Bogni, C.; Nader-Macías, M.F. Lactic acid bacteria from raw milk as potentially beneficial strains to prevent bovine mastitis. Anaerobe 2012, 18, 103–109. [Google Scholar] [CrossRef] [PubMed]
  165. Đuričić, D.; Sukalić, T.; Marković, F.; Kočila, P.; Žura Žaja, I.; Menčik, S.; Dobranić, T.; Benić, M.; Samardžija, M. Effects of dietary vibroactivated clinoptilolite supplementation on the intramammary microbiological findings in dairy cows. Animals 2020, 10, 202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Benić, M.; Maćešić, N.; Cvetnić, L.; Habrun, B.; Cvetnić, Ž.; Turk, R.; Đuričić, D.; Lojkić, M.; Dobranić, V.; Valpotić, H. Bovine mastitis: A persistent and evolving problem requiring novel approaches for its control-a review. Vet. Arh. 2018, 88, 535–557. [Google Scholar] [CrossRef]
  167. Gruet, P.; Maincent, P.; Berthelot, X.; Kaltsatos, V. Bovine mastitis and intramammary drug delivery: Review and perspectives. Adv. Drug Deliv. Rev. 2001, 50, 245–259. [Google Scholar] [CrossRef]
  168. Alluwaimi, A.M. The cytokines of bovine mammary gland: Prospects for diagnosis and therapy. Res. Vet. Sci. 2004, 77, 211–222. [Google Scholar] [CrossRef]
  169. Oviedo-Boyso, J.; Valdez-Alarcón, J.J.; Cajero-Juárez, M.; Ochoa-Zarzosa, A.; López-Meza, J.E.; Bravo-Patino, A.; Baizabal-Aguirre, V.M. Innate immune response of bovine mammary gland to pathogenic bacteria responsible for mastitis. J. Infect. 2007, 54, 399–409. [Google Scholar] [CrossRef]
  170. Daley, M.; Williams, T.; Coyle, P.; Furda, G.; Dougherty, R.; Hayes, P. Prevention and treatment of Staphylococcus aureus infections with recombinant cytokines. Cytokine 1993, 5, 276–284. [Google Scholar] [CrossRef]
  171. Enginler, S.Ö.; Sabuncu, A.; Kahraman, B.B.; Koçak, Ö.; Yıldar, E.; Güzel, Ö. Comparison of intramammary ozone administration doses in dairy cows with clinical mastitis. Acta Sci. Vet. 2015, 43, 1–7. [Google Scholar]
  172. Ogata, A.; Nagahata, H. Intramammary application of ozone therapy to acute clinical mastitis in dairy cows. J. Vet. Med. Sci. 2000, 62, 681–686. [Google Scholar] [CrossRef] [Green Version]
  173. Kwon, H.-J.; Liu, J.; Jo, S.-N.; Song, K.-H.; Kim, D.-H.; Jun, M.-H.; Cho, S.-W.; Kim, M.-C.; Yoon, H.-I. Therapeutic effect of ozone gas on bovine mastitis. J. Vet. Clin. 2005, 22, 314–317. [Google Scholar]
  174. Chacón, O.; Forno, N.; Lapierre, L.; Muñoz, R.; Fresno, M.; San Martín, B. Effect of Aloe barbadensis Miller (Aloe vera) associated with beta-lactam antibiotics on the occurrence of resistance in strains of Staphylococcus aureus and Streptococcus uberis. Eur. J. Integr. Med. 2019, 32, 100996. [Google Scholar] [CrossRef]
  175. Forno-Bell, N.; Munoz, M.A.; Chacón, O.; Pachá, P.; Iragüen, D.; Cornejo, J.; San Martín, B. Efficacy Prediction of Four Pharmaceutical Formulations for Intramammary Administration Containing Aloe vera (L.) Burm. f. Combined With Ceftiofur or Cloxacillin in Lactating Cows as an Alternative Therapy to Treat Mastitis Caused by Staphylococcus aureus. Front. Vet. Sci. 2021, 8, 572568. [Google Scholar] [CrossRef]
  176. Šubová, E.; Sasáková, N.; Zigo, F.; Mindžáková, I.; Vargová, M.; Kachnič, J.; Laktičová, K.V. Amendment of livestock manure with natural zeolite-clinoptilolite and its effect on decomposition processes during composting. Agriculture 2021, 11, 980. [Google Scholar] [CrossRef]
  177. Jäger, T.; Mokos, A.; Prasianakis, N.I.; Leyer, S. first_page settings Order Article Reprints Open AccessArticle Pore-Level Multiphase Simulations of Realistic Distillation Membranes for Water Desalination. Membranes 2022, 12, 1112. [Google Scholar] [CrossRef]
  178. Đuričić, D.; Benić, M.; Maćešić, N.; Valpotić, H.; Turk, R.; Dobranić, V.; Cvetnić, L.; Gračner, D.; Vince, S.; Grizelj, J. Dietary zeolite clinoptilolite supplementation influences chemical composition of milk and udder health in dairy cows. Vet. Stanica 2017, 48, 257–265. [Google Scholar]
  179. Maity, S.; Rubić, I.; Kuleš, J.; Horvatić, A.; Đuričić, D.; Samardžija, M.; Ljubić, B.B.; Turk, R.; Gračner, D.; Maćešić, N. Integrated metabolomics and proteomics dynamics of serum samples reveals dietary zeolite clinoptilolite supplementation restores energy balance in high yielding dairy cows. Metabolites 2021, 11, 842. [Google Scholar] [CrossRef]
  180. Alic Ural, D. Efficacy of clinoptilolite supplementation on milk yield and somatic cell count. Rev. MVZ Córdoba 2014, 19, 4242–4248. [Google Scholar] [CrossRef] [Green Version]
  181. Li, Y.; Zhu, Y.; Chu, B.; Liu, N.; Chen, S.; Wang, J. Lactobacillus rhamnosus GR-1 prevents Escherichia coli-induced apoptosis through PINK1/Parkin-mediated mitophagy in bovine mastitis. Front. Immunol. 2021, 12, 715098. [Google Scholar] [CrossRef] [PubMed]
  182. Nader-Macías, M.E.F.; Bogni, C.; Sesma, F.J.M.; Espeche, M.C.; Pellegrino, M.; Saavedra, L.; Frola, I. Alternative Approaches for the Prevention of Bovine Mastitis. Probiotics, Bioactive Compounds And vaccines. In Bioactives Compounds; Nova Science Publishing: New York, NY, USA, 2011; pp. 1–34. [Google Scholar]
  183. Pellegrino, M.S.; Frola, I.D.; Natanael, B.; Gobelli, D.; Nader-Macias, M.E.; Bogni, C.I. In vitro characterization of lactic acid bacteria isolated from bovine milk as potential probiotic strains to prevent bovine mastitis. Probiotics Antimicrob. Proteins 2019, 11, 74–84. [Google Scholar] [CrossRef]
  184. Klostermann, K.; Crispie, F.; Flynn, J.; Ross, R.P.; Hill, C.; Meaney, W. Intramammary infusion of a live culture of Lactococcus lactis for treatment of bovine mastitis: Comparison with antibiotic treatment in field trials. J. Dairy Res. 2008, 75, 365–373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tomanić, D.; Samardžija, M.; Kovačević, Z. Alternatives to Antimicrobial Treatment in Bovine Mastitis Therapy: A Review. Antibiotics 2023, 12, 683. https://doi.org/10.3390/antibiotics12040683

AMA Style

Tomanić D, Samardžija M, Kovačević Z. Alternatives to Antimicrobial Treatment in Bovine Mastitis Therapy: A Review. Antibiotics. 2023; 12(4):683. https://doi.org/10.3390/antibiotics12040683

Chicago/Turabian Style

Tomanić, Dragana, Marko Samardžija, and Zorana Kovačević. 2023. "Alternatives to Antimicrobial Treatment in Bovine Mastitis Therapy: A Review" Antibiotics 12, no. 4: 683. https://doi.org/10.3390/antibiotics12040683

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop