Next Article in Journal
The Relaxin-3 Receptor, RXFP3, Is a Modulator of Aging-Related Disease
Next Article in Special Issue
A Lupin (Lupinus angustifolius) Protein Hydrolysate Exerts Anxiolytic-Like Effects in Western Diet-Fed ApoE−/− Mice
Previous Article in Journal
Phenotype of Mrps5-Associated Phylogenetic Polymorphisms Is Intimately Linked to Mitoribosomal Misreading
Previous Article in Special Issue
Enzymes-Assisted Extraction of Plants for Sustainable and Functional Applications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Natural Products for the Prevention and Treatment of Oral Mucositis—A Review

by
Ana Sofia Ferreira
1,†,
Catarina Macedo
1,†,
Ana Margarida Silva
1,
Cristina Delerue-Matos
1,
Paulo Costa
2,3 and
Francisca Rodrigues
1,*
1
REQUIMTE/LAQV—Instituto Superior de Engenharia do Porto, Rua Dr. António Bernardino de Almeida 431, 4249-015 Porto, Portugal
2
UCIBIO—Applied Molecular Biosciences Unit, MedTech-Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
3
Associate Laboratory i4HB—Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(8), 4385; https://doi.org/10.3390/ijms23084385
Submission received: 14 March 2022 / Revised: 11 April 2022 / Accepted: 14 April 2022 / Published: 15 April 2022

Abstract

:
Cancer, a major world public health problem, is associated with chemotherapy treatments whose administration leads to secondary concerns, such as oral mucositis (OM). The OM disorder is characterized by the presence of ulcers in the oral mucosa that cause pain, bleeding, and difficulty in ingesting fluids and solids, or speaking. Bioactive compounds from natural sources have arisen as an effective approach for OM. This review aims to summarize the new potential application of different natural products in the prevention and treatment of OM in comparison to conventional ones, also providing a deep insight into the most recent clinical studies. Natural products, such as Aloe vera, Glycyrrhiza glabra, Camellia sinensis, Calendula officinalis, or honeybee crops, constitute examples of sources of bioactive compounds with pharmacological interest due to their well-reported activities (e.g., antimicrobial, antiviral, anti-inflammatory, analgesic, or wound healing). These activities are associated with the bioactive compounds present in their matrix (such as flavonoids), which are associated with in vivo biological activities and minimal or absent toxicity. Finally, encapsulation has arisen as a future opportunity to preserve the chemical stability and the drug bioa vailability of bioactive compounds and, most importantly, to improve the buccal retention period and the therapeutic effects.

1. Introduction

Cancer is currently a major public health problem all over the world. In 2020, almost 19.3 millions new cases were diagnosed worldwide [1]. Treatment of malignancies with cytotoxic chemotherapy (CT), radiation (RT), or a combination of the two is becoming more effective, as it is associated with short- and long-term adverse effects, including mucositis [2,3]. This secondary reaction may occur in any area of the gastrointestinal tract’s mucosal layer, from the mouth to the anus, with the oral cavity being the most prevalent location. The cytotoxicity is caused by a variety of mechanisms, including inhibition of DNA replication and repair, cell-cycle arrest, DNA damage, and cell death [4]. However, the precise and complex molecular pathways underlying the oral epithelial damage are not completely known [5,6].
Oral mucositis (OM) is a painful inflammatory and frequently ulcerative disorder of the oral mucosa that severely reduces the patient’s quality of life [3,7,8]. OM occurs in 20 to 40% of the patients submitted to conventional CT, 80% of patients on high-dose CT, 75 to 100% of patients receiving hematopoietic cell transplants, and practically all patients with head and neck squamous carcinoma (HNSC) undergoing RT [4,5,9,10,11]. Common symptoms of OM include pain, bleeding, ulcers, and difficulty ingesting fluids or solids and speaking, as well as severe complications, such as secondary infections and significant weight loss, which may compromise the treatment of the primary disease and its outcome [2,12,13]. In addition, OM may result in the need for enteral or parenteral nutrition [14,15] and systemic analgesics [16,17,18], thus increasing hospitalizations [13,19], the use of resources and higher costs [19,20], and, in some cases, the risk of sepsis [8,21]. However, when mucositis progresses, topical analgesics become less effective and systemic opioids may be required [22,23,24]. Different strategies have been used to attempt the prevention or amelioration of this condition, and some clinical trials were effective [8,16,17,25]. For example, cryotherapy [11,26] and keratinocyte growth factor [11,27] demonstrated some benefits in preventing mucositis. Zinc [28,29] and vitamin E [28,30,31] were effective in reducing the severity of OM, but Aloe vera [32], amifostine [4,33], glutamine [28,30,34], honey [32,35,36,37,38], photobiomodulation (PBM) therapy [39,40,41], and antibiotics [21] demonstrated lower evidence of benefits. The studies reviewed were evaluated in patients with different types of cancer who underwent different treatment approaches.
While there are a growing number of innovative anticancer agents, few therapeutic alternatives for the prevention or treatment of oral mucositis have been reported. Most important, the scarce alternatives that have been successfully achieved are still unsatisfactory [6,16]. Therefore, the search for alternative compounds obtained from natural sources could be an option and a challenge for this research field. Natural compounds, in contrast to synthetic ones, are often thought to have fewer side effects, are easy to access, and present beneficial bioactive properties (e.g., anti-inflammatory, antioxidant, and antimicrobial properties), making them interesting solutions as promising therapeutics. Aside from the protective results of natural products against toxicity induced by radiation or antineoplastic drugs, one of the most promising preventive measures in patients during therapy may be the employment of natural products. The aim of this review is to provide an overview of the use of natural compounds for the prevention and eventual treatment of OM in cancer patients and their potential applications in drug delivery systems to overcome the specific limitations of the oral cavity environment.

2. Oral Mucositis

As previously stated, mucositis is an inflammatory response condition of the oral mucous membrane that is frequently observed in malignant neoplastic patients undergoing CT, RT, or both. This condition develops due to interactions among an oral tissue injury, the oral cavity environment, bone marrow suppression, and innate predisposing factors in the patient [18,42,43]. The symptoms of OM, such as oral mucosal atrophy, swelling, erythema and subsequent pain, bleeding, ulceration, difficulty in feeding and even swallowing saliva, or a combination thereof, may be diverse [2,11,44]. Difficulties with eating reduce the nutritional intake, resulting in a decline in the patient’s nutritional status. This can also seriously affect their speech due to an uncomfortably dry mouth and a decrease or increase in salivation [11,16,44]. OM may also be aggravated by injuries induced by sharpened teeth, bruxism, food, and microorganisms [44,45]. Naturally, additional ulcers provide an easy access point for microorganisms, including bacteria, fungi, and viruses, to enter the bloodstream because of the loss of mucosal integrity, culminating in systemic infections that may cause the treatment for fighting the primary disease to be discontinued or even threaten the patient’s survival. Moreover, the dysphagia, xerostomia, and changes in taste caused by OM can increase the systemic symptoms, such as lethargy and anorexia, as well as psychological issues. Consequently, OM is associated with increased resource needs and potentially major economic impacts—depending on its severity—due to the more frequent and prolonged hospitalizations for support and nutritional care and analgesic treatments.
Three tools are available for assessing the severity of OM. The most extensively used is the World Health Organization’s Oral Mucositis Grading Scale (WHO-OMGS), which incorporates clinical criteria to evaluate the OM lesion and eating capacity [46]. On the other hand, the Common Terminology Criteria for Adverse Events in its fifth revision (CTCAE v5.0) considers the following factors when assessing the impact of OM: pain intensity, ability to eat, and need for intervention [46]. Finally, Radiation Therapy Oncology Group (RTOG) defines the severity of RT-induced OM using a four-point scale [46]. OM is classified according to these three criteria, as summarized in Table 1.

2.1. Physiopathology of OM

In the last decades, substantial evolution has taken place in the understanding of the complex mechanism behind the development of mucositis [6]. A five-phase model that begins with an (i) initiation involving cell injury, (ii) elevation of inflammatory cytokines, a (iii) primary damage response, and (iv) signaling and amplification of the inflammatory cascade, followed by (v) ulceration and mucosal repair through epithelial proliferation, has been reported by different authors [2,3,16,47]. Thus, OM is characterized by a cascade of events that occur simultaneously and are mechanistically related (Figure 1). Therefore, each factor that drives each phase may constitute a possible therapeutic target [16].
The mucositis initiation phase—initiation—corresponds to the injury of oral mucosal cells caused by CT and/or RT. This phase begins instantaneously as the antineoplastic treatment is being administered [5,6,48,49]. The second phase—upregulation with messenger generation—involves the cytotoxic effect, resulting in the generation of reactive oxygen and nitrogen species (ROS and RNS, respectively) and DNA damage, leading to basal and suprabasal epithelial cell death [2,3,6]. Particularly, when DNA strands breaks, the apoptotic process is activated, with p53 and nuclear factor κB (NF- κB) playing major roles [50,51]. At this point, inflammatory cytokines, chemokines, and adhesion molecules are generated when NF- κB, the key mediator of pro-inflammatory gene expression, is activated, which is clinically manifested as mucosal damage. The release of pro-inflammatory cytokines, such as tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), is mediated through transcription factor activation, and this promotes connective tissue and endothelial damage, limiting the tissue oxygenation and stimulating epithelial basal cell death [2,18,50,51,52,53]. The third phase—signaling and amplification—is the consequence of tissue damage, apoptosis, enzyme activation, and vascular permeability, which amplify the molecules of the innate immune response as pro-inflammatory cytokines in a positive feedback mechanism, leading to more tissue damage [18,54]. In the fourth phase—ulceration—clinical signs of mucositis become visible, as the integrity of the mucosa and submucosa is disrupted, causing pain control to be required [3,5,16]. In neutropenic patients, the immune cells cannot respond properly, and the ulcerative lesions allow several microorganisms to penetrate into the connective tissue, triggering the production of more pro-inflammatory cytokines and increasing the tissue damage [50,51]. Bacteremia and sepsis are mostly caused by herpes simplex virus, Candida albicans, or other fungal genera, such as Aspergillus [10]. Healing usually occurs naturally after the cancer treatment is ceased, and it is marked by epithelial proliferation, migration, and differentiation promoted by the extracellular matrix [2,3,49]. The oral mucosa recovers, but the patient remains at risk for recurrent episodes due to residual angiogenesis [16,18,49,55].
CT patients often experience acute symptoms 3–5 days following its administration, with ulcerative lesions appearing a few days later and resolving within 2 weeks [3,44,51]. On the other hand, RT mucositis is a chronic condition that lasts up to 7 weeks. The radiation doses range from 2 to 70 Gy per day and cause ulcerations that remain for 3–4 weeks after the treatment is ceased [9,11,18]. The lack of taste develops because the oral mucosa is exposed to radiation after few weeks, compromising nutrition and psychological status, while the recovery begins 6–8 weeks after the completion of the treatment [5,9].

2.2. Risk Factors

The risk factors of OM can be classified as patient-related, tumor-related, and treatment-related variables, as summarized in Table 2 [16,45].
In the patient-related factors, gender has been linked to mucositis, since women are associated with a higher risk, which could be due to dosimetric considerations [12,25,58,62]. However, other studies reported the absence of evidence that gender and OM are correlated [16,45,58,63]. Although age is frequently reported as a mucositis risk factor, there are few consistent reports that link younger and older patients and mucositis severity [45,58]. Likewise, the effect of body mass index (BMI) on mucositis risk is inconsistent, with data suggesting that a low BMI and a BMI higher than 25 are related with a superior risk, as body composition can affect drug metabolism, as can smoking and poor oral hygiene [18,45,58]. Genetic variants, previous treatment, and comorbidities (such as renal dysfunction and diabetes mellitus) have been indicated as possible factors for chronic OM associated with RT [16,61].
In what concerns the tumor’s nature, its location, size, and stadium may also influence the grade of OM [45]. For instance, in HNSC patients, the standard protocol includes RT with a specific area and prescription dose, which influences the exposure to radiation and the subsequent mucosal damage [16,45]. However, in recent years, there was an increased investment in intraoral medical devices that enable the minimization of excessive irradiation of normal tissues [64].
Although the risk factors of OM are not completely understood, the characteristics of anticancer therapeutics (mechanism of action, dose, planning, and number of cycles) are closely associated with the prevalence and severity of the lesions, as their effects accumulate [12,18,45]. It is well known that female patients using methotrexate and melphalan have a greater chance of developing this local inflammatory condition [16].
Along with investigating intrinsic patient characteristics, such as pre-existing medical conditions, altered oral dynamics, and general health, age, oral health (hygiene prior to treatment), nutritional status, and liver and kidney function are critical, as they are parameters that a medical team must consider [2,18]. Aside from that, it is necessary to emphasize that OM is frequently documented only in its advanced phases owing to the requirements for clinical therapy and assistance [13,14,19,65]. Therefore, the search for new active ingredients that could be used in the prevention (and even treatment) of OM is of utmost importance.

2.3. Prevention and Management of OM

OM management strategies include either preventive or symptom control strategies [8,18,23,25]. The primary key measure in preventing OM is the preservation of tissue during RT treatment planning and the use of RT procedures that conserve the uninvolved oral mucosal surface [17,66]. Some strategies are addressed in the evidence-based guidelines developed by the Multinational Association of Supportive Care in Cancer and the International Society of Oral Oncology (MASCC/ISOO), which present three categories: a recommendation, a suggestion, and a situation where no guideline is possible [54,63]. These guidelines can be adjusted at any time to compensate for possible restraints in the clinic and patient choices [63]. Table 3 summarizes the recommended or suggested strategies for most of the groups of cancer patients.
Proper oral health and hygiene are essential for mitigating the risk and severity of OM [11,60]. Before initiating CT or RT, all potential causes of mucosal irritation should be removed, as they may worsen and prolong the development of oral mucositis [60]. Teeth with sharp surfaces must be restored, orthodontics and protheses should be removed, and the maintenance of a stable oral microbiome is also an important aspect. The presence of a balanced nutrition is another variable that may help in the relief of discomfort from mucositis [9,11,60]. Alcohol, smoking, and foods that are crunchy, acidic, spicy, or sweetened should be limited or eliminated [81].
As previously stated, OM can make the ingestion process a challenge, as it is normally unpleasant and, in extreme cases, impossible due to painful symptoms; therefore, a liquid diet is the only solution [2,18]. Therefore, soft and liquid diets may be necessary, and, in the case of patients that cannot tolerate a liquid diet, the solution is parenteral nutrition [17]. The patients’ complaints can be reduced with the use of specific mouthwashes with topical analgesics, anesthetics, antibiotics, and steroids [67,74,80,82], as topical analgesics and anesthetics are intended to relieve localized pain [23].
According to the MASCC/ISOO guidelines (Table 3), a benzydamine mouthwash may be useful due to its anti-inflammatory properties, which inhibit the production of TNF-α and IL-1β [46,53,63,74]. However, the use of saline, sodium bicarbonate, and antimicrobial (e.g., chlorhexidine 0.12%) rinses can ameliorate the symptoms of moderate mucositis [53,83]. In clinical practice, topical analgesics (e.g., morphine, benzocaine, and menthol) are applied to provide temporary relief in some patients, but their concentrations are not well established [22,84].
Currently, palifermin was the only agent that has been approved by the European Medical Agency (EMA) and the American Food and Drug Administration (FDA) for the prevention of OM in HSCT patients receiving CT and RT. However, on 1 April 2016, the European Commission withdrew the marketing authorization for this drug in the European Union (EU). The withdrawal was at the request of the marketing authorisation holder, which notified the European Commission of its decision to permanently discontinue the marketing of the product for commercial reasons. It has also been tested in HNSC patients in terms of its reduction of the state of pathogenic severity [10]. The MASSC/ISOO guidelines also indicate cryotherapy and photobiomodulation (PBM) protocols for more advanced phases. In particular, cryotherapy has been reported to reduce the symptoms of oral mucositis in patients undergoing CT as a result of its vasoconstriction, decrease in the blood flow, and reduction of the local distribution of the chemotherapeutic agent (e.g., fluorouracil (5-FU) and melphalan) [23,71]. Thirty minutes of ice chips used prior to the administration CT are the recommended and tolerable period [71,73].
PBM is another method employed to stabilize and inhibit the development of OM [39,85]. It has anti-inflammatory effects, diminishes the pain, and improves the healing rate of the basal wound. The energy applied to the specific area must be adapted according to the patient’s lesion. To relieve the most common complaints, PBM can be used both prophylactically and therapeutically, that is, it can be used before and after an antineoplastic treatment [40,86,87]. Mucositis may also be treated with supplementary vitamins and minerals. For instance, vitamin E, a potent antioxidant, may reduce the grade of mucositis by preventing the damage caused by ROS [2]. A blood test performed in severe OM patients demonstrated a lack of some vitamins (such as vitamins E, A, and D), which inhibited the pro-inflammatory pathways [34]. Different studies also showed that oral zinc supplements may be applied as a prophylactic treatment [18,29,30].
Therefore, for most of the strategies recommended or suggested in Table 3, the research in the literature displays minimal evidence or even contradictory results, thus invalidating the definitions of the guidelines [7,8,54,63]. Consequently, the search for new active ingredients with potential therapeutic effects for preventing or treating OM is a challenge. Natural compounds, the majority of which are rich in polyphenols, are an option that should be explored.

3. Natural Compounds and Their Properties for Preventing/Treating OM

Currently, the protocols and therapeutical agents available from the different authorities have the purpose of ameliorating the OM grade, as mentioned in the previous section, but no treatments with reasonable results have been established [42,53,88]. Aside from that, many of these compounds have been associated with adverse effects and high costs [8,19]. Thus, natural products, such as honey, Aloe vera, curcumin, or propolis, are of huge interest for the nutraceutical and pharmaceutical industries, as they are easily accessible and allow more cost-effective treatments with minimal or no toxicity when compared to conventional strategies [89,90]. Their richness in bioactive compounds with anti-inflammatory, antioxidant, antiseptic, analgesic, and wound-healing properties that may interfere with many cellular signaling pathways could play an important role in the progression of OM and the activity of carcinogenic cells (e.g., HNSC) [10].

3.1. Bee Products

Honey is a natural product generated by bees and has been used since ancient times in traditional medicine. The huge diversity of studies has shown the multiplicity of beneficial applications of honey based on its antioxidant, anti-inflammatory, antibacterial, antiviral, antifungal, antitumoral, antimutagenic, and wound-healing properties [32,35,37,38,77]. The composition of honey is difficult to exactly define, as the components and relative amounts are conditioned by the flora of the geographical area from which honeybees collect pollen [91]. In a general way, honey is a heterogeneous mixture of water, nectar sugars, and glandular secretions produced by honeybees that contain proteins, vitamins, and enzymes [92]. One of the enzymes present is glucose oxidase, which, when in contact with body tissue, may stimulate the production of hydrogen peroxide, which acts as a messenger and promote wound healing and rapid epithelization at low concentrations by stimulating the proliferation of fibroblasts and epithelial cells [92,93]. It is also suggested that matrix metalloproteases of connective tissue and neutrophil serine proteases may be activated by hydrogen peroxide [94]. Furthermore, the levulose and fructose present in honey may improve local nutrition and promote epithelialization [93,94]. Honey also has immunomodulatory effects, as it influences the activation of macrophages and the proliferation of B-lymphocytes and T-lymphocytes [95], in addition to decreasing the inflammatory process by inhibiting cyclooxygenase pathway and reducing prostaglandin synthesis [96]. The beneficial effects of honey may also be due to its moisturizing effect, low pH, and viscosity which inhibit the proliferation of bacteria [35].
Charalambous et al. conducted a randomized, controlled trial to evaluate the potential effect of thyme honey rinses on HNSC patients [35]. In this study that involved 72 participants, a solution of 20 mL of thyme honey diluted in 100 mL of purified water was given to the patients to gargle in the oral cavity three times per day (15 min before and after the RT session and 6 h later) for 7 weeks, starting from the first day of the fourth week of RT. The results showed a significant improvement (p < 0.001) in the patients’ quality of life, leading to fewer symptoms and maintenance of the body weight (p = 0.001) when compared to saline rinses [35]. Honey mouthwash also proved to be effective in a randomized, single-blind controlled trial that enrolled 53 patients [97]. The honey solution (honey-to-water ratio of 1:20) at 37 °C was gargled and kept in the mouth before and after each meal and before sleeping for 30 s by the treatment group, while the patients in the control group received routine care, such as ingestion of fluconazole capsules, nursing care, and mouth hygiene training [97]. According to the authors, the solution reduced or eliminated weight loss, leading to some weight gain and preventing and reducing the severity of OM in the acute myeloid leukemia patients receiving CT (p < 0.001 at the fourth week of treatment) [97]. In another randomized, controlled trial with a parallel design involving 150 children, Sener et al. treated 25 OM patients with honey (with vitamin E as the most effective compound) for 21 days, applying the amount of 1–1.5 g of honey per weight (kg) of the child twice per day (every 12 h) [31]. Honey was found to be more effective in the management of OM (p < 0.05) when compared to chlorhexidine, a wide-spectrum antifungal and bactericidal antiseptic solution that is frequently used in oral care [31]. Motallbnejad et al. also conducted a randomized single-blind (examiner-blind) clinical trial to evaluate the effect of pure honey on radiation-induced mucositis in a total of 40 patients with head and neck cancer receiving RT [95]. Twenty patients were instructed to rinse and gradually swallow 20 mL of pure honey 15 min before radiation, then again at intervals of 15 min and six hours after radiation, while the control group was advised to rinse with 20 mL of saline before and after radiation. This procedure was repeated weekly from the beginning of the treatment until the end of the RT. The honey-receiving patients exhibited a significant reduction in OM (p < 0.001) when compared to the control group [95]. In a unicenter randomized, controlled clinical human study involving 82 patients with head and neck cancer treated with RT over 4–6 weeks, the treatment group was instructed to take 20 mL of Ziziphus honey 15 min before and after the radiation and before sleeping at night, while the control group repeated the process using 20 mL of 0.9% saline [92]. The results showed that the proportion of mucositis (Grades 3 and 4) was lower in the honey-treated group (p = 0.016 and p = 0.032 for Grades 3 and 4 of mucositis, respectively) than in the control group at the end of 6 weeks of RT [92]. In 2010, Khanal et al. conducted a single-blinded, randomized, controlled clinical trial over 6 weeks on 40 oral carcinoma patients receiving RT [91]. Radiation was given once per day for 5 days a week, and the application was performed 15 min before and after radiation and once before going to bed. Honey extracted from beehives of the Western Ghats forests or lignocaine gel 2% (control group) was swished around the oral cavity for 2 min and expectorated. Only one of the 20 patients of the treatment group developed intolerable mucositis (p < 0.0001) compared to 15 of the 20 patients of the lignocaine group [91]. Caffeine, a natural alkaloid with hypoalgesic, antioxidant, and anti-inflammatory effects, has also been screened as a potential ingredient to work against oral mucositis [98,99,100,101]. In a double-blinded randomized clinical trial involving 75 patients (that randomly fell into three treatment groups) presenting OM after CT, the therapeutic effects of coffee plus honey were compared with those of topical steroids that are usually used in the treatment of OM after CT [98]. A syrup-like solution was prepared for each treatment group: 300 g of honey plus 20 g of instant coffee for the honey-plus-coffee group; 300 mg of honey for the honey group; the control group was treated with 20 eight-milligram ampoules of betamethasone solution. All groups were instructed to sip 10 mL of the prescribed product and swallow every 3 h for 1 week. While all treatment regimens decreased the severity of the lesions, the best result was achieved in the honey–coffee group (p < 0.05), followed by the honey-and-steroid groups [98].

3.1.1. Propolis

Propolis is a resinous material produced by bees and is frequently used as natural nutritional supplement [102]. It is composed of a mix of plant buds and exudates, bee enzymes, pollen, and wax, and it has been widely used by different civilizations to treat colds, wounds, and ulcers due to its anesthetic, antimicrobial, anti-inflammatory, antitumor, immunomodulatory, and antioxidant properties [102]. Similarly to honey, the chemical composition of propolis is highly dependent on the diversity of the flora and bee species [103,104]. It is mainly composed of proteins, amino acids, vitamins (A, B1, B2, B3, and B7), minerals, essential oils, phenolic acids, alcohols, fatty acids, and flavonoids [102,105,106,107,108]. Regarding OM, the bioactivity of propolis is mainly associated with flavonoids, as these molecules are capable of sequestering or inhibiting the formation of free radicals, and they promote immunomodulatory, antioxidant, wound-healing, and anti-inflammatory activities [109]. The anti-inflammatory properties are directly related with the inhibition of the synthesis of prostaglandins and promotion of phagocytic activity [110]. In addition, propolis promotes healing effects in epithelial tissues, while the presence of iron and zinc improves the synthesis of collagen [108].
Akhavan-Karbassi et al. conducted a randomized double-blind placebo-controlled trial to evaluate the potential effect of propolis mouthwash on head and neck tumor patients undergoing CT [111]. In the treatment group (n = 20), 5 mL of propolis mouth rinse (30% extract) was administered every 8 h for 7 consecutive days. The solution was swished in the patients’ mouths for 60 s, gargled, and expectorated. In the control group (n = 20), the process was repeated with a placebo mouth rinse. OM, erythema, and eating and drink ability were evaluated. When compared to the control group, the treatment group presented significant improvement in OM, wound healing, and erythema at day 7 (p = 0.006), but no significant differences in eating and drinking ability were observed (p = 0.21). Moreover, 65% of the patients in the propolis group were completely healed by day 7 of the trial [111].

3.1.2. Royal Jelly

Royal jelly is a secretory product of the cephalic glands of nurse bees that serves as the diet of honeybee larvae in their first 2–3 days, while for the queen, it is the specific food for her whole life period [112]. It is widely used in folk and mainstream medicines and as a dietary supplement due to its antioxidant, anti-inflammatory, hypoglycemic, antibiotic, antitumor, antiallergic, antiaging, immunomodulatory, neurotrophic, hypocholesterolemic, hepatoprotective, hypotensive, and blood pressure regulatory activities [112,113,114,115,116,117,118,119,120].
Similarly to the aforementioned bee products, the composition of royal jelly is dependent on the geography and climate [121]. It is a complex substance with a unique combination of sugars (mainly glucose and fructose, as well as traces of sucrose, maltose, trehalose, melibiose, ribose, and erlose), proteins (which represent >50% of the dry weight of royal jelly), amino acids, nucleotides, ascorbic acid, phenols, waxes, fatty acids, steroids, and phospholipids [121]. The impact that royal jelly has on OM may be closely related to its anti-inflammatory and wound-healing activities. However, the active compounds of royal jelly and the mechanisms underlying these activities are still largely unknown. In vitro studies performed on mice revealed that supernatants of royal jelly suspensions added to a mouse peritoneal macrophage culture stimulated with lipopolysaccharides and IFN-γ efficiently suppressed the secretion of pro-inflammatory cytokines TNF-α, IL-6, and IL-1, which was probably due to protein factors such as Major Royal Jelly Protein 3 (MRJP3) [119]. MRJP2, MRJP3, and MRJP7 are thought to be responsible for the wound-healing bioactivity of royal jelly, as they stimulate cell migration and proliferation [122], along with the antioxidant compounds present in royal jelly, which, when taken orally, lowered the levels of 8-hydroxy-2-deoxyguanosine, a marker of oxidative stress in mouse kidney DNA and serum [123].
Suemaru et al. evaluated the effects of royal jelly, honey, and propolis on OM induced with 5-fluorouracil and mild abrasions made on the cheek pouch in hamsters [124]. The bee products were topically administered to the oral mucosa. Royal jelly ointments at 3%, 10%, and 30% improved the recovery from 5-fluorouracil-induced damage in a dose-dependent manner, while the results of ointments of honey at 1%, 10%, and 100% and propolis at 0.3%, 1%, and 3% were not statically different from those of the Vaseline-treated control group [124]. In a more in-depth trial in Golden Syrian hamsters, the influence of royal jelly on 5-fluorouracil-induced OM was assessed using oral mucosal adhesive films containing royal jelly [125]. The 5-fluorouracil was administered through intraperitoneal injections on days 0 and 2, and the left cheek pouches of hamsters (n = 12 per group) were everted and scratched with a small wire brush on days 1 and 2. Royal-jelly-containing sodium alginate–chitosan films (10% or 30%) were applied to the cheek pouches every day from day 3. Royal-jelly-containing films (both 10% and 30%) improved the recovery from 5-fluorouracil-induced OM, which presented lower erythema and absence of ulceration and abscesses on day 8. They also reduced the myelo-peroxidase (MPO) activity and the expression of pro-inflammatory cytokines. The data suggest that these effects were caused by the anti-inflammatory or antioxidative properties of royal jelly [125]. In humans, the effect of royal jelly on OM in patients with different types of malignancies undergoing RT and CT was evaluated by Erdem et al. in a randomized, controlled trial [126]. In this clinical trial that involved 103 patients, all patients received a mouthwash therapy with benzydamine hydrochloride and nystatin rinses. In addition, patients in the experimental group received royal jelly two times per day for a total daily dose of 1 g. Royal jelly was orally swished for 30 s and swallowed. The treatment group showed a mean resolution time of OM that was significantly shorter than that of the control group (OM Grade 1: p = 0.0001; OM Grade 2: p = 0.0001; OM Grade 3: p = 0.05) [126]. In a single-blind clinical trial that involved 13 patients with head and neck cancer receiving CT, 1 g of royal jelly was given three times per day to the treatment group during the RT period [127]. Royal jelly was shown to have a preventive effect on the progression of CT-induced OM from the early phase (p < 0.001) [127].

3.2. Spondias Mombin

The leaves of Spondias mombin, commonly known as the cashew tree, are a rich source of interesting bioactive compounds, with particular emphasis on tannins, saponins, triterpenes, and flavonoids [128]. Traditionally, the leaves have been used to treat inflammatory pathologies, making them a promising source for the development of new therapeutic agents for OM [128]. Gomes et al. assessed the effects of a hydroethanolic extract of S. mombin leaves on 5-fluorouracil-induced OM in Golden Syrian male hamsters [128]. The animals were orally pre-treated with the hydroethanolic extract of S. mombin leaves (50, 100, or 200 mg/kg) for ten days [128]. The treatment with the highest dose of the extract (200 mg/kg) showed the best healing effect, with hamsters displaying reduced oxidative stress and inflammation and no evidence of ulceration. Further analysis showed re-epithelialization, absence of hemorrhage, discrete mononuclear inflammatory infiltration, and lower expression levels of different molecules involved in the modulation of inflammation, such as MMP-2, COX-2, TNF-α, NF-κB p50 NLS, iNOS, and IL-1β, as well as an increase in glutathione (GSH) levels [128]. Although the mechanisms behind these effects remain under investigation, the hydroethanolic extract of S. mombin leaves is rich in potent antioxidant phenolic phytochemicals, such as ellagic acid (12 mg/g) and chlorogenic acid (19.4 mg/g), which could justify these activities [129]. Studies have demonstrated that chlorogenic acid acts on the reduction of COX-2 expression in macrophages, as well as in the inhibition of the production of pro-inflammatory cytokines, such as IL-1β and TNF-α, and of NF-κB activation [129]. On the other hand, chlorogenic acid was proven to promote wound healing in rats [130], while ellagic acid acted by down-regulating MMP-2 expression and inhibiting NF-κB-mediated transcriptional activation [129]. These activities may justify the results achieved in the previously detailed trial.

3.3. Camellia sinensis

Camellia sinensis (green tea) is one of the most popular drinks in the world and is widely known for its antimicrobial, antitumoral, antioxidant, and anti-inflammatory activities [67]. Different compounds with therapeutic effects have been discovered in this plant. The majority of the health-promoting properties are associated with polyphenols [131], which represent almost 30% of the fresh-leaf dry weight, including flavandiols, flavonols, flavonoids, and phenolic acids [132]. However, most of the polyphenols present in leaves of C. sinensis are catechins, namely, (+)-catechin, (−)-epicatechin, (+)-gallocatechin, (−)-epigallocatechin (EGC), (−)-epicatechin gallate, and (−)-epigallocatechin gallate (EGCG) [133]. Catechins are mainly responsible for the ROS scavenging and antioxidant activities of C. sinensis [134,135]. EGCG, in particular, efficiently inhibits the transcription of NF-κB, resulting in a decrease in the expression of different pro-inflammatory genes [136]. The anti-inflammatory effect of catechins may be due to the activation of endothelial nitric oxide synthase (eNOS) [137,138].
The effect of green tea on OM was evaluated in oral cancer patients [67]. For that, a single-blind randomized, controlled trial was made with 63 participants. For 6 months, after the tooth-brushing procedure, the intervention group rinsed the mouth with a solution of 5 g of green tea dissolved in 100 mL of water for 60 s, and the control group rinsed the mouth with 100 mL of tap water for the same period. The results demonstrated an improvement in oral health status and the preservation of the oral mucosa at the end of the follow-up period (6 months), with a higher reduction of the oral health status score in the intervention group than in the control group (p = 0.008) [67]. In another randomized study, the effect of Baxidil Onco® mouthwash (Sanitas Farmaceutici Srl, Tortona, Italy), composed of C. Sinensis leaf extract and palmitoyil hydrolyzed wheat protein, was tested in 60 hematologic patients undergoing hematopoietic stem cell transplantation (HCST) [139]. Twenty mL of Baxidil Onco® was used to rinse the mouths of 28 patients four times per day for at least one minute without swallowing, while the remaining 32 patients were treated with standard prophylactic schedules and served as control. The results demonstrated that the incidence, severity, and duration of OM were significantly reduced (p = 0.022) by the oral rinsing with Baxidil Onco® [139].

3.4. Plantago Major

In traditional Persian medicine, Plantago major was used as a wound-healing herb, as it possesses a wide range of bioactive properties, such as anti-inflammatory, antiulcerogenic, antioxidant, antimicrobial, analgesic, wound-healing, and immunomodulatory effects [140].
Soltani et al. conducted a randomized, double-blind, placebo-controlled clinical trial to assess the effects of P. major syrup as a natural agent against OM for 7 weeks [141]. The participants were HNSC patients who were going to receive RT. The 23 patients of the intervention group received 7.5 cc of P. major syrup three times per day, starting from three days before the start of RT until the end of it, while the placebo group received 7.5 cc of placebo syrup. The P. major syrup was shown to be effective in the reduction of the mucositis and the severity of pain caused by RT (p < 0.001) [141]. A multicenter randomized, controlled trial developed by Cabrera-Jaime et al. evaluated the efficacy of P. major extract vs. chlorhexidine vs. sodium bicarbonate in the treatment of CT-induced OM in solid-tumor cancer patients with grade II–III mucositis [140]. A total of 45 patients were randomized for one of the treatments, consisting of a 5% aqueous solution of sodium bicarbonate together with (i) an additional dose of 5% sodium bicarbonate, (ii) P. major extract, or (iii) 0.12% chlorhexidine. The solutions were applied over 14 days. The differences in healing time and the lower pain levels among the three groups were not statistically significant (p = 0.702) [140].
The properties of P. major leaves are dependent on the different compounds present. The leaves are rich in different bioactive molecules, such as aucubin, a glycoside with anti-toxin activity, and ursolic, oleanolic, and α-linoleic acids, which inhibit COX-2-catalyzed prostaglandin production [142,143,144]. Extracts of P. major leaves have remarkable antioxidant and antiradical capacities due to the presence of baicalein, lutolin, salicylic acid, citric acid, ascorbic acid, apigenin, ferulic acid, benzoic acid, chlorogenic acid, oleanolic acid, and ursolic acids [145,146]. According to different studies, the bioactivity of P. major is due to the decrease in the inflammatory reaction through the modulation of NF-κB, NO, COX-2, and B4 leukotriene (LB4) levels [140].

3.5. Aloe vera

A. vera is a plant that has been used for medical purposes for thousands of years. It is widely employed for the treatment of various medical conditions, such as oral ulcers, psoriasis, skin burns, and frostbite, since it presents analgesic, liver-protection, antifungal, antidiabetic, anti-inflammatory, antiproliferative, anticarcinogenic, antiaging, and immunomodulatory properties [147,148,149]. In addition, it can scavenge free radicals, improve wound oxygenation, promote wound healing, increase collagen formation, and inhibit metalloproteinase and collagenase activity [150,151,152,153,154]. Different studies have shown the potent free-radical and superoxide anion activity of three derivatives from A. vera, namely, isorabaichromone, feruoylaloesin, and p-coumaroylaloesin [150,151]. The beneficial effects, assumed to be exerted in the oral cavity, may also be due to its moisturizing effect, which is provided by the polysaccharide components (principally mannose, glucose, xylose, arabinose, galactose, and rhamnose), which provide and sustain moisture in tissues [155]. One of the sugars present in a higher quantity, mannose-6-phosphate, acted as an active-growth substance and anti-inflammatory agent in in vivo studies on mice [156]. The anti-inflammatory effects of A. vera extracts are attributable to the inhibitory action on the arachidonic acid pathway via COX-2 inhibition [150,157], as well as the reduction of leukocyte adhesion molecules and TNF-α levels [158]. In vitro and animal assays suggest that A. vera promotes wound healing through the reduction of the vasoconstriction and the platelet aggregation at the wound site [152].
An initial assessment of A. vera’s potential in preventing RT-induced OM did not yield promising results in a single-institution, double-blind, prospective, randomized trial that involved 58 head and neck cancer patients [159]. The patients were instructed to take a 20 mL swish (A. vera solution or placebo) and swallow four times daily, beginning on the first day and continuing throughout the course of RT. However, no significant differences were observed between treatments (p = 0.07) [159]. Better results were achieved in other studies. Mansouri et al. evaluated the effect of A. vera on CT-induced OM in patients with acute lymphocytic leukemia and acute myeloid leukemia [160]. In this randomized, controlled clinical trial, 64 patients were divided into an intervention group and a control group. The first group was instructed to wash their mouths with 5 mL of A. vera solution for 2 min three times per day for 14 days. The control group repeated the procedure using mouthwashes that are typically recommended by hematologic centers, including normal saline, nystatin, and chlorhexidine. An evaluation of the patients’ mouths was performed on days 1, 3, 5, 7, and 14. Even though, regarding the intensity of stomatitis and pain, no significant differences were found between the two groups on the first day, a significant difference was observed in this regard on the other days (p < 0.001) [160]. In a similar study, an assessment of the effect of A. vera mouthwash on CT-induced OM was performed in a double-blinded randomized clinical trial on 120 patients, who were divided into three groups [161]. Until 2 weeks after the CT sessions, group 1 received tablets with 10 mg of atorvastatin daily plus a placebo mouthwash, group 2 received placebo tablets and A. vera mouthwash, and group 3 received placebo tablets and placebo mouthwash. The analysis of the results showed that 50% of the placebo patients (group 3) experienced mucositis, while that value decreased to 2.5% in group 2 (p < 0.042), with no significant differences between groups 1 and 3 (p < 0.674) [161]. Likewise, the efficacy of A. vera use for prevention of CT-induced OM was evaluated in a randomized, controlled clinical trial in 26 children with acute lymphoblastic leukemia [162]. Depending on the treatment group, a 70% A. vera solution or a 5% sodium bicarbonate solution was applied twice per day to oral tissues with spongeous sticks. The application started 3 days before the CT therapy. The application of A. vera solution showed to be effective in the prevention and reduction of OM severity (p < 0.001) [162]. A triple-blind randomized and controlled interventional quality-of-life clinical trial on the efficacy of A. vera and a benzydamine mouthwash in the alleviation of RT-induced OM was performed by Sahebjamee et al. in a study with 26 head and neck cancer patients [163]. The intervention group rinsed the mouth three times per day with 5 mL of an A. vera mouthwash, while the control group repeated the procedure with benzydamine mouthwash. The protocol was applied from the first day of RT until the end of the treatment, demonstrating that A. vera mouthwash was as efficient as benzydamine at reducing the severity of RT-induced OM, without differences between them (p < 0.09) [163].

3.6. Curcuma Longa

Curcuma longa, also known as turmeric, is an herb that is extensively grown in Asia [164] and is often used culinarily as a spice and in traditional Asian medical treatments for depression, stress, infection, and dermatological diseases [165,166]. Various compounds were identified in this plant, including polyphenols, sesquiterpenes, diterpenes, triterpenoids, sterols, and alkaloids [165,167]. Among these, the most studied component of C. longa is curcumin, a lipophilic polyphenol extracted from the rhizomes of C. longa, which represent 2–5% of turmeric [164,165].
Due to the antioxidant, anti-inflammatory, and anticancer effects of curcumin, it has an important role in the prevention of depression, cancer, and pro-inflammatory, neurodegenerative, diabetic, autoimmune, and cardiovascular diseases [168,169,170,171,172]. Furthermore, curcumin has antimicrobial, insecticidal, larvicidal, and radioprotective activities [165]. Curcumin mediates its effects through direct or indirect interactions with growth factors, kinases, enzymes, transcription factors, receptors, and proteins that regulate cell proliferation and apoptosis [168,173,174,175]. In the case of OM, the beneficial effects of curcumin may be related with the upregulation of TGF-β-1, which promotes re-epithelialization through the stimulation of fibronectin and collagen production by fibroblasts, while increasing the rate of granulation [168,176,177]. TGF-β-1also promotes the removal of dead tissue by enhancing the recruitment of macrophages [177]. Aside from that, curcumin potently inhibits the activation of nuclear factor-κB (NF-κB), but activates others, such as the nuclear factor erythroid 2-related factor 2 (Nrf2) [168,176,177]. COX-2, the inducible form of COX, can be selectively induced by mitogenic and inflammatory stimuli, resulting in enhanced synthesis of prostaglandins, such as IL-6. The activation of NF-κB significantly upregulates superoxide dismutase (SOD) expression [168,176,177]. Curcumin also enhanced the expression of antioxidant enzymes such as SOD, catalase (CAT), glutathione (GSH), and glutathione peroxidase (GSH-px) through the regulation of Nrf2 [168,176,177].
The wound-healing ability of curcumin is accelerated by its antioxidant activity, as it decreases the levels of lipid peroxides (LPs) and increases the activity levels of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) [178].
In a placebo-controlled study, an assessment of the tolerability of a curcumin mouthwash for the prevention of OM in pediatric patients undergoing CT was performed in a group of seven pediatric and young-adult patients [179]. In this study, which was developed without a control group for ethical reasons, in addition to the standard preventive oral care consisting of 0.2% chlorhexidine mouthwash for 30 s twice per day, the patients also used a mouthwash with 10 drops of Curcumall® (a dietary supplement containing turmeric, curcumin and ginger) twice per day during the CT treatment. The researcher concluded that curcumin mouthwash was safe and well tolerated by the patients [179]. The efficiency of curcumin mouthwash in cancer patients undergoing RT and suffering from OM was evaluated in a randomized trial involving 20 patients [180]. The study group used 0.004% curcumin mouthwash diluted at a ratio of 1:5 for 1 min three times per day for 20 days, while the control group was treated with standard preventive oral care using a commercially available 0.2% chlorhexidine mouthwash to be used in a 1:1 dilution for 1 min three times per day for 20 days. Curcumin promoted faster wound healing and better patient compliance in the management of RT-induced OM (p < 0.001) [180]. In another double-blind randomized clinical trial, the effects of curcumin encapsulated in nanomicelles on OM in 32 head and neck cancer patients receiving RT were evaluated [181]. During the RT, patients in the treatment group received daily one capsule of SinaCurcumin® (Exir Nano Sina Company, Tehran, Iran), which contained 80 mg of curcumin-loaded nanomicelles. The control group received placebo tablets containing lactose. There were statistically significant differences (p < 0.05) between the two groups in the severity of OM, as all of the patients in the placebo group developed OM versus the 32% of the case group [181].

3.7. Olea Europaea

Olive leaf extract is a natural product extracted from Olea europaea, which is traditionally used to treat and prevent hypertension and diabetes due to its antioxidant, anti-inflammatory, anticancer, antiapoptotic, antimicrobial, hypoglycemic, and diuretic properties [182,183,184,185,186,187]. The leaves of O. europaea contain a high concentration of phenolic compounds (1450 mg/100 g of fresh leaf), with secoiridoid oleuropein, verbascoside, rutin, luteolin-7-glucoside, and hydroxytyrosol as the main phenolic constituents [188]. Oleuropein is possibly the main active compound promoting the wound-healing activity of olive leaf extract, as it increases collagen fiber deposition and advanced re-epithelialization [189,190]. Furthermore, it has been demonstrated that oleuropein decreases oxidative stress and inflammation through the modulation of the COX-2, AMPF, eNOS, MAPK, and apoptosis cell signaling pathways in in vivo studies on mice [187]. In addition, olive leaf extract also inhibited the aggregation platelets in in vitro studies [186].
In 2013, the effect of a mouth rinse containing olive leaf extract on the prevention of severe OM in CT-receiving patients, as well as an estimation of its effect on the salivary levels of pro-inflammatory cytokines, was assessed in a prospective, randomized, double-blind, placebo-controlled cross-over study design involving 25 cancer patients [182]. The studied drugs (olive leaf extract at 333 mg/mL, benzydamine hydrochloride at 0.15 g/100 mL, or normal saline) were self-administered 3–4 times daily for 14 days, starting on the first day of chemotherapy. The patients were evaluated weekly until 15 days after CT for each cycle. The findings indicated that the olive leaf extract could effectively reduce the OM rates (p < 0.001) by decreasing the salivary levels of IL-1β and TNF-α [182]. Briefly, Ahmed et al. performed an experimental animal study and a prospective, randomized, double-blind, placebo-controlled cross-over study to evaluate the management of OM with mouthwashes containing olive leaf extract [191]. In the animal study, 45 male albino rats received two intraperitoneal injections of 5-fluorouracil (60 mg/kg) on day 0 and day 2. The first group received normal saline, the second group received olive leaf extract (333 mg/mL), and the third group received benzydamine hydrochloride (0.15 g/100 mL). By the end of the study (day 14), the control group presented ulcerated connective tissue that was not completely covered by epithelium, and there was evidence of necrosis and degeneration. The animals with the olive leaf extract and benzydamine hydrochloride presented a totally re-epithelialized mucosal surface with hyperkeratinization and hyperplasia, while the sub-epithelia were more organized, with decreased cellularity of fibrous tissue [191]. In a clinical study, 62 CT-receiving patients were divided to receive olive leaf extract, benzydamine hydrochloride, or a placebo in the form of a mouth rinse, and the treatment was changed in the next chemotherapy cycle for each patient (cross-over design) [191]. Mouth rinses were self-administered 3–4 times per day for 14 days from the start of the CT. When compared to the benzydamine hydrochloride and the control, the olive leaf extract more efficiently reduced the oral pain, dysphagia, and functional impairment of eating (p < 0.001) [191].

3.8. Glycyrrhiza glabra

Glycyrrhiza glabra, commonly known as licorice, is one of the most important herbal medicines for traditional Chinese medicine and Japanese Kampo medicine [192]. It is traditionally used to relieve inflammation, gastric and peptic ulcers, arthritis, eye and liver disorders, hyperacidity, and sex-hormone imbalance [193,194,195,196,197,198,199,200,201]. This plant has attracted the attention of the pharmacological field due to its antimicrobial, antiviral, and anti-inflammatory properties [202,203,204,205]. The roots of G. glabra have been found to possess many secondary metabolites, with numerous pharmacological properties that contribute to their medicinal use, including flavonoids (such as liquirtin, rhamnoliquirilin, liquiritigenin, and prenyllicoflavone A) and volatile components (including pentanol, hexanol, tetramethyl pyrazine, linalool, and terpinen-4-ol) [206]. The essential oil extracted from the roots of G. glabra contains propionic acid, 1-methyl-2-formylpyrrole, benzoic acid, 2,3-butanediol, and ethyl linoleate, among other compounds. The roots of G. glabra are also composed of 20% moisture, 3–16% sugars, 30% starch, and 6% ash [207].
The main biologically active components of G. glabra are dipotassium glycyrrhizinate, glycyrrhizin, also known as glycyrrhizic acid, and its aglycone, glycyrrhetinic acid [206]. Dipotassium glycyrrhizinate has similar properties to those of corticosteroids, namely, anti-inflammatory, antiallergic, and antibiotic activities, without the side effects of allergic reactions on the skin [208]. This property is due to dipotassium glycyrrhizinate’s ability to efficiently inhibit the activity of phospholipase A2 enzyme, which is necessary for several inflammatory processes [209,210,211]. Moreover, it is able to avoid damage to the extracellular matrix by inhibiting the activity of hyaluronidase enzyme, histamine release, inflammatory chemical mediators, leukotrienes, and prostaglandins [212]. Glycyrrhizic acid inhibits prostaglandin E2 synthesis by suppressing the activity of COX-2, resulting in the augmentation of NO production through the enhancement of iNOS mRNA secretion and indirectly preventing platelet aggregation [211,213,214]. The anti-inflammatory activity of glycyrrhizic and glycyrrhetinic acids is realized through cytokines such as 1β, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12, and IL-17, IFN-γ, and TNF-α [215,216,217]. Moreover, these compounds also present immunomodulatory activity through their interaction with different transcription factors, such as NF-κB, as well as signal transducers and activator of transcriptions (STAT- 3 and STAT-6) [215].
Najafi et al. conducted a double-blind clinical trial to evaluate the potential effect of G. glabra extract on cancer patients under head and neck radiotherapy [207]. The experimental group received a 50% extract of Glycyrrhiza (hydroalcoholic extract) and the placebo group received a brown-colored water. The patients were asked to use 20 cc twice per day for 14 days after the beginning of RT. According to the results obtained, Glycyrrhiza extract efficiently decreased the OM, wound size, and irritation (p < 0.001) [207]. The effect of G. glabra on head and neck cancer patients receiving RT was also evaluated in a small randomized study with six patients who were assigned to receive a licorice mucoadhesive film or a placebo mucoadhesive film [218]. The level of pain and the mucositis severity were significantly lower in the licorice-mucoadhesive-film-receiving patients in the last 2 weeks of the clinical trial (weeks 3 and 4) (p < 0.05) [218].
The efficiency of a G. glabra root extract in preventing CT-induced OM in colon cancer patients was evaluated in a double-blind randomized clinical trial that involved 72 patients [219]. The treatment group received 5% licorice root extract, and the control group received a combined mouthwash composed of aluminum, magnesium, diphenhydramine, nystatin powder, and 2% lidocaine. For one week, from the first day of CT, both mouthwashes were used daily, every 8 h, at a dose of 10 cc. The researchers did not observe differences between the two groups in terms of the incidence and severity of OM (p > 0.05) [219].

3.9. Matricaria Recutita

The chamomile plant, Chamomilla recutita or Matricaria recutita, one of the most common medicinal plants, is characterized by flowers with anti-inflammatory, antibacterial, and antifungal properties [220]. It is mainly used to treat different inflammatory conditions of the skin and mucosa, as it promotes faster a wound-healing process in comparison to corticosteroids [220,221]. M. recutita owes its therapeutic activity to chamazulene, α-bisabolol, bisabolol oxides, spiroethers, and flavonoids [220]. Flavonoids—in particular, apigenin-7-glucoside—have been found to be responsible for the anti-inflammatory activity that may be involved in recuperation from OM [222]. Pre-clinical studies showed evidence of the anti-inflammatory action of M. recutita through the inhibition of COX-2 and IL-6 production [223,224].
In a small comparative study with random assignment, dos Reis et al. evaluated the efficacy of M. recutita infusion cryotherapy for the prevention and reduction of the intensity of OM in gastric and colorectal cancer patients [225]. The study was performed during the first course (5 days) of CT. The patients in the M. recutita group received a cup of ice chips made with an M. recutita infusion at 2.5%, while the control group received a cup of ice chips made with pure water. The patients in both groups were instructed to swish the ice chips around in their mouths for at least 30 min, starting 5 min before the CT infusion. The M. recutita group presented less pain and had no ulcerations when compared to the control group [225]. The effects and the percentage of extract necessary to reduce the incidence and intensity of OM in patients undergoing hematopoietic stem cell transplantation were assessed in a randomized, controlled, phase II clinical trial [221]. All 40 patients received standard oral care, while the treatment group received an additional mouthwash containing a liquid extract of M. recutita at 0.5%, 1%, or 2%. When compared with the control group, the M. recutita group at 1% (equivalent to 0.108 mg of apigenin-7-glucoside/mL) demonstrated have reduced incidence, intensity, and duration of OM in patients undergoing hematopoietic stem cell transplantation (p < 0.01) [221]. Shabanloei et al. performed a randomized, double-blind clinical trial between alloporinol and M. recutita extract in the prevention of OM in CT-receiving patients [226]. Group 1 received 5 mg/mL of allopurinol, group 2 received a solution of 8 g of M. recutita in 50 cc, and the control group received a normal saline solution as a mouthwash. All patients gargled daily, four times per day, for the 16 days following the beginning of CT. The researchers concluded that both the allopurinol and M. recutita mouthwashes were effective in reducing post-CT OM, with no significant differences in the mean stomatitis (p = 0.59) and stomatitis pain (p = 0.071) [226].

3.10. Calendula officinalis

Calendula officinalis, commonly known as marigold, has been used for centuries as a topical and oral herbal remedy due to its bactericidal, antioxidant, anti-inflammatory, antiseptic, hepatoprotective, and anti-metastatic effects, with applications in blood purification and treatment of herpes, keratolytic radiation dermatitis, wounds, and scars, and as an antispasmodic [227,228,229]. The main compounds that contribute to its medicinal use are triterpenoids, flavonoids, oleanolic acid, faradiol, glycosides, quinones, tannins, coumarins, carotenoids, saponins, alkaloids, phenolic acids, and amino acids [227]. Triterpenoids provide anti-inflammatory and anti-edematous effects, in addition to stimulating the proliferation of fibroblasts, possibly through the inhibition of COX-2, C3-convertase, and 5-lipoxygenase [230,231,232]. Flavonoids are reported to have anti-inflammatory, antioxidant, and anti-edematous properties, in addition to their inhibition of lipoxygenase enzymes and mast cells [233].
The potential of C. officinalis extract for the healing of 5-fluorouracil-induced OM was studied in hamsters [229]. OM was induced in 60 male hamsters on days 0, 5, and 10 through the intraperitoneal administration of 5-fluorouracil (60 mg/kg). The cheek pouch was scratched with a needle once per day, from day 1 until day 12, when erythematous changes were noted. The treatment of OM started on days 12–17 with the topical application of a gel once a day. The animals were divided into four groups: 12 without treatment as control animals, 15 treated with 5% C. officinalis gel, 15 treated with 10% C. officinalis gel, and 15 treated with the gel base. The C. officinalis gel (5% and 10%) significantly reduced the microscopic and macroscopic scores of OM when compared with the gel base and the control group. Moreover, the animals of the treatment groups gained more weight than those in the gel base and the control groups [229]. In humans, the effect of C. officinalis on OM was evaluated in a placebo-controlled clinical trial with 40 patients with neck and head cancers under RT or concurrent CT [234]. Patients were given 5 mL of either placebo or a 2% C. officinalis extract gel mouthwash to be held for at least 1 min in the oral cavity two times per day. Compared to the placebo group, the intensity of OM was significantly lower in the C. officinalis mouthwash group at weeks 2, 3, and 6 (p < 0.048). According to the same study, the high content of flavonoids and phenolic compounds and the antioxidant activity may be responsible for the protective effect of C. officinalis in RT-induced OM [234].

3.11. Other Compounds

In addition to the compounds mentioned above, different experiments were also performed to evaluate the potential of other natural compounds for preventing/treating OM. However, due to the low number of studies published, not only regarding the OM application, but also with respect to the molecular mechanism of action enrolled, a section was not dedicated to them in this review. Table 4 summarizes the different natural products in these circumstances.

4. Conclusions and Future Perspectives

OM is a common and incapacitating side effect of antineoplastic therapies. The increased knowledge of its pathogenesis allows a better prediction of a patient’s risk with the aim of adapting the management protocols and improving the development of new therapies. Nevertheless, standard guidelines for preventing and treating OM do not display significant effectiveness. The interest in natural products as potential therapeutic drugs has increased in recent years, as they have the advantage of being accessible and generating minimal side effects, with potential properties that include anti-inflammatory, antioxidant, antimicrobial, antiulcerative, and wound-healing capacities. In addition, over recent years, there have been multiple efforts to develop naturally based therapies, with natural compounds being tested in model organisms and clinical trials that are currently ongoing. However, the environment of the oral cavity is a complex system that is divided into two functional layers—the epithelium (thick and avascular) and the underlying tissue (vascular)—that are anatomically different, which affects their permeability to drugs and the capacity for maintaining a system for a certain period [255]. The buccal mucosa, which is composed of epithelial cells, provides a large surface area of almost 100 cm2 [256]. This area is ideal for attaching a drug delivery system, providing a permeability that is 4 to 4000 times higher than that of skin [256,257,258,259]. Oral administration provides the advantage of a simple administration that does not suffer from the first-pass metabolism and that is safe and increases the drug availability. In addition, this route has a rapid action, reduced side effects, easy access to the local condition, and great patient compliance [255,260]. These characteristics make the buccal mucosa an optimal solution for the systemic and local treatment of OM [261]. However, it also has limitations that are associated with a functionalized protective barrier. The presence of saliva and its enzymatic action, as well as the constant mechanical pressure caused by eating and speaking movements, may compromise the penetration of the drug present in the delivery system; as such, the application of mucoadhesive components may be required to solve this issue, but this can compromise the therapeutic effectiveness [255,257,258,259].
Due to the characteristics of the oral cavity, it is necessary to develop novel strategies for overcoming topical delivery, such as mucoadhesive dosage forms (e.g., films, tablets). For the treatment of oral diseases, the most suitable formulations investigated were in the form of tablets, films, sprays, mouthwashes, gels, and pastes [24,259,262]. Gel and film formulations were evaluated in hamsters with CT-induced mucositis. By the 28th day, the hamsters’ mucosa appeared to be healed, as no erythema or edema was visible. These results proved their efficiency, as the animals’ survival was higher than in the control group, and these treatments showed promising potential for a function as an occlusive patch and for delivering therapeutic compounds [261]. Films containing ethanolic propolis extract also presented optimal mucoadhesion capacity, ensuring the release of propolis compounds, a good stability, a high swelling capacity, and antimicrobial effects against S. aureus [263]. In addition, the incorporation of nanoparticles in the forms of dosage for buccal drug delivery has recently been encouraged [24,256,264]. Furthermore, nanoparticles could transport many therapeutic agents [24,256]. Functional and biocompatible carriers that display chemical stability are sought in the innovation of buccal drug delivery systems [264,265,266,267]. Chitosan is an example of a biopolymer that is biologically safe and bioadhesive, and it has been used in several studies for the development of drug delivery systems, as it has longer retention periods in the oral mucosa [261,268,269]. In addition, it inhibits the attachment of C. albicans to human oral mucosal cells [261,268,269].
A SWOT diagram (Figure 2) was constructed with the aim of summarizing the previously described strengths, weaknesses, opportunities, and threats of employing natural products for the prevention/treatment of OM.
Despite the significant advances made in this area, more investigations are needed to ensure that these formulations reach the pharmaceutical market, and few have been published regarding this topic with natural products.

Author Contributions

Conceptualization, F.R.; methodology, A.S.F., C.M., A.M.S. and F.R.; validation, F.R.; investigation, A.S.F. and C.M.; resources, F.R.; writing—original draft preparation, A.S.F., C.M. and A.M.S.; writing—review and editing, F.R., P.C. and C.D.-M.; supervision, F.R., C.D.-M. and P.C.; project administration, F.R.; funding acquisition, F.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the project EXPL/BAA-GR/0663/2021—Kiwi4Health—Exploring the Eco-Innovative Re-Use of Kiwiberry, supported by national funds from Fundação para a Ciência e a Tecnologia (Portugal), as well as the project MTS/SAS/0077/2020-Honey+-New reasons to care honey from the Natural Park of Montesinho: A bioindicator of environmental quality & its therapeutic potential.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available on request due to restrictions, e.g., privacy or ethical restrictions.

Acknowledgments

Ana Sofia Ferreira (SFRH/BD/7519/2020) and Ana Margarida Silva (SFRH/BD/144994/2019) are thankful for their Ph.D. grants financed by POPH-QREN and subsidized by the European Science Foundation and Ministério da Ciência, Tecnologia e Ensino Superior. Catarina Macedo is thankful for her scholarship from the project EXPL/BAA-GR/0663/2021. Francisca Rodrigues (CEECIND/01886/2020) is thankful for her contract financed by FCT/MCTES—CEEC Individual Program Contract. This work was financially supported by Portuguese national funds through projects UIDB/50006/2020, UIDP/50006/2020, and LA/P/0008/2020, from the Fundação para a Ciência e a Tecnologia (FCT)/Ministério da Ciência, Tecnologia e Ensino Superior (MCTES). This work was also financed by national funds from FCT—Fundação para a Ciência e a Tecnologia, I.P., in the scope of the project UIDP/04378/2020 and UIDB/04378/2020 of the Research Unit on Applied Molecular Biosciences—UCIBIO and the project LA/P/0140/2020 of the Associate Laboratory Institute for Health and Bioeconomy—i4HB.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Cinausero, M.; Aprile, G.; Ermacora, P.; Basile, D.; Vitale, M.G.; Fanotto, V.; Parisi, G.; Calvetti, L.; Sonis, S.T. New Frontiers in the Pathobiology and Treatment of Cancer Regimen-Related Mucosal Injury. Front. Pharmacol. 2017, 8, 354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Sonis, S.T. Mucositis: The impact, biology and therapeutic opportunities of oral mucositis. Oral Oncol. 2009, 45, 1015–1020. [Google Scholar] [CrossRef]
  4. Oronsky, B.; Goyal, S.; Kim, M.M.; Cabrales, P.; Lybeck, M.; Caroen, S.; Oronsky, N.; Burbano, E.; Carter, C.; Oronsky, A. A Review of Clinical Radioprotection and Chemoprotection for Oral Mucositis. Transl. Oncol. 2018, 11, 771–778. [Google Scholar] [CrossRef] [PubMed]
  5. Shetty, S.S.; Maruthi, M.; Dhara, V.; de Arruda, J.A.A.; Abreu, L.G.; Mesquita, R.A.; Teixeira, A.L.; Silva, T.A.; Merchant, Y. Oral mucositis: Current knowledge and future directions. Dis. Mon. 2021; 101300, in press. [Google Scholar] [CrossRef] [PubMed]
  6. Sonis, S.T. A hypothesis for the pathogenesis of radiation-induced oral mucositis: When biological challenges exceed physiologic protective mechanisms. Implications for pharmacological prevention and treatment. Support. Care Cancer 2021, 29, 4939–4947. [Google Scholar] [CrossRef]
  7. Kawashita, Y.; Soutome, S.; Umeda, M.; Saito, T. Oral management strategies for radiotherapy of head and neck cancer. Jpn. Dent. Sci. Rev. 2020, 56, 62–67. [Google Scholar] [CrossRef]
  8. Lalla, R.V.; Saunders, D.P.; Peterson, D.E. Chemotherapy or radiation-induced oral mucositis. Dent. Clin. N. Am. 2014, 58, 341–349. [Google Scholar] [CrossRef]
  9. Singh, V.; Singh, A.K. Oral mucositis. Natl. J. Maxillofac. Surg. 2020, 11, 159–168. [Google Scholar] [CrossRef]
  10. Pulito, C.; Cristaudo, A.; Porta, C.; Zapperi, S.; Blandino, G.; Morrone, A.; Strano, S. Oral mucositis: The hidden side of cancer therapy. J. Exp. Clin. Cancer Res. 2020, 39, 210. [Google Scholar] [CrossRef]
  11. Daugelaite, G.; Uzkuraityte, K.; Jagelaviciene, E.; Filipauskas, A. Prevention and Treatment of Chemotherapy and Radiotherapy Induced Oral Mucositis. Medicina 2019, 55, 25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Chen, S.C.; Lai, Y.H.; Huang, B.S.; Lin, C.Y.; Fan, K.H.; Chang, J.T. Changes and predictors of radiation-induced oral mucositis in patients with oral cavity cancer during active treatment. Eur. J. Oncol. Nurs. 2015, 19, 214–219. [Google Scholar] [CrossRef] [PubMed]
  13. De Sanctis, V.; Bossi, P.; Sanguineti, G.; Trippa, F.; Ferrari, D.; Bacigalupo, A.; Ripamonti, C.I.; Buglione, M.; Pergolizzi, S.; Langendjik, J.A.; et al. Mucositis in head and neck cancer patients treated with radiotherapy and systemic therapies: Literature review and consensus statements. Crit. Rev. Oncol. Hematol. 2016, 100, 147–166. [Google Scholar] [CrossRef] [PubMed]
  14. Fidan, O.; Arslan, S. Development and Validation of the Oral Mucositis Risk Assessment Scale in Hematology Patients. Semin. Oncol. Nurs. 2021, 37, 151159. [Google Scholar] [CrossRef]
  15. Hearnden, V.; Sankar, V.; Hull, K.; Juras, D.V.; Greenberg, M.; Kerr, A.R.; Lockhart, P.B.; Patton, L.L.; Porter, S.; Thornhill, M.H. New developments and opportunities in oral mucosal drug delivery for local and systemic disease. Adv. Drug Deliv. Rev. 2012, 64, 16–28. [Google Scholar] [CrossRef]
  16. Elad, S.; Yarom, N.; Zadik, Y.; Kuten-Shorrer, M.; Sonis, S.T. The broadening scope of oral mucositis and oral ulcerative mucosal toxicities of anticancer therapies. CA Cancer J. Clin. 2022, 72, 57–77. [Google Scholar] [CrossRef]
  17. Kawashita, Y.; Koyama, Y.; Kurita, H.; Otsuru, M.; Ota, Y.; Okura, M.; Horie, A.; Sekiya, H.; Umeda, M. Effectiveness of a comprehensive oral management protocol for the prevention of severe oral mucositis in patients receiving radiotherapy with or without chemotherapy for oral cancer: A multicentre, phase II, randomized controlled trial. Int. J. Oral Maxillofac. Surg. 2019, 48, 857–864. [Google Scholar] [CrossRef]
  18. Basile, D.; Di Nardo, P.; Corvaja, C.; Garattini, S.K.; Pelizzari, G.; Lisanti, C.; Bortot, L.; Da Ros, L.; Bartoletti, M.; Borghi, M.; et al. Mucosal Injury during Anti-Cancer Treatment: From Pathobiology to Bedside. Cancers 2019, 11, 857. [Google Scholar] [CrossRef] [Green Version]
  19. Elting, L.S.; Chang, Y.C. Costs of Oral Complications of Cancer Therapies: Estimates and a Blueprint for Future Study. J. Natl. Cancer Inst. Monogr. 2019, 2019, lgz010. [Google Scholar] [CrossRef]
  20. Rodrigues-Oliveira, L.; Kowalski, L.P.; Santos, M.; Marta, G.N.; Bensadoun, R.J.; Martins, M.D.; Lopes, M.A.; Castro, G., Jr.; William, W.N., Jr.; Chaves, A.L.F.; et al. Direct costs associated with the management of mucositis: A systematic review. Oral Oncol. 2021, 118, 105296. [Google Scholar] [CrossRef]
  21. Sampson, M.M.; Nanjappa, S.; Greene, J.N. Mucositis and oral infections secondary to gram negative rods in patients with prolonged neutropenia. IDCases 2017, 9, 101–103. [Google Scholar] [CrossRef] [PubMed]
  22. Alkhouli, M.; Laflouf, M.; Comisi, J.C. Assessing the topical application efficiency of two biological agents in managing chemotherapy-induced oral mucositis in children: A randomized clinical trial. J. Oral Biol. Craniofac. Res. 2021, 11, 373–378. [Google Scholar] [CrossRef] [PubMed]
  23. Kusiak, A.; Jereczek-Fossa, B.A.; Cichonska, D.; Alterio, D. Oncological-Therapy Related Oral Mucositis as an Interdisciplinary Problem-Literature Review. Int. J. Environ. Res. Public Health 2020, 17, 2464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Tran, P.H.L.; Duan, W.; Tran, T.T.D. Recent developments of nanoparticle-delivered dosage forms for buccal delivery. Int. J. Pharm. 2019, 571, 118697. [Google Scholar] [CrossRef] [PubMed]
  25. Moslemi, D.; Nokhandani, A.M.; Otaghsaraei, M.T.; Moghadamnia, Y.; Kazemi, S.; Moghadamnia, A.A. Management of chemo/radiation-induced oral mucositis in patients with head and neck cancer: A review of the current literature. Radiother. Oncol. 2016, 120, 13–20. [Google Scholar] [CrossRef]
  26. Patel, P.; Robinson, P.D.; Baggott, C.; Gibson, P.; Ljungman, G.; Massey, N.; Ottaviani, G.; Phillips, R.; Revon-Riviere, G.; Treister, N.; et al. Clinical practice guideline for the prevention of oral and oropharyngeal mucositis in pediatric cancer and hematopoietic stem cell transplant patients: 2021 update. Eur. J. Cancer 2021, 154, 92–101. [Google Scholar] [CrossRef]
  27. Ayago Flores, D.; Ferriols Lisart, R. Effectiveness of palifermin in the prevention of oral mucositis in patients with haematological cancers. Farm. Hosp. 2010, 34, 163–169. [Google Scholar] [CrossRef]
  28. de Sousa Melo, A.; de Lima Dantas, J.B.; Medrado, A.; Lima, H.R.; Martins, G.B.; Carrera, M. Nutritional supplements in the management of oral mucositis in patients with head and neck cancer: Narrative literary review. Clin. Nutr. ESPEN 2021, 43, 31–38. [Google Scholar] [CrossRef]
  29. Yarom, N.; Ariyawardana, A.; Hovan, A.; Barasch, A.; Jarvis, V.; Jensen, S.B.; Zadik, Y.; Elad, S.; Bowen, J.; Lalla, R.V. Systematic review of natural agents for the management of oral mucositis in cancer patients. Support. Care Cancer 2013, 21, 3209–3221. [Google Scholar] [CrossRef]
  30. Yarom, N.; Hovan, A.; Bossi, P.; Ariyawardana, A.; Jensen, S.B.; Gobbo, M.; Saca-Hazboun, H.; Kandwal, A.; Majorana, A.; Ottaviani, G.; et al. Correction to: Systematic review of natural and miscellaneous agents, for the management of oral mucositis in cancer patients and Clinical Practice Guidelines—Part 1: Vitamins, minerals and nutritional supplements. Support. Care Cancer 2021, 29, 4175–4176. [Google Scholar] [CrossRef]
  31. Konuk Sener, D.; Aydin, M.; Cangur, S.; Guven, E. The Effect of Oral Care with Chlorhexidine, Vitamin E and Honey on Mucositis in Pediatric Intensive Care Patients: A Randomized Controlled Trial. J. Pediatric Nurs. 2019, 45, e95–e101. [Google Scholar] [CrossRef] [PubMed]
  32. Lima, I.; de Fatima Souto Maior, L.; Gueiros, L.A.M.; Leao, J.C.; Higino, J.S.; Carvalho, A.A.T. Clinical applicability of natural products for prevention and treatment of oral mucositis: A systematic review and meta-analysis. Clin. Oral Investig. 2021, 25, 4115–4124. [Google Scholar] [CrossRef] [PubMed]
  33. Yang, C.; Tang, H.; Wang, L.; Peng, R.; Bai, F.; Shan, Y.; Yu, Z.; Zhou, P.; Cong, Y. Dimethyl Sulfoxide Prevents Radiation-Induced Oral Mucositis through Facilitating DNA Double-Strand Break Repair in Epithelial Stem Cells. Int. J. Radiat. Oncol. Biol. Phys. 2018, 102, 1577–1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Anderson, P.M.; Lalla, R.V. Glutamine for Amelioration of Radiation and Chemotherapy Associated Mucositis during Cancer Therapy. Nutrients 2020, 12, 1675. [Google Scholar] [CrossRef]
  35. Charalambous, M.; Raftopoulos, V.; Paikousis, L.; Katodritis, N.; Lambrinou, E.; Vomvas, D.; Georgiou, M.; Charalambous, A. The effect of the use of thyme honey in minimizing radiation-induced oral mucositis in head and neck cancer patients: A randomized controlled trial. Eur. J. Oncol. Nurs. 2018, 34, 89–97, (clinicaltrials.gov identifier NCT01465308). [Google Scholar] [CrossRef]
  36. Charalambous, M.; Raftopoulos, V.; Lambrinou, E.; Charalambous, A. The effectiveness of honey for the management of radiotherapy-induced oral mucositis in head and neck cancer patients: A systematic review of clinical trials. Eur. J. Integr. Med. 2013, 5, 217–225. [Google Scholar] [CrossRef]
  37. Ramsay, E.I.; Rao, S.; Madathil, L.; Hegde, S.K.; Baliga-Rao, M.P.; George, T.; Baliga, M.S. Honey in oral health and care: A mini review. J. Oral Biosci. 2019, 61, 32–36. [Google Scholar] [CrossRef]
  38. Munstedt, K.; Momm, F.; Hubner, J. Honey in the management of side effects of radiotherapy- or radio/chemotherapy-induced oral mucositis. A systematic review. Complement. Ther. Clin. Pract. 2019, 34, 145–152. [Google Scholar] [CrossRef]
  39. Pires Marques, E.C.; Piccolo Lopes, F.; Nascimento, I.C.; Morelli, J.; Pereira, M.V.; Machado Meiken, V.M.; Pinheiro, S.L. Photobiomodulation and photodynamic therapy for the treatment of oral mucositis in patients with cancer. Photodiagn. Photodyn. Ther. 2020, 29, 101621. [Google Scholar] [CrossRef]
  40. de Carvalho, P.A.G.; Lessa, R.C.; Carraro, D.M.; Assis Pellizzon, A.C.; Jaguar, G.C.; Alves, F.A. Three photobiomodulation protocols in the prevention/treatment of radiotherapy-induced oral mucositis. Photodiagn. Photodyn. Ther. 2020, 31, 101906. [Google Scholar] [CrossRef]
  41. Cotomacio, C.C.; Calarga, C.C.; Yshikawa, B.K.; Arana-Chavez, V.E.; Simoes, A. Wound healing process with different photobiomodulation therapy protocols to treat 5-FU-induced oral mucositis in hamsters. Arch. Oral Biol. 2021, 131, 105250. [Google Scholar] [CrossRef] [PubMed]
  42. Blakaj, A.; Bonomi, M.; Gamez, M.E.; Blakaj, D.M. Oral mucositis in head and neck cancer: Evidence-based management and review of clinical trial data. Oral Oncol. 2019, 95, 29–34. [Google Scholar] [CrossRef] [PubMed]
  43. Campos, M.I.; Campos, C.N.; Aarestrup, F.M.; Aarestrup, B.J. Oral mucositis in cancer treatment: Natural history, prevention and treatment. Mol. Clin. Oncol. 2014, 2, 337–340. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Abt, E. Probiotics May Lower the Risk of Oral Mucositis in Cancer Patients. J. Evid. Based Dent. Pract. 2021, 21, 101639. [Google Scholar] [CrossRef]
  45. Wardill, H.R.; Sonis, S.T.; Blijlevens, N.M.A.; Van Sebille, Y.Z.A.; Ciorba, M.A.; Loeffen, E.A.H.; Cheng, K.K.F.; Bossi, P.; Porcello, L.; Castillo, D.A.; et al. Prediction of mucositis risk secondary to cancer therapy: A systematic review of current evidence and call to action. Support. Care Cancer 2020, 28, 5059–5073. [Google Scholar] [CrossRef]
  46. Elad, S.; Cheng, K.K.F.; Lalla, R.V.; Yarom, N.; Hong, C.; Logan, R.M.; Bowen, J.; Gibson, R.; Saunders, D.P.; Zadik, Y.; et al. MASCC/ISOO clinical practice guidelines for the management of mucositis secondary to cancer therapy. Cancer 2020, 126, 4423–4431. [Google Scholar] [CrossRef]
  47. Logan, R.M.; Stringer, A.M.; Bowen, J.M.; Yeoh, A.S.; Gibson, R.J.; Sonis, S.T.; Keefe, D.M. The role of pro-inflammatory cytokines in cancer treatment-induced alimentary tract mucositis: Pathobiology, animal models and cytotoxic drugs. Cancer Treat. Rev. 2007, 33, 448–460. [Google Scholar] [CrossRef]
  48. Bertolini, M.; Sobue, T.; Thompson, A.; Dongari-Bagtzoglou, A. Chemotherapy Induces Oral Mucositis in Mice without Additional Noxious Stimuli. Transl. Oncol. 2017, 10, 612–620. [Google Scholar] [CrossRef]
  49. Raber-Durlacher, J.E.; Elad, S.; Barasch, A. Oral mucositis. Oral Oncol. 2010, 46, 452–456. [Google Scholar] [CrossRef]
  50. Bailly, C. Potential use of edaravone to reduce specific side effects of chemo-, radio- and immuno-therapy of cancers. Int. Immunopharmacol. 2019, 77, 105967. [Google Scholar] [CrossRef]
  51. Vigarios, E.; Epstein, J.B.; Sibaud, V. Oral mucosal changes induced by anticancer targeted therapies and immune checkpoint inhibitors. Support. Care Cancer 2017, 25, 1713–1739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Kudrimoti, M.; Curtis, A.; Azawi, S.; Worden, F.; Katz, S.; Adkins, D.; Bonomi, M.; Elder, J.; Sonis, S.T.; Straube, R.; et al. Dusquetide: A novel innate defense regulator demonstrating a significant and consistent reduction in the duration of oral mucositis in preclinical data and a randomized, placebo-controlled phase 2a clinical study. J. Biotechnol. 2016, 239, 115–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Ariyawardana, A.; Cheng, K.K.F.; Kandwal, A.; Tilly, V.; Al-Azri, A.R.; Galiti, D.; Chiang, K.; Vaddi, A.; Ranna, V.; Nicolatou-Galitis, O.; et al. Systematic review of anti-inflammatory agents for the management of oral mucositis in cancer patients and clinical practice guidelines. Support. Care Cancer 2019, 27, 3985–3995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Davy, C.; Heathcote, S. A systematic review of interventions to mitigate radiotherapy-induced oral mucositis in head and neck cancer patients. Support. Care Cancer 2021, 29, 2187–2202. [Google Scholar] [CrossRef]
  55. Peterson, D.E.; Lalla, R.V. Oral mucositis: The new paradigms. Curr. Opin. Oncol. 2010, 22, 318–322. [Google Scholar] [CrossRef]
  56. Satheeshkumar, P.S.; El-Dallal, M.; Mohan, M.P. Feature selection and predicting chemotherapy-induced ulcerative mucositis using machine learning methods. Int. J. Med. Inform. 2021, 154, 104563. [Google Scholar] [CrossRef]
  57. Pratiwi, E.S.; Ismawati, N.D.S.; Ruslin, M. Prevalence and risk factors of oral mucositis in children with acute lymphoblastic leukemia in Dr. Soetomo Hospital Surabaya Indonesia. Enfermería Clínica 2020, 30, 289–292. [Google Scholar] [CrossRef]
  58. Soutome, S.; Yanamoto, S.; Nishii, M.; Kojima, Y.; Hasegawa, T.; Funahara, M.; Akashi, M.; Saito, T.; Umeda, M. Risk factors for severe radiation-induced oral mucositis in patients with oral cancer. J. Dent. Sci. 2021, 16, 1241–1246. [Google Scholar] [CrossRef]
  59. Parkhideh, S.; Zeraatkar, M.; Moradi, O.; Hajifathali, A.; Mehdizadeh, M.; Tavakoli-Ardakani, M. Azithromycin oral suspension in prevention and management of oral mucositis in patients undergoing hematopoietic stem cell transplantation: A randomized controlled trial. Support. Care Cancer 2022, 30, 251–257. [Google Scholar] [CrossRef]
  60. Aspinall, S.R.; Parker, J.K.; Khutoryanskiy, V.V. Oral care product formulations, properties and challenges. Colloids Surf. B 2021, 200, 111567. [Google Scholar] [CrossRef]
  61. Yamaguchi, T.; Makiguchi, T.; Nakamura, H.; Yamatsu, Y.; Hirai, Y.; Shoda, K.; Suzuki, K.; Kim, M.; Kurozumi, S.; Motegi, S.I.; et al. Impact of muscle volume loss on acute oral mucositis in patients undergoing concurrent chemoradiotherapy after oral cancer resection. Int. J. Oral Maxillofac. Surg. 2021, 50, 1195–1202. [Google Scholar] [CrossRef] [PubMed]
  62. Orlandi, E.; Iacovelli, N.A.; Rancati, T.; Cicchetti, A.; Bossi, P.; Pignoli, E.; Bergamini, C.; Licitra, L.; Fallai, C.; Valdagni, R.; et al. Multivariable model for predicting acute oral mucositis during combined IMRT and chemotherapy for locally advanced nasopharyngeal cancer patients. Oral Oncol. 2018, 86, 266–272. [Google Scholar] [CrossRef] [PubMed]
  63. Hong, C.H.L.; Gueiros, L.A.; Fulton, J.S.; Cheng, K.K.F.; Kandwal, A.; Galiti, D.; Fall-Dickson, J.M.; Johansen, J.; Ameringer, S.; Kataoka, T.; et al. Systematic review of basic oral care for the management of oral mucositis in cancer patients and clinical practice guidelines. Support. Care Cancer 2019, 27, 3949–3967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Inoue, Y.; Yamagata, K.; Nakamura, M.; Ohnishi, K.; Tabuchi, K.; Bukawa, H. Are Intraoral Stents Effective for Reducing the Severity of Oral Mucositis during Radiotherapy for Maxillary and Nasal Cavity Cancer? J. Oral Maxillofac. Surg. 2020, 78, 1214.e1–1214.e8. [Google Scholar] [CrossRef]
  65. Duzkaya, D.S.; Uysal, G.; Bozkurt, G.; Yakut, T. The Effect of Oral Care Using an Oral Health Care Guide on Preventing Mucositis in Pediatric Intensive Care. J. Pediatr. Nurs. 2017, 36, 98–102. [Google Scholar] [CrossRef]
  66. Edmans, J.G.; Murdoch, C.; Santocildes-Romero, M.E.; Hatton, P.V.; Colley, H.E.; Spain, S.G. Incorporation of lysozyme into a mucoadhesive electrospun patch for rapid protein delivery to the oral mucosa. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 112, 110917. [Google Scholar] [CrossRef]
  67. Liao, Y.C.; Hsu, L.F.; Hsieh, L.Y.; Luo, Y.Y. Effectiveness of green tea mouthwash for improving oral health status in oral cancer patients: A single-blind randomized controlled trial. Int. J. Nurs. Stud. 2021, 121, 103985, (clinicaltrials.gov identifier NCT04615780). [Google Scholar] [CrossRef]
  68. Freires, I.A.; Rosalen, P.L. How Natural Product Research has Contributed to Oral Care Product Development? A Critical View. Pharm. Res. 2016, 33, 1311–1317. [Google Scholar] [CrossRef]
  69. Yen, S.H.; Wang, L.W.; Lin, Y.H.; Jen, Y.M.; Chung, Y.L. Phenylbutyrate mouthwash mitigates oral mucositis during radiotherapy or chemoradiotherapy in patients with head-and-neck cancer. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, 1463–1470. [Google Scholar] [CrossRef]
  70. Nigro, O.; Tuzi, A.; Tartaro, T.; Giaquinto, A.; Vallini, I.; Pinotti, G. Biological effects of verbascoside and its anti-inflammatory activity on oral mucositis: A review of the literature. Anticancer Drugs 2020, 31, 1–5. [Google Scholar] [CrossRef]
  71. Baysal, E.; Sari, D.; Vural, F.; Cagirgan, S.; Saydam, G.; Tobu, M.; Sahin, F.; Soyer, N.; Gediz, F.; Acarlar, C.; et al. The Effect of Cryotherapy on the Prevention of Oral Mucositis and on the Oral pH Value in Multiple Myeloma Patients Undergoing Autologous Stem Cell Transplantation. Semin. Oncol. Nurs. 2021, 37, 151146. [Google Scholar] [CrossRef] [PubMed]
  72. Askarifar, M.; Lakdizaji, S.; Ramzi, M.; Rahmani, A.; Jabbarzadeh, F. The Effects of Oral Cryotherapy on Chemotherapy-Induced Oral Mucositis in Patients Undergoing Autologous Transplantation of Blood Stem Cells: A Clinical Trial. Iran. Red Crescent Med. J. 2016, 18, e24775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Migliorati, C.A.; Oberle-Edwards, L.; Schubert, M. The role of alternative and natural agents, cryotherapy, and/or laser for management of alimentary mucositis. Support. Care Cancer 2006, 14, 533–540. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, X.; Sun, D.; Qin, N.; Liu, M.; Zhang, J.; Li, X. Comparative prevention potential of 10 mouthwashes on intolerable oral mucositis in cancer patients: A Bayesian network analysis. Oral Oncol. 2020, 107, 104751. [Google Scholar] [CrossRef]
  75. Karavana Hizarcioglu, S.Y.; Sezer, B.; Guneri, P.; Veral, A.; Boyacioglu, H.; Ertan, G.; Epstein, J.B. Efficacy of topical benzydamine hydrochloride gel on oral mucosal ulcers: An in vivo animal study. Int. J. Oral Maxillofac. Surg. 2011, 40, 973–978. [Google Scholar] [CrossRef]
  76. El-Salamouni, N.S.; Hanafy, A.S. Hyaluronic-benzydamine oromucosal films outperform conventional mouth rinse in ulcer healing. J. Drug Deliv. Sci. Technol. 2021, 65, 102690. [Google Scholar] [CrossRef]
  77. Yang, C.; Gong, G.; Jin, E.; Han, X.; Zhuo, Y.; Yang, S.; Song, B.; Zhang, Y.; Piao, C. Topical application of honey in the management of chemo/radiotherapy-induced oral mucositis: A systematic review and network meta-analysis. Int. J. Nurs. Stud. 2019, 89, 80–87. [Google Scholar] [CrossRef]
  78. Saunders, D.P.; Epstein, J.B.; Elad, S.; Allemano, J.; Bossi, P.; van de Wetering, M.D.; Rao, N.G.; Potting, C.; Cheng, K.K.; Freidank, A.; et al. Systematic review of antimicrobials, mucosal coating agents, anesthetics, and analgesics for the management of oral mucositis in cancer patients. Support. Care Cancer 2013, 21, 3191–3207. [Google Scholar] [CrossRef] [Green Version]
  79. Nielsen, B.N.; Aagaard, G.; Henneberg, S.W.; Schmiegelow, K.; Hansen, S.H.; Romsing, J. Topical morphine for oral mucositis in children: Dose finding and absorption. J. Pain Symptom Manag. 2012, 44, 117–123. [Google Scholar] [CrossRef]
  80. Sanz, R.; Calpena, A.C.; Mallandrich, M.; Gimeno, A.; Halbaut, L.; Clares, B. Development of a buccal doxepin platform for pain in oral mucositis derived from head and neck cancer treatment. Eur. J. Pharm. Biopharm. 2017, 117, 203–211. [Google Scholar] [CrossRef]
  81. Chung, M.K.; Wang, S.; Oh, S.L.; Kim, Y.S. Acute and Chronic Pain from Facial Skin and Oral Mucosa: Unique Neurobiology and Challenging Treatment. Int. J. Mol. Sci. 2021, 22, 5810. [Google Scholar] [CrossRef] [PubMed]
  82. Miranzadeh, S.; Adib-Hajbaghery, M.; Soleymanpoor, L.; Ehsani, M. Effect of adding the herb Achillea millefolium on mouthwash on chemotherapy induced oral mucositis in cancer patients: A double-blind randomized controlled trial. Eur. J. Oncol. Nurs. 2015, 19, 207–213. [Google Scholar] [CrossRef] [PubMed]
  83. Santos, H.T.C.; Coimbra, M.C.; Meri Junior, A.E.; Gomes, A.J.P.S. Effectiveness of topically applied chamomile in the treatment of oral mucositis: A literature review. Res. Soc. Dev. 2021, 10, e433101422081. [Google Scholar] [CrossRef]
  84. Honda, H.; Onda, T.; Hayashi, K.; Shibahara, T.; Takano, M. Comparison of topical agents that are effective against oral mucositis associated with chemotherapy using a rat anticancer agent-induced oral mucositis model. J. Oral Maxillofac. Surg. Med. Pathol. 2021; in press. [Google Scholar] [CrossRef]
  85. da Silva, V.C.R.; da Motta Silveira, F.M.; Barbosa Monteiro, M.G.; da Cruz, M.M.D.; Caldas Junior, A.F.; Pina Godoy, G. Photodynamic therapy for treatment of oral mucositis: Pilot study with pediatric patients undergoing chemotherapy. Photodiagn. Photodyn. Ther. 2018, 21, 115–120. [Google Scholar] [CrossRef] [PubMed]
  86. Simoes, A.; Benites, B.M.; Benassi, C.; Torres-Schroter, G.; de Castro, J.R.; Campos, L. Antimicrobial photodynamic therapy on treatment of infected radiation-induced oral mucositis: Report of two cases. Photodiagn. Photodyn. Ther. 2017, 20, 18–20. [Google Scholar] [CrossRef] [PubMed]
  87. de Oliveira, A.B.; Ferrisse, T.M.; Basso, F.G.; Fontana, C.R.; Giro, E.M.A.; Brighenti, F.L. A systematic review and meta-analysis of the effect of photodynamic therapy for the treatment of oral mucositis. Photodiagn. Photodyn. Ther. 2021, 34, 102316. [Google Scholar] [CrossRef]
  88. Yarom, N.; Hovan, A.; Bossi, P.; Ariyawardana, A.; Jensen, S.B.; Gobbo, M.; Saca-Hazboun, H.; Kandwal, A.; Majorana, A.; Ottaviani, G.; et al. Systematic review of natural and miscellaneous agents, for the management of oral mucositis in cancer patients and clinical practice guidelines—Part 2: Honey, herbal compounds, saliva stimulants, probiotics, and miscellaneous agents. Support. Care Cancer 2020, 28, 2457–2472. [Google Scholar] [CrossRef]
  89. Nagi, R.; Patil, D.J.; Rakesh, N.; Jain, S.; Sahu, S. Natural agents in the management of oral mucositis in cancer patients-systematic review. J. Oral Biol. Craniofac. Res. 2018, 8, 245–254. [Google Scholar] [CrossRef]
  90. Aghamohamamdi, A.; Hosseinimehr, S.J. Natural Products for Management of Oral Mucositis Induced by Radiotherapy and Chemotherapy. Integr. Cancer Ther. 2016, 15, 60–68. [Google Scholar] [CrossRef]
  91. Khanal, B.; Baliga, M.; Uppal, N. Effect of topical honey on limitation of radiation-induced oral mucositis: An intervention study. Int. J. Oral Maxillofac. Surg. 2010, 39, 1181–1185. [Google Scholar] [CrossRef] [PubMed]
  92. Amanat, A.; Ahmed, A.; Kazmi, A.; Aziz, B. The Effect of Honey on Radiation-induced Oral Mucositis in Head and Neck Cancer Patients. Indian J. Palliat. Care 2017, 23, 317–320. [Google Scholar] [CrossRef] [PubMed]
  93. Lusby, P.E.; Coombes, A.; Wilkinson, J.M. Honey: A potent agent for wound healing? J. Wound Ostomy Cont. Nurs. 2002, 29, 295–300. [Google Scholar] [CrossRef]
  94. Molan, P.C. Re-introducing honey in the management of wounds and ulcers-theory and practice. Ostomy Wound Manag. 2002, 48, 28–40. [Google Scholar]
  95. Motallebnejad, M.; Akram, S.; Moghadamnia, A.; Moulana, Z.; Omidi, S. The effect of topical application of pure honey on radiation-induced mucositis: A randomized clinical trial. J. Contemp. Dent. Pract. 2008, 9, 40–47. [Google Scholar] [CrossRef] [Green Version]
  96. Al-Waili, N.S. An alternative treatment for pityriasis versicolor, tinea cruris, tinea corporis and tinea faciei with topical application of honey, olive oil and beeswax mixture: An open pilot study. Complement. Ther. Med. 2004, 12, 45–47. [Google Scholar] [CrossRef]
  97. Khanjani Pour-Fard-Pachekenari, A.; Rahmani, A.; Ghahramanian, A.; Asghari Jafarabadi, M.; Onyeka, T.C.; Davoodi, A. The effect of an oral care protocol and honey mouthwash on mucositis in acute myeloid leukemia patients undergoing chemotherapy: A single-blind clinical trial. Clin. Oral Investig. 2019, 23, 1811–1821, (WHO Trial registration IRCT2015121419919N7). [Google Scholar] [CrossRef]
  98. Raeessi, M.A.; Raeessi, N.; Panahi, Y.; Gharaie, H.; Davoudi, S.M.; Saadat, A.; Karimi Zarchi, A.A.; Raeessi, F.; Ahmadi, S.M.; Jalalian, H. “Coffee plus honey” versus “topical steroid” in the treatment of chemotherapy-induced oral mucositis: A randomised controlled trial. BMC Complement. Altern. Med. 2014, 14, 293, (Iranian Registry of Clinical Trials IRCT: 201104074737N3). [Google Scholar] [CrossRef] [Green Version]
  99. Davis, J.K.; Green, J.M. Caffeine and anaerobic performance: Ergogenic value and mechanisms of action. Sports Med. 2009, 39, 813–832. [Google Scholar] [CrossRef]
  100. Miłek, M.; Młodecki, Ł.; Dżugan, M. Caffeine content and antioxidant activity of various brews of specialty grade coffee. Acta Sci. Pol. Technol. Aliment. 2021, 20, 179–188. [Google Scholar]
  101. Barcelos, R.P.; Lima, F.D.; Carvalho, N.R.; Bresciani, G.; Royes, L.F. Caffeine effects on systemic metabolism, oxidative-inflammatory pathways, and exercise performance. Nutr. Res. 2020, 80, 1–17. [Google Scholar] [CrossRef] [PubMed]
  102. Sforcin, J.M. Biological Properties and Therapeutic Applications of Propolis. Phytother. Res. 2016, 30, 894–905. [Google Scholar] [CrossRef] [PubMed]
  103. Melliou, E.; Chinou, I. Chemical analysis and antimicrobial activity of Greek propolis. Planta Med. 2004, 70, 515–519. [Google Scholar] [CrossRef] [PubMed]
  104. Huang, X.Y.; Guo, X.L.; Luo, H.L.; Fang, X.W.; Zhu, T.G.; Zhang, X.L.; Chen, H.W.; Luo, L.P. Fast Differential Analysis of Propolis Using Surface Desorption Atmospheric Pressure Chemical Ionization Mass Spectrometry. Int. J. Anal. Chem. 2015, 2015, 176475. [Google Scholar] [CrossRef] [Green Version]
  105. Franco, T. Chemical composition of propolis: Vitamins and aminoacids. Rev. Bras. Farmacogn. 1985, 1, 12–19. [Google Scholar]
  106. Kamburoğlu, K.; Özen, T. Analgesic effect of Anatolian propolis in mice. Agri 2011, 23, 47–50. [Google Scholar]
  107. Montero, J.C.; Mori, G.G. Assessment of ion diffusion from a calcium hydroxide-propolis paste through dentin. Braz. Oral Res. 2012, 26, 318–322. [Google Scholar] [CrossRef]
  108. Ozan, F.; Sümer, Z.; Polat, Z.A.; Er, K.; Ozan, U.; Deger, O. Effect of mouthrinse containing propolis on oral microorganisms and human gingival fibroblasts. Eur. J. Dent. 2007, 1, 195–201. [Google Scholar]
  109. Cavalcante, D.R.; Oliveira, P.S.; Góis, S.M.; Soares, A.F.; Cardoso, J.C.; Padilha, F.F.; Albuquerque, R.L., Jr. Effect of green propolis on oral epithelial dysplasia in rats. Braz. J. Otorhinolaryngol. 2011, 77, 278–284. [Google Scholar] [CrossRef] [Green Version]
  110. Mirzoeva, O.K.; Calder, P.C. The effect of propolis and its components on eicosanoid production during the inflammatory response. Prostaglandins Leukot. Essent. Fat. Acids 1996, 55, 441–449. [Google Scholar] [CrossRef]
  111. AkhavanKarbassi, M.H.; Yazdi, M.F.; Ahadian, H.; SadrAbad, M.J. Randomized DoubleBlind Placebo Controlled Trial of Propolis for Oral Mucositis in Patients Receiving Chemotherapy for Head and Neck Cancer. Asian Pac. J. Cancer Prev. 2016, 17, 3611–3614. [Google Scholar] [PubMed]
  112. Pavel, C.; Mărghitaş, A.L.; Bobis, O.; Dezmirean, D.; Şapcaliu, A.; Radoi, I.; Mădaş, M. Biological Activities of Royal Jelly-Review. Lucr. Stiintifice 2011, 44, 108–118. [Google Scholar]
  113. Nagai, T.; Inoue, R. Preparation and the functional properties of water extract and alkaline extract of royal jelly. Food Chem. 2004, 84, 181–186. [Google Scholar] [CrossRef]
  114. Nagai, T.; Inoue, R.; Suzuki, N.; Nagashima, T. Antioxidant properties of enzymatic hydrolysates from royal jelly. J. Med. Food 2006, 9, 363–367. [Google Scholar] [CrossRef] [Green Version]
  115. Izuta, H.; Chikaraishi, Y.; Shimazawa, M.; Mishima, S.; Hara, H. 10-Hydroxy-2-decenoic acid, a major fatty acid from royal jelly, inhibits VEGF-induced angiogenesis in human umbilical vein endothelial cells. Evid. Based Complement. Altern. Med. 2009, 6, 489–494. [Google Scholar] [CrossRef]
  116. Šimúth, J.; Bíliková, K.; Kováčová, E.; Kuzmová, Z.; Schroder, W. Immunochemical Approach to Detection of Adulteration in Honey:  Physiologically Active Royal Jelly Protein Stimulating TNF-α Release is a Regular Component of Honey. J. Agric. Food Chem. 2004, 52, 2154–2158. [Google Scholar] [CrossRef]
  117. Matsui, T.; Yukiyoshi, A.; Doi, S.; Sugimoto, H.; Yamada, H.; Matsumoto, K. Gastrointestinal enzyme production of bioactive peptides from royal jelly protein and their antihypertensive ability in SHR. J. Nutr. Biochem. 2002, 13, 80–86. [Google Scholar] [CrossRef]
  118. Fujiwara, S.; Imai, J.; Fujiwara, M.; Yaeshima, T.; Kawashima, T.; Kobayashi, K. A potent antibacterial protein in royal jelly. Purification and determination of the primary structure of royalisin. J. Biol. Chem. 1990, 265, 11333–11337. [Google Scholar] [CrossRef]
  119. Kohno, K.; Okamoto, I.; Sano, O.; Arai, N.; Iwaki, K.; Ikeda, M.; Kurimoto, M. Royal jelly inhibits the production of proinflammatory cytokines by activated macrophages. Biosci. Biotechnol. Biochem. 2004, 68, 138–145. [Google Scholar] [CrossRef] [Green Version]
  120. Fujii, A.; Kobayashi, S.; Kuboyama, N.; Furukawa, Y.; Kaneko, Y.; Ishihama, S.; Yamamoto, H.; Tamura, T. Augmentation of wound healing by royal jelly (RJ) in streptozotocin-diabetic rats. Jpn. J. Pharmacol. 1990, 53, 331–337. [Google Scholar] [CrossRef] [Green Version]
  121. Xue, X.; Wu, L.; Wang, K. Chemical Composition of Royal Jelly. In Bee Products-Chemical and Biological Properties; Alvarez-Suarez, J.M., Ed.; Springer International Publishing: Cham, Switzerland, 2017; pp. 181–190. [Google Scholar]
  122. Lin, Y.; Shao, Q.; Zhang, M.; Lu, C.; Fleming, J.; Su, S. Royal jelly-derived proteins enhance proliferation and migration of human epidermal keratinocytes in an in vitro scratch wound model. BMC Complement. Altern. Med. 2019, 19, 175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Inoue, S.; Koya-Miyata, S.; Ushio, S.; Iwaki, K.; Ikeda, M.; Kurimoto, M. Royal Jelly prolongs the life span of C3H/HeJ mice: Correlation with reduced DNA damage. Exp. Gerontol. 2003, 38, 965–969. [Google Scholar] [CrossRef]
  124. Suemaru, K.; Cui, R.; Li, B.; Watanabe, S.; Okihara, K.; Hashimoto, K.; Yamada, H.; Araki, H. Topical application of royal jelly has a healing effect for 5-fluorouracil-induced experimental oral mucositis in hamsters. Methods Find. Exp. Clin. Pharmacol. 2008, 30, 103–106. [Google Scholar] [CrossRef] [PubMed]
  125. Watanabe, S.; Suemaru, K.; Takechi, K.; Kaji, H.; Imai, K.; Araki, H. Oral mucosal adhesive films containing royal jelly accelerate recovery from 5-fluorouracil-induced oral mucositis. J. Pharmacol. Sci. 2013, 121, 110–118. [Google Scholar] [CrossRef] [Green Version]
  126. Erdem, O.; Güngörmüş, Z. The effect of royal jelly on oral mucositis in patients undergoing radiotherapy and chemotherapy. Holist. Nurs. Pract. 2014, 28, 242–246. [Google Scholar] [CrossRef]
  127. Yamauchi, K.; Kogashiwa, Y.; Moro, Y.; Kohno, N. The effect of topical application of royal jelly on chemoradiotherapy-induced mucositis in head and neck cancer: A preliminary study. Int. J. Otolaryngol. 2014, 2014, 974967. [Google Scholar] [CrossRef] [Green Version]
  128. Gomes, M.S.; Lins, R.D.A.U.; Langassner, S.M.Z.; da Silveira, E.J.D.; de Carvalho, T.G.; de Sousa Lopes, M.L.D.; de Souza Araujo, L.; de Medeiros, C.A.C.X.; de Carvalho Leitão, R.F.; Guerra, G.C.B.; et al. Anti-inflammatory and antioxidant activity of hydroethanolic extract of Spondias mombin leaf in an oral mucositis experimental model. Arch. Oral Biol. 2020, 111, 104664. [Google Scholar] [CrossRef]
  129. Cabral, B.; Siqueira, E.M.S.; Bitencourt, M.A.O.; Lima, M.C.J.S.; Lima, A.K.; Ortmann, C.F.; Chaves, V.C.; Fernandes-Pedrosa, M.F.; Rocha, H.A.O.; Scortecci, K.C.; et al. Phytochemical study and anti-inflammatory and antioxidant potential of Spondias mombin leaves. Rev. Bras. Farmacogn. 2016, 26, 304–311. [Google Scholar] [CrossRef] [Green Version]
  130. Nworu, C.S.; Akah, P.A.; Okoye, F.B.C.; Toukam, D.K.; Udeh, J.; Esimone, C.O. The leaf extract of Spondias mombin L. displays an anti-inflammatory effect and suppresses inducible formation of tumor necrosis factor-α and nitric oxide (NO). J. Immunotoxicol. 2011, 8, 10–16. [Google Scholar] [CrossRef]
  131. Khan, N.; Mukhtar, H. Tea polyphenols for health promotion. Life Sci. 2007, 81, 519–533. [Google Scholar] [CrossRef] [Green Version]
  132. Lin, J.K.; Liang, Y.C.; Lin-Shiau, S.Y. Cancer chemoprevention by tea polyphenols through mitotic signal transduction blockade. Biochem. Pharmacol. 1999, 58, 911–915. [Google Scholar] [CrossRef]
  133. Moyers, S.B.; Kumar, N.B. Green Tea Polyphenols and Cancer Chemoprevention: Multiple Mechanisms and Endpoints for Phase II Trials. Nutr. Rev. 2004, 62, 204–211. [Google Scholar] [CrossRef] [PubMed]
  134. Cardoso, R.R.; Neto, R.O.; dos Santos D’Almeida, C.T.; do Nascimento, T.P.; Pressete, C.G.; Azevedo, L.; Martino, H.S.D.; Cameron, L.C.; Ferreira, M.S.L.; de Barros, F.A.R. Kombuchas from green and black teas have different phenolic profile, which impacts their antioxidant capacities, antibacterial and antiproliferative activities. Food Res. Int. 2020, 128, 108782. [Google Scholar] [CrossRef] [PubMed]
  135. Steinmann, J.; Buer, J.; Pietschmann, T.; Steinmann, E. Anti-infective properties of epigallocatechin-3-gallate (EGCG), a component of green tea. Br. J. Pharmacol. 2013, 168, 1059–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Musial, C.; Kuban-Jankowska, A.; Gorska-Ponikowska, M. Beneficial Properties of Green Tea Catechins. Int. J. Mol. Sci. 2020, 21, 1744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Okabe, S.; Ochiai, Y.; Aida, M.; Park, K.; Kim, S.J.; Nomura, T.; Suganuma, M.; Fujiki, H. Mechanistic aspects of green tea as a cancer preventive: Effect of components on human stomach cancer cell lines. Jpn. J. Cancer Res. 1999, 90, 733–739. [Google Scholar] [CrossRef]
  138. Pervin, M.; Unno, K.; Takagaki, A.; Isemura, M.; Nakamura, Y. Function of green tea catechins in the brain: Epigallocatechin gallate and its metabolites. Int. J. Mol. Sci. 2019, 20, 3630. [Google Scholar] [CrossRef] [Green Version]
  139. Carulli, G.; Rocco, M.; Panichi, A.; Chios, C.F.; Ciurli, E.; Mannucci, C.; Sordi, E.; Caracciolo, F.; Papineschi, F.; Benedetti, E.; et al. Treatment of oral mucositis in hematologic patients undergoing autologous or allogeneic transplantation of peripheral blood stem cells: A prospective, randomized study with a mouthwash containing camelia sinensis leaf extract. Hematol. Rep. 2013, 5, 21–25. [Google Scholar] [CrossRef] [Green Version]
  140. Cabrera-Jaime, S.; Martinez, C.; Ferro-Garcia, T.; Giner-Boya, P.; Icart-Isern, T.; Estrada-Masllorens, J.M.; Fernandez-Ortega, P. Efficacy of Plantago major, chlorhexidine 0.12% and sodium bicarbonate 5% solution in the treatment of oral mucositis in cancer patients with solid tumour: A feasibility randomised triple-blind phase III clinical trial. Eur. J. Oncol. Nurs. 2018, 32, 40–47. [Google Scholar] [CrossRef]
  141. Soltani, G.M.; Hemati, S.; Sarvizadeh, M.; Kamalinejad, M.; Tafazoli, V.; Latifi, S.A. Efficacy of the Plantago major L. syrup on radiation induced oral mucositis in head and neck cancer patients: A randomized, double blind, placebo-controlled clinical trial. Complement. Ther. Med. 2020, 51, 102397, (Iranian Registry of Clinical Trials IRCT: 20190312043027N1). [Google Scholar] [CrossRef]
  142. Yazdian, M.A.; Khodadoost, M.; Gheisari, M.; Kamalinejad, M.; Ehsani, A.; Barikbin, B. A Hypothesis on the Possible Potential of Plantago major in the Treatment of Urticaria. Galen Med. J. 2014, 3, 123–126. [Google Scholar] [CrossRef]
  143. Ringbom, T.; Segura, L.; Noreen, Y.; Perera, P.; Bohlin, L. Ursolic acid from Plantago major, a selective inhibitor of cyclooxygenase-2 catalyzed prostaglandin biosynthesis. J. Nat. Prod. 1998, 61, 1212–1215. [Google Scholar] [CrossRef] [PubMed]
  144. Stenholm, A.; Göransson, U.; Bohlin, L. Bioassay-guided supercritical fluid extraction of cyclooxygenase-2 inhibiting substances in Plantago major L. Phytochem. Anal. 2013, 24, 176–183. [Google Scholar] [CrossRef] [PubMed]
  145. Pourmorad, F.; Hosseinimehr, S.J.; Shahabimajd, N. Antioxidant activity of phenol and flavonoid contents of some Iranian medicinal plants. Afr. J. Adv. Biotechnol. 2005, 5, 1684–5315. [Google Scholar]
  146. Samuelsen, A.B.; Paulsen, B.S.; Wold, J.K.; Otsuka, H.; Yamada, H.; Espevik, T. Isolation and partial characterization of biologically active polysaccharides from Plantago major L. Phytother. Res. 1995, 9, 211–218. [Google Scholar] [CrossRef]
  147. Baechler, B.J.; Nita, F.; Jones, L.; Frestedt, J.L. A novel liquid multi-phytonutrient supplement demonstrates DNA-protective effects. Plant Foods Hum. Nutr. 2009, 64, 81–85. [Google Scholar] [CrossRef] [Green Version]
  148. Langmead, L.; Makins, R.J.; Rampton, D.S. Anti-inflammatory effects of Aloe vera gel in human colorectal mucosa in vitro. Aliment. Pharmacol. Ther. 2004, 19, 521–527. [Google Scholar] [CrossRef]
  149. Heggers, J.; Pineless, G.; Robson, M. Dermaide Aloe vera gel-comparison of the anti-microbial effects. J. Am. Med. Inform. Assoc. 1979, 41, 293–294. [Google Scholar]
  150. Yagi, A.; Kabash, A.; Okamura, N.; Haraguchi, H.; Moustafa, S.M.; Khalifa, T.I. Antioxidant, free radical scavenging and anti-inflammatory effects of aloesin derivatives in Aloe vera. Planta Med. 2002, 68, 957–960. [Google Scholar] [CrossRef]
  151. Hu, Y.; Xu, J.; Hu, Q. Evaluation of antioxidant potential of Aloe vera (Aloe barbadensis miller) extracts. J. Agric. Food Chem. 2003, 51, 7788–7791. [Google Scholar] [CrossRef]
  152. Heggie, S.; Bryant, G.P.; Tripcony, L.; Keller, J.; Rose, P.; Glendenning, M.; Heath, J. A Phase III study on the efficacy of topical Aloe vera gel on irradiated breast tissue. Cancer Nurs. 2002, 25, 442–451. [Google Scholar] [CrossRef] [PubMed]
  153. Davis, R.H.; Leitner, M.G.; Russo, J.M.; Byrne, M.E. Wound healing. Oral and topical activity of Aloe vera. J. Am. Podiatr. Med. Assoc. 1989, 79, 559–562. [Google Scholar] [PubMed]
  154. Davis, R.H.; Leitner, M.G.; Russo, J.M.; Byrne, M.E. Anti-inflammatory activity of Aloe vera against a spectrum of irritants. J. Am. Podiatr. Med. Assoc. 1989, 79, 263–276. [Google Scholar] [CrossRef] [PubMed]
  155. Meadows, T.P. Aloe as a humectant in new skin preparations. Cosmet. Toilet. 1980, 95, 51–56. [Google Scholar]
  156. Davis, R.H.; Donato, J.J.; Hartman, G.M.; Haas, R.C. Anti-inflammatory and wound healing activity of a growth substance in Aloe vera. J. Am. Podiatr. Med. Assoc. 1994, 84, 77–81. [Google Scholar] [PubMed]
  157. Vázquez, B.; Avila, G.; Segura, D.; Escalante, B. Anti-inflammatory activity of extracts from Aloe vera gel. J. Ethnopharmacol. 1996, 55, 69–75. [Google Scholar] [CrossRef]
  158. Wei, A.; Shibamoto, T. Antioxidant/lipoxygenase inhibitory activities and chemical compositions of selected essential oils. J. Agric. Food Chem. 2010, 58, 7218–7225. [Google Scholar] [CrossRef]
  159. Su, C.K.; Mehta, V.; Ravikumar, L.; Shah, R.; Pinto, H.; Halpern, J.; Koong, A.; Goffinet, D.; Le, Q.-T. Phase II double-blind randomized study comparing oral Aloe vera versus placebo to prevent radiation-related mucositis in patients with head-and-neck neoplasms. Int. J. Radiat. Oncol. Biol. Phys. 2004, 60, 171–177. [Google Scholar] [CrossRef]
  160. Mansouri, P.; Haghighi, M.; Beheshtipour, N.; Ramzi, M. The effect of Aloe vera solution on Chemotherapy-induced stomatitis in clients with lymphoma and leukemia: A randomized controlled Clinical Trial. Int. J. Community Based Nurs. Midwifery 2016, 4, 119–126, (Iranian Registry of Clinical Trials: 2014092819318N1). [Google Scholar]
  161. Karbasizade, S.; Ghorbani, F.; Ghasemi Darestani, N.; Mansouri-Tehrani, M.M.; Kazemi, A.H. Comparison of therapeutic effects of statins and Aloe vera mouthwash on chemotherapy induced oral mucositis. Int. J. Physiol. Pathophysiol. Pharmacol. 2021, 13, 110–116. [Google Scholar]
  162. Alkhouli, M.; Laflouf, M.; Alhaddad, M. Efficacy of Aloe-vera use for prevention of chemotherapy-induced oral mucositis in children with acute lymphoblastic leukemia: A randomized controlled clinical trial. Compr. Child Adolesc. Nurs. 2021, 44, 49–62, (Australian New Zealand Clinical Trials Registry: 12618001931268). [Google Scholar] [CrossRef] [PubMed]
  163. Sahebjamee, M.; Mansourian, A.; Hajimirzamohammad, M.; Zadeh, M.T.; Bekhradi, R.; Kazemian, A.; Manifar, S.; Ashnagar, S.; Doroudgar, K. Comparative efficacy of Aloe vera and benzydamine mouthwashes on radiation-induced oral mucositis: A triple-blind, randomised, controlled clinical trial. Oral Health Prev. Dent. 2015, 13, 309–315, (Iranian Registry of Clinical Trials: 2012072410377N1). [Google Scholar] [PubMed]
  164. Kocaadam, B.; Şanlier, N. Curcumin, an active component of turmeric (Curcuma longa), and its effects on health. Crit. Rev. Food Sci. Nutr. 2017, 57, 2889–2895. [Google Scholar] [CrossRef] [PubMed]
  165. Gupta, S.C.; Sung, B.; Kim, J.H.; Prasad, S.; Li, S.; Aggarwal, B.B. Multitargeting by turmeric, the golden spice: From kitchen to clinic. Mol. Nutr. Food Res. 2013, 57, 1510–1528. [Google Scholar] [CrossRef]
  166. Deogade, S.C.; Ghate, S. Curcumin: Therapeutic applications in systemic and oral health. Int. J. Biol. Pharm. Res. 2015, 6, 281–290. [Google Scholar]
  167. Li, S. Chemical composition and product quality control of turmeric (Curcuma longa L.). Pharm. Crops 2011, 5, 28–54. [Google Scholar] [CrossRef]
  168. Shehzad, A.; Lee, Y. Curcumin: Multiple molecular targets mediate multiple pharmacological actions: A review. Drugs Future 2010, 35, 113. [Google Scholar] [CrossRef]
  169. Srinivasan, M. Effect of curcumin on blood sugar as seen in a diabetic subject. Indian J. Med. Sci. 1972, 26, 269–270. [Google Scholar]
  170. Srimal, R.C.; Dhawan, B.N. Pharmacology of diferuloyl methane (curcumin), a non-steroidal anti-inflammatory agent. J. Pharm. Pharmacol. 1973, 25, 447–452. [Google Scholar] [CrossRef]
  171. Sharma, O.P. Antioxidant activity of curcumin and related compounds. Biochem. Pharmacol. 1976, 25, 1811–1812. [Google Scholar] [CrossRef]
  172. Aggarwal, B.B.; Harikumar, K.B. Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. Int. J. Biochem. Cell Biol. 2009, 41, 40–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Gandhy, S.U.; Kim, K.; Larsen, L.; Rosengren, R.J.; Safe, S. Curcumin and synthetic analogs induce reactive oxygen species and decreases specificity protein (Sp) transcription factors by targeting microRNAs. BMC Cancer 2012, 12, 564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Kim, S.G.; Veena, M.S.; Basak, S.K.; Han, E.; Tajima, T.; Gjertson, D.W.; Starr, J.; Eidelman, O.; Pollard, H.B.; Srivastava, M.; et al. Curcumin treatment suppresses IKKβ kinase activity of salivary cells of patients with head and neck cancer: A pilot study. Clin. Cancer Res. 2011, 17, 5953–5961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Das, L.; Vinayak, M. Curcumin attenuates carcinogenesis by down regulating proinflammatory cytokine interleukin-1 (IL-1α and IL-1β) via modulation of AP-1 and NF-IL6 in lymphoma bearing mice. Int. Immunopharmacol. 2014, 20, 141–147. [Google Scholar] [CrossRef] [PubMed]
  176. Rujirachotiwat, A.; Suttamanatwong, S. Curcumin upregulates transforming growth factor-β1, its receptors, and vascular endothelial growth factor expressions in an in vitro human gingival fibroblast wound healing model. BMC Oral Health 2021, 21, 535. [Google Scholar] [CrossRef]
  177. Mani, H.; Sidhu, G.S.; Kumari, R.; Gaddipati, J.P.; Seth, P.; Maheshwari, R.K. Curcumin differentially regulates TGF-beta1, its receptors and nitric oxide synthase during impaired wound healing. Biofactors 2002, 16, 29–43. [Google Scholar] [CrossRef]
  178. Panchatcharam, M.; Miriyala, S.; Gayathri, V.S.; Suguna, L. Curcumin improves wound healing by modulating collagen and decreasing reactive oxygen species. Mol. Cell. Biochem. 2006, 290, 87–96. [Google Scholar] [CrossRef]
  179. Elad, S.; Meidan, I.; Sellam, G.; Simaan, S.; Zeevi, I.; Waldman, E.; Weintraub, M.; Revel-Vilk, S. Topical curcumin for the prevention of oral mucositis in pediatric patients: Case series. Altern. Ther. Health Med. 2013, 19, 21–24. [Google Scholar]
  180. Patil, K.; Guledgud, M.V.; Kulkarni, P.K.; Keshari, D.; Tayal, S. Use of Curcumin mouthrinse in radio-chemotherapy induced Oral Mucositis patients: A pilot study. J. Clin. Diagn. Res. 2015, 9, ZC59–ZC62. [Google Scholar] [CrossRef]
  181. Delavarian, Z.; Pakfetrat, A.; Ghazi, A.; Jaafari, M.R.; Homaei Shandiz, F.; Dalirsani, Z.; Mohammadpour, A.H.; Rahimi, H.R. Oral administration of nanomicelle curcumin in the prevention of radiotherapy-induced mucositis in head and neck cancers. Spec. Care Dent. 2019, 39, 166–172. [Google Scholar] [CrossRef]
  182. Ahmed, K.M. The effect of olive leaf extract in decreasing the expression of two pro-inflammatory cytokines in patients receiving chemotherapy for cancer. A randomized clinical trial. Saudi Dent. J. 2013, 25, 141–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Lee-Huang, S.; Huang, P.L.; Zhang, D.; Lee, J.W.; Bao, J.; Sun, Y.; Chang, Y.T.; Zhang, J.; Huang, P.L. Discovery of small-molecule HIV-1 fusion and integrase inhibitors oleuropein and hydroxytyrosol: Part I. fusion inhibition. Biochem. Biophys. Res. Commun. 2007, 354, 872–878. [Google Scholar] [CrossRef] [PubMed]
  184. Markin, D.; Duek, L.; Berdicevsky, I. In vitro antimicrobial activity of olive leaves. Mycoses 2003, 46, 132–136. [Google Scholar] [CrossRef] [PubMed]
  185. Coni, E.; Di Benedetto, R.; Di Pasquale, M.; Masella, R.; Modesti, D.; Mattei, R.; Carlini, E.A. Protective effect of oleuropein, an olive oil biophenol, on low density lipoprotein oxidizability in rabbits. Lipids 2000, 35, 45–54. [Google Scholar] [CrossRef] [PubMed]
  186. Singh, I.; Mok, M.; Christensen, A.M.; Turner, A.H.; Hawley, J.A. The effects of polyphenols in olive leaves on platelet function. Nutr. Metab. Cardiovasc. Dis. 2008, 18, 127–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Campolo, M.; Di Paola, R.; Impellizzeri, D.; Crupi, R.; Morittu, V.M.; Procopio, A.; Perri, E.; Britti, D.; Peli, A.; Esposito, E.; et al. Effects of a polyphenol present in olive oil, oleuropein aglycone, in a murine model of intestinal ischemia/reperfusion injury. J. Leukoc. Biol. 2013, 93, 277–287. [Google Scholar] [CrossRef] [PubMed]
  188. Silva, S.; Gomes, L.; Leitão, F.; Coelho, A.; Boas, L.V. Phenolic compounds and antioxidant activity of Olea europaea L. fruits and leaves. Food Sci. Technol. Int. 2006, 12, 385–395. [Google Scholar] [CrossRef]
  189. Koca, U.; Süntar, I.; Akkol, E.K.; Yilmazer, D.; Alper, M. Wound repair potential of Olea europaea L. leaf extracts revealed by in vivo experimental models and comparative evaluation of the extracts’ antioxidant activity. J. Med. Food 2011, 14, 140–146. [Google Scholar] [CrossRef]
  190. Mehraein, F.; Sarbishegi, M.; Aslani, A. Evaluation of effect of oleuropein on skin wound healing in aged male BALB/c mice. Cell J. 2014, 16, 25–30. [Google Scholar]
  191. Ahmed, K. Olive leaf extract as a new topical management for oral mucositis following chemotherapy: A microbiological examination, experimental animal study and clinical trial. Pharm. Anal. Acta 2013, 4, 4–9. [Google Scholar]
  192. Kondo, K.; Shiba, M.; Nakamura, R.; Morota, T.; Shoyama, Y. Constituent properties of licorices derived from Glycyrrhiza uralensis, G. glabra, or G. inflata identified by genetic information. Biol. Pharm. Bull. 2007, 30, 1271–1277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  193. Gupta, V.K.; Fatima, A.; Faridi, U.; Negi, A.S.; Shanker, K.; Kumar, J.K.; Rahuja, N.; Luqman, S.; Sisodia, B.S.; Saikia, D.; et al. Antimicrobial potential of Glycyrrhiza glabra roots. J. Ethnopharmacol. 2008, 116, 377–380. [Google Scholar] [CrossRef] [PubMed]
  194. Saxena, R.C.; Garg, K.C.; Bhargava, K.P.; Gupta, G.P. A clinical trial of glycyrrhitinic acid in allergic conditions of the eye. J. Indian Med. Prof. 1965, 12, 5487–5490. [Google Scholar] [PubMed]
  195. Lv, H.; Yang, H.; Wang, Z.; Feng, H.; Deng, X.; Cheng, G.; Ci, X. Nrf2 signaling and autophagy are complementary in protecting lipopolysaccharide/d-galactosamine-induced acute liver injury by licochalcone A. Cell Death Dis. 2019, 10, 313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Feng, Y.; Mei, L.; Wang, M.; Huang, Q.; Huang, R. Anti-inflammatory and pro-apoptotic effects of 18beta-glycyrrhetinic acid in vitro and in vivo models of rheumatoid arthritis. Front. Pharmacol. 2021, 12, 681525. [Google Scholar] [CrossRef] [PubMed]
  197. Meng, X.; Zhang, X.; Su, X.; Liu, X.; Ren, K.; Ning, C.; Zhang, Q.; Zhang, S. Daphnes Cortex and its licorice-processed products suppress inflammation via the TLR4/NF-κB/NLRP3 signaling pathway and regulation of the metabolic profile in the treatment of rheumatoid arthritis. J. Ethnopharmacol. 2022, 283, 114657. [Google Scholar] [CrossRef]
  198. Sadinpour, A.; Seyedi, Z.S.; Arabdolatabadi, A.; Razavi, Y.; Ajdary, M. The synergistic effect of Paeonia spp and Glycyrrhiza glabra on polycystic ovary induced in mice. Pak. J. Pharm. Sci. 2020, 33, 1665–1670. [Google Scholar]
  199. Hsu, Y.W.; Chen, H.Y.; Chiang, Y.F.; Chang, L.C.; Lin, P.H.; Hsia, S.M. The effects of isoliquiritigenin on endometriosis in vivo and in vitro study. Phytomedicine 2020, 77, 153214. [Google Scholar] [CrossRef]
  200. Zadeh, J.B.; Kor, Z.M.; Goftar, M.K. Licorice (Glycyrrhiza glabra Linn) as a valuable medicinal plant. Int. J. Adv. Biol. Biomed. Res. 2013, 1, 1281–1288. [Google Scholar]
  201. Pastorino, G.; Cornara, L.; Soares, S.; Rodrigues, F.; Oliveira, M. Liquorice (Glycyrrhiza glabra): A phytochemical and pharmacological review. Phytother. Res. 2018, 32, 2323–2339. [Google Scholar] [CrossRef]
  202. Vispute, S.; Khopade, A. Glycyrrhiza glabra Linn.-“Klitaka”: A Review. Int. J. Pharma Bio Sci. 2011, 2, 42–51. [Google Scholar]
  203. Damle, M. Glycyrrhiza glabra (Liquorice)-a potent medicinal herbInt. J. Herb. Med. 2014, 2, 132–136. [Google Scholar]
  204. Kaur, R.; Kaur, H.; Dhindsa, A.S. Glycyrrhiza glabra: A phytopharmacological review. Int. J. Pharm. Sci. Res. 2013, 4, 2470. [Google Scholar]
  205. Tewari, D.; Mocan, A.; Parvanov, E.D.; Sah, A.N.; Nabavi, S.M.; Huminiecki, L.; Ma, Z.F.; Lee, Y.Y.; Horbańczuk, J.O.; Atanasov, A.G. Ethnopharmacological approaches for therapy of jaundice: Part II. Highly used plant species from Acanthaceae, Euphorbiaceae, Asteraceae, Combretaceae, and Fabaceae families. Front. Pharmacol. 2017, 8, 519. [Google Scholar] [CrossRef] [Green Version]
  206. El-Saber Batiha, G.; Magdy Beshbishy, A.; El-Mleeh, A.; Abdel-Daim, M.M.; Prasad Devkota, H. Traditional uses, bioactive chemical constituents, and pharmacological and toxicological activities of Glycyrrhiza glabra L. (Fabaceae). Biomolecules 2020, 10, 352. [Google Scholar] [CrossRef] [Green Version]
  207. Najafi, S.; Koujan, S.E.; Manifar, S.; Kharazifard, M.J.; Kidi, S.; Hajheidary, S. Preventive effect of Glycyrrhiza glabra extract on oral mucositis in patients under head and neck radiotherapy: A randomized clinical trial. J. Dent. 2017, 14, 267–274. [Google Scholar]
  208. Lee, S.H.; Bae, I.H.; Choi, H.; Choi, H.W.; Oh, S.; Marinho, P.A.; Min, D.J.; Kim, D.Y.; Lee, T.R.; Lee, C.S.; et al. Ameliorating effect of dipotassium glycyrrhizinate on an IL-4- and IL-13-induced atopic dermatitis-like skin-equivalent model. Arch. Dermatol. Res. 2019, 311, 131–140. [Google Scholar] [CrossRef]
  209. Shim, J.Y.; Yim, S.B.; Chung, J.H.; Hong, K.S. Antiplaque and antigingivitis effects of a mouthrinse containing cetylpyridinium chloride, triclosan and dipotassium glycyrrhizinate. J. Periodontal Implant Sci. 2012, 42, 33–38. [Google Scholar] [CrossRef]
  210. Vitali, R.; Palone, F.; Cucchiara, S.; Negroni, A.; Cavone, L.; Costanzo, M.; Aloi, M.; Dilillo, A.; Stronati, L. Dipotassium glycyrrhizate inhibits hmgb1-dependent inflammation and ameliorates colitis in mice. PLoS ONE 2013, 8, e66527. [Google Scholar] [CrossRef] [Green Version]
  211. Okimasu, E.; Moromizato, Y.; Watanabe, S.; Sasaki, J.; Shiraishi, N.; Morimoto, Y.M.; Miyahara, M.; Utsumi, K. Inhibition of phospholipase A2 and platelet aggregation by glycyrrhizin, an antiinflammation drug. Acta Med. Okayama 1983, 37, 385–391. [Google Scholar]
  212. Leite, C.D.S.; Pires, O.C.; Tenis, D.G.; Ziegler, J.V.N.; Priolli, D.G.; Rocha, T. Effects of dipotassium glycyrrhizinate on wound healing. Acta Cir. Bras. 2021, 36, e360801. [Google Scholar] [CrossRef] [PubMed]
  213. Harwansh, R.; Patra, K. Pharmacological studies on Glycyrrhiza glabra. Pharmacologyonline 2011, 2, 1032–1038. [Google Scholar]
  214. Bhattacharjee, S.; Bhattacharjee, A.; Majumder, S.; Majumdar, S.B.; Majumdar, S. Glycyrrhizic acid suppresses Cox-2-mediated anti-inflammatory responses during Leishmania donovani infection. J. Antimicrob. Chemother. 2012, 67, 1905–1914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  215. Richard, S.A. Exploring the pivotal immunomodulatory and anti-inflammatory potentials of glycyrrhizic and glycyrrhetinic acids. Mediat. Inflamm. 2021, 2021, 6699560. [Google Scholar] [CrossRef] [PubMed]
  216. Okamoto, T. The protective effect of glycyrrhizin on anti-Fas antibody-induced hepatitis in mice. Eur. J. Pharmacol. 2000, 387, 229–232. [Google Scholar] [CrossRef]
  217. Abe, N.; Ebina, T.; Ishida, N. Interferon induction by glycyrrhizin and glycyrrhetinic acid in mice. Microbiol. Immunol. 1982, 26, 535–539. [Google Scholar] [CrossRef] [Green Version]
  218. Pakravan, F.; Salehabad, N.H.; Karimi, F.; Isfahani, M. N, Comparative study of the effect of licorice muco-adhesive film on radiotherapy induced oral mucositis, a randomized controlled clinical trial. Gulf J. Oncol. 2021, 1, 42–47. [Google Scholar]
  219. Sattari, A.; Shariati, A.; Maram, N.; Ehsanpour, A.; Maraghi, E. Comparative study of the effect of licorice root extract mouthwash and combined mouthwash on the incidence and severity of chemotherapy-induced mucositis symptoms in colon cancer patients admitted to intensive care units. Jundishapur J. Health Sci. 2019; in press. [Google Scholar] [CrossRef]
  220. Martins, M.D.; Marques, M.M.; Bussadori, S.K.; Martins, M.A.; Pavesi, V.C.; Mesquita-Ferrari, R.A.; Fernandes, K.P. Comparative analysis between Chamomilla recutita and corticosteroids on wound healing. An in vitro and in vivo study. Phytother. Res. 2009, 23, 274–278. [Google Scholar] [CrossRef]
  221. Braga, F.T.M.M.; Santos, A.C.F.; Bueno, P.C.P.; Silveira, R.C.C.P.; Santos, C.B.; Bastos, J.K.; Carvalho, E.C. Use of Chamomilla recutita in the prevention and treatment of oral mucositis in patients undergoing hematopoietic stem cell transplantation: A randomized, controlled, phase ii clinical trial. Cancer Nurs. 2015, 38, 322–329. [Google Scholar] [CrossRef]
  222. Srivastava, J.K.; Shankar, E.; Gupta, S. Chamomile: A herbal medicine of the past with bright future. Mol. Med. Rep. 2010, 3, 895–901. [Google Scholar] [PubMed]
  223. Srivastava, J.K.; Pandey, M.; Gupta, S. Chamomile, a novel and selective COX-2 inhibitor with anti-inflammatory activity. Life Sci. 2009, 85, 663–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Smolinski, A.T.; Pestka, J.J. Modulation of lipopolysaccharide-induced proinflammatory cytokine production in vitro and in vivo by the herbal constituents apigenin (chamomile), ginsenoside Rb(1) (ginseng) and parthenolide (feverfew). Food Chem. Toxicol. 2003, 41, 1381–1390. [Google Scholar] [CrossRef]
  225. dos Reis, P.E.D.; Ciol, M.A.; de Melo, N.S.; de Souza Figueiredo, P.T.; Leite, A.F.; de Melo Manzi, N. Chamomile infusion cryotherapy to prevent oral mucositis induced by chemotherapy: A pilot study. Support. Care Cancer 2016, 24, 4393–4398. [Google Scholar] [CrossRef] [PubMed]
  226. Shabanloei, R.; Ahmadi, F.; Vaez, J.; Ansarin, K.; Hajizadeh, E.; Javadzadeh, Y.; Dolathkhah, R.; Gholchin, M. Alloporinol, chamomile and normal saline mouthwashes for the prevention of chemotherapy-induced stomatitis. J. Clin. Diagn. Res. 2009, 3, 1537–1542. [Google Scholar]
  227. Muley, B.; Khadabadi, S.S.; Banarase, N. Phytochemical constituents and pharmacological activities of Calendula officinalis Linn (Asteraceae): A review. Trop. J. Pharm. Res. 2009, 8, 455–465. [Google Scholar] [CrossRef] [Green Version]
  228. Hadfield, R.A.; Vlahovic, T.C.; Khan, M.T. The use of marigold therapy for podiatric skin conditions. Foot Ankle J. 2008, 1, 1–8. [Google Scholar] [CrossRef]
  229. Tanideh, N.; Tavakoli, P.; Saghiri, M.A.; Garcia-Godoy, F.; Amanat, D.; Tadbir, A.A.; Samani, S.M.; Tamadon, A. Healing acceleration in hamsters of oral mucositis induced by 5-fluorouracil with topical Calendula officinalis. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2013, 115, 332–338. [Google Scholar] [CrossRef]
  230. Fronza, M.; Heinzmann, B.; Hamburger, M.; Laufer, S.; Merfort, I. Determination of the wound healing effect of Calendula extracts using the scratch assay with 3T3 fibroblasts. J. Ethnopharmacol. 2009, 126, 463–467. [Google Scholar] [CrossRef]
  231. Dzubak, P.; Hajduch, M.; Vydra, D.; Hustova, A.; Kvasnica, M.; Biedermann, D.; Markova, L.; Urban, M.; Sarek, J. Pharmacological activities of natural triterpenoids and their therapeutic implications. Nat. Prod. Rep. 2006, 23, 394–411. [Google Scholar] [CrossRef]
  232. Givol, O.; Kornhaber, R.; Visentin, D.; Cleary, M.; Haik, J.; Harats, M. A systematic review of Calendula officinalis extract for wound healing. Wound Repair Regen. 2019, 27, 548–561. [Google Scholar] [CrossRef] [PubMed]
  233. Middleton, E., Jr.; Kandaswami, C.; Theoharides, T.C. The effects of plant flavonoids on mammalian cells: Implications for inflammation, heart disease, and cancer. Pharmacol. Rev. 2000, 52, 673–751. [Google Scholar] [PubMed]
  234. Babaee, N.; Moslemi, D.; Khalilpour, M.; Vejdani, F.; Moghadamnia, Y.; Bijani, A.; Baradaran, M.; Kazemi, M.T.; Khalilpour, A.; Pouramir, M.; et al. Antioxidant capacity of Calendula officinalis flowers extract and prevention of radiation induced oropharyngeal mucositis in patients with head and neck cancers: A randomized controlled clinical study. Daru 2013, 21, 18, (Iranian Registry of Clinical Trials: 201106076734N1). [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Maddocks-Jennings, W.; Wilkinson, J.M.; Cavanagh, H.M.; Shillington, D. Evaluating the effects of the essential oils Leptospermum scoparium (manuka) and Kunzea ericoides (kanuka) on radiotherapy induced mucositis: A randomized, placebo controlled feasibility study. Eur. J. Oncol. Nurs. 2009, 13, 87–93. [Google Scholar] [CrossRef]
  236. You, W.C.; Hsieh, C.C.; Huang, J.T. Effect of extracts from indigowood root (Isatis indigotica Fort.) on immune responses in radiation-induced mucositis. J. Altern. Complement. Med. 2009, 15, 771–778. [Google Scholar] [CrossRef]
  237. Loo, W.T.; Jin, L.J.; Chow, L.W.; Cheung, M.N.; Wang, M. Rhodiola algida improves chemotherapy-induced oral mucositis in breast cancer patients. Expert Opin. Investig. Drugs 2010, 19, S91–S100. [Google Scholar] [CrossRef] [PubMed]
  238. Mutluay Yayla, E.; Izgu, N.; Ozdemir, L.; Aslan Erdem, S.; Kartal, M. Sage tea-thyme-peppermint hydrosol oral rinse reduces chemotherapy-induced oral mucositis: A randomized controlled pilot study. Complement. Ther. Med. 2016, 27, 58–64. [Google Scholar] [CrossRef] [PubMed]
  239. Tanideh, N.; Badie, A.D.; Habibagahi, R.; Koohi-Hosseinabadi, O.; Haghnegahdar, S.; Andisheh-tadbir, A. Effect of topical 2% eucalyptus extract on 5-fu-induced oral mucositis in male golden hamsters. Braz. Dent. J. 2020, 31, 310–318. [Google Scholar] [CrossRef]
  240. Koohi-Hosseinabadi, O.; Andisheh-Tadbir, A.; Bahadori, P.; Sepehrimanesh, M.; Mardani, M.; Tanideh, N. Comparison of the therapeutic effects of the dietary and topical forms of Zizyphus jujuba extract on oral mucositis induced by 5-fluorouracil: A golden hamster model. J. Clin. Exp. Dent. 2015, 7, e304–e309. [Google Scholar] [CrossRef] [Green Version]
  241. Aghamohammadi, A.; Moslemi, D.; Akbari, J.; Ghasemi, A.; Azadbakht, M.; Asgharpour, A.; Hosseinimehr, S.J. The effectiveness of Zataria extract mouthwash for the management of radiation-induced oral mucositis in patients: A randomized placebo-controlled double-blind study. Clin. Oral Investig. 2018, 22, 2263–2272. [Google Scholar] [CrossRef]
  242. Soares, A.D.S.; Wanzeler, A.M.V.; Cavalcante, G.H.S.; Barros, E.; Carneiro, R.C.M.; Tuji, F.M. Therapeutic effects of andiroba (Carapa guianensis Aubl) oil, compared to low power laser, on oral mucositis in children underwent chemotherapy: A clinical study. J. Ethnopharmacol. 2021, 264, 113365. [Google Scholar] [CrossRef] [PubMed]
  243. Wanzeler, A.M.V.; Júnior, S.M.A.; Gomes, J.T.; Gouveia, E.H.H.; Henriques, H.Y.B.; Chaves, R.H.; Soares, B.M.; Salgado, H.L.C.; Santos, A.S.; Tuji, F.M. Therapeutic effect of andiroba oil (Carapa guianensis Aubl.) against oral mucositis: An experimental study in golden Syrian hamsters. Clin. Oral Investig. 2018, 22, 2069–2079. [Google Scholar] [CrossRef] [PubMed]
  244. Hasheminasab, F.S.; Hashemi, S.M.; Dehghan, A.; Sharififar, F.; Setayesh, M.; Sasanpour, P.; Tasbandi, M.; Raeiszadeh, M. Effects of a Plantago ovata-based herbal compound in prevention and treatment of oral mucositis in patients with breast cancer receiving chemotherapy: A double-blind, randomized, controlled crossover trial. J. Integr. Med. 2020, 18, 214–221, (Iranian Registry of Clinical Trials IRCT: 20180923041093N1). [Google Scholar] [CrossRef] [PubMed]
  245. Bertoglio, J.C.; Folatre, I.; Bombardelli, E.; Riva, A.; Morazzoni, P.; Ronchi, M.; Petrangolini, G. Management of gastrointestinal mucositis due to cancer therapies in pediatric patients: Results of a case series with SAMITAL®. Future Oncol. 2012, 8, 1481–1486. [Google Scholar] [CrossRef] [PubMed]
  246. Davarmanesh, M.; Miri, R.; Haghnegahdar, S.; Tadbir, A.A.; Tanideh, N.; Saghiri, M.A.; Garcia-Godoy, F.; Asatourian, A. Protective effect of bilberry extract as a pretreatment on induced oral mucositis in hamsters. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2013, 116, 702–708. [Google Scholar] [CrossRef]
  247. Morazzoni, P.; Petrangolini, G.; Bombardelli, E.; Ronchi, M.; Cabri, W.; Riva, A. SAMITAL®: A new botanical drug for the treatment of mucositis induced by oncological therapies. Future Oncol. 2013, 9, 1717–1725. [Google Scholar] [CrossRef]
  248. Mardani, M.; Afra, S.M.; Tanideh, N.; Tadbir, A.A.; Modarresi, F.; Koohi-Hosseinabadi, O.; Iraji, A.; Sepehrimanesh, M. Hydroalcoholic extract of Carum carvi L. in oral mucositis: A clinical trial in male golden hamsters. Oral Dis. 2016, 22, 39–45. [Google Scholar] [CrossRef]
  249. Tanideh, N.; Davarmanesh, M.; Andisheh-Tadbir, A.; Ranjbar, Z.; Mehriar, P.; Koohi-Hosseinabadi, O. Healing acceleration of oral mucositis induced by 5-fluorouracil with Pistacia atlantica (bene) essential oil in hamsters. J. Oral Pathol. Med. 2017, 46, 725–730. [Google Scholar] [CrossRef]
  250. Tanideh, N.; Zareh, A.A.; Fani, M.M.; Mardani, M.; Farrokhi, F.; Talati, A.; Koohi Hosseinabadi, O.; Kamali, M. Evaluation of the effect of a topical gel form of Pistacia atlantica and Trachyspermum ammi on induced oral mucositis in male golden hamsters by bio-marker indices and stereological assessment. J. Dent. 2019, 20, 240–248. [Google Scholar]
  251. Tanideh, N.; Namazi, F.; Andisheh Tadbir, A.; Ebrahimi, H.; Koohi-Hosseinabadi, O. Comparative assessment of the therapeutic effects of the topical and systemic forms of Hypericum perforatum extract on induced oral mucositis in golden hamsters. Int. J. Oral Maxillofac. Surg. 2014, 43, 1286–1292. [Google Scholar] [CrossRef]
  252. Koohi-Hosseinabadi, O.; Ranjbar, Z.; Sepehrimanesh, M.; AndisheTadbir, A.; Poorbaghi, S.L.; Bahranifard, H.; Tanideh, N.; Koohi-Hosseinabadi, M.; Iraji, A. Biochemical, hematological, and pathological related healing effects of Elaeagnus angustifolia hydroalcoholic extract in 5-fluorouracil-induced oral mucositis in male golden hamster. Environ. Sci. Pollut. Res. Int. 2017, 24, 24447–24453. [Google Scholar] [CrossRef] [PubMed]
  253. Kuduban, O.; Mazlumoglu, M.R.; Kuduban, S.D.; Erhan, E.; Cetin, N.; Kukula, O.; Yarali, O.; Cimen, F.K.; Cankaya, M. The effect of Hippophae rhamnoides extract on oral mucositis induced in rats with methotrexate. J. Appl. Oral Sci. 2016, 24, 423–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Erhan, E.; Terzi, S.; Celiker, M.; Yarali, O.; Cankaya, M.; Cimen, F.K.; Malkoc, I.; Suleyman, B. Effect of Hippophae rhamnoides extract on oxidative oropharyngeal mucosal damage induced in rats using methotrexate. Clin. Exp. Otorhinolaryngol. 2017, 10, 181–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Shahiwala, A. Applications of Polymers in Buccal Drug Delivery. In Applications of Polymers in Drug Delivery; Elsevier: Amsterdam, The Netherlands, 2021; pp. 43–76. [Google Scholar]
  256. Sandri, G.; Ruggeri, M.; Rossi, S.; Bonferoni, M.C.; Vigani, B.; Ferrari, F. (Trans) buccal drug delivery. In Nanotechnology for Oral Drug Delivery; Academic Press: London, UK, 2020; pp. 225–250. [Google Scholar]
  257. Moroz, E.; Matoori, S.; Leroux, J.C. Oral delivery of macromolecular drugs: Where we are after almost 100 years of attempts. Adv. Drug Deliv. Rev. 2016, 101, 108–121. [Google Scholar] [CrossRef] [Green Version]
  258. Li, T.; Lalla, R.V.; Burgess, D.J. Enhanced drug loading of in situ forming gels for oral mucositis pain control. Int. J. Pharm. 2021, 595, 120225. [Google Scholar] [CrossRef]
  259. Paderni, C.; Compilato, D.; Giannola, L.I.; Campisi, G. Oral local drug delivery and new perspectives in oral drug formulation. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2012, 114, e25–e34. [Google Scholar] [CrossRef]
  260. Tedesco, M.P.; Monaco-Lourenco, C.A.; Carvalho, R.A. Characterization of oral disintegrating film of peanut skin extract-Potential route for buccal delivery of phenolic compounds. Int. J. Biol. Macromol. 2017, 97, 418–425. [Google Scholar] [CrossRef]
  261. Aksungur, P.; Sungur, A.; Unal, S.; Iskit, A.B.; Squier, C.A.; Senel, S. Chitosan delivery systems for the treatment of oral mucositis: In Vitro and in vivo studies. J. Control. Release 2004, 98, 269–279. [Google Scholar] [CrossRef]
  262. Fonseca-Santos, B.; Chorilli, M. An overview of polymeric dosage forms in buccal drug delivery: State of art, design of formulations and their in vivo performance evaluation. Mater. Sci. Eng. C Mater. Biol. Appl. 2018, 86, 129–143. [Google Scholar] [CrossRef] [Green Version]
  263. Borges, J.G.; De Carvalho, R.A. Orally disintegrating films containing propolis: Properties and release profile. J. Pharm. Sci. 2015, 104, 1431–1439. [Google Scholar] [CrossRef]
  264. Agrawal, U.; Sharma, R.; Gupta, M.; Vyas, S.P. Is nanotechnology a boon for oral drug delivery? Drug Discov. Today 2014, 19, 1530–1546. [Google Scholar] [CrossRef] [PubMed]
  265. Morantes, S.J.; Buitrago, D.M.; Ibla, J.F.; García, Y.M.; Lafaurie, G.I.; Parraga, J.E. Composites of hydrogels and nanoparticles. In Biopolymer-Based Composites; Woodhead Publishing: Duxford, UK, 2017; pp. 107–138. [Google Scholar]
  266. Allaker, R.P.; Yuan, Z. Nanoparticles and the control of oral biofilms. In Nanobiomaterials in Clinical Dentistry; Elsevier: Amsterdam, The Netherlands, 2019; pp. 243–275. [Google Scholar]
  267. Campos, J.C.; Cunha, D.; Ferreira, D.C.; Reis, S.; Costa, P.J. Oromucosal precursors of in loco hydrogels for wound-dressing and drug delivery in oral mucositis: Retain, resist, and release. Mater. Sci. Eng. C Mater. Biol. Appl. 2021, 118, 111413. [Google Scholar] [CrossRef] [PubMed]
  268. Ryu, J.H.; Choi, J.S.; Park, E.; Eom, M.R.; Jo, S.; Lee, M.S.; Kwon, S.K.; Lee, H. Chitosan oral patches inspired by mussel adhesion. J. Control. Release 2020, 317, 57–66. [Google Scholar] [CrossRef] [PubMed]
  269. Takeuchi, I.; Kamiki, Y.; Makino, K. Therapeutic efficacy of rebamipide-loaded PLGA nanoparticles coated with chitosan in a mouse model for oral mucositis induced by cancer chemotherapy. Colloids Surf. B 2018, 167, 468–473. [Google Scholar] [CrossRef]
Figure 1. Diagram representing the mucosal cells and clinical manifestations of oral mucositis.
Figure 1. Diagram representing the mucosal cells and clinical manifestations of oral mucositis.
Ijms 23 04385 g001
Figure 2. SWOT analysis for the possible use of natural products to prevent/treat OM.
Figure 2. SWOT analysis for the possible use of natural products to prevent/treat OM.
Ijms 23 04385 g002
Table 1. Available clinical scales for oral mucositis assessment. Adapted from [16]. NA—Not applicable.
Table 1. Available clinical scales for oral mucositis assessment. Adapted from [16]. NA—Not applicable.
ScaleGrade 0Grade 1
(Mild)
Grade 2
(Moderate)
Grade 3
(Severe)
Grade 4
(Life-Threatening)
Grade 5
(Death)
WHONo findingsOral erythema and soreness; no ulcersOral erythema, ulcers; solid diet toleratedOral ulcers; liquid diet onlyOral alimentation impossibleNA
CTCAENoneAsymptomatic or mild symptoms; intervention not indicatedModerate pain or ulcer that does not interfere with oral intake; modified diet indicatedSevere pain, interfering with oral intakeLife-threatening consequences; urgent intervention indicatedDeath
RTOGNo change over baselineIrritation; may experience mild pain, not requiring analgesicsPatchy mucositis that may produce an inflammatory serosanguinous discharge; may experience moderate pain requiring analgesiaConfluent, fibrinous mucositis; may include severe pain requiring narcoticsUlceration, hemorrhage, or necrosisNA
Table 2. Risk factors related to patients, tumors, and treatments in the development of oral mucositis.
Table 2. Risk factors related to patients, tumors, and treatments in the development of oral mucositis.
Risk FactorCriteriaReferences
Related to patient
AgeExtremities[9,12,25,45,56,57,58]
GenderFemale[2,3,10,23]
Body mass index (BMI)Low and high body mass index[2,3,10,23]
Dental prosthesisOrthodontics and prosthesis[9,11]
EducationLack of health literacy[7,55,59,60]
Oral hygieneOral hygiene less than 2 times/day
Periodontal disease
[2,3,10,23]
ComorbiditiesDiabetes mellitus, renal and hepatic dysfunction[16,54,61]
LeucocytesNeutropenic patients are immunocompromised[2,18,45]
AlcoholUse of alcohol prior to and during treatment[2,3,18,45]
SmokingSmoking prior to and during treatment increases the severity[5,45]
GeneticsGenetic polymorphisms (e.g., TNF-α)[10,45]
Mucosal traumaSharpened teeth[5]
Related to tumor
Types of cancerSolid tumors have higher risk, mainly those located near oral cavity[12,45,47]
Related to treatment
Type of treatment5-fluorouracil, Doxorubicin, Methotrexate, Cisplatin, Vinblastine, Mitomycin, Transtuzumabe, Docetaxel, Melphalan[10,16,54]
DoseHigh doses over short periods and their extension[10,16,54]
Type of administrationIntravenous[2,10,16,45,54]
Microbiota
Table 3. Management of Oral Mucositis Guidelines created by the Multinational Association for Supportive Care in Cancer and the International Society of Oral Oncology.
Table 3. Management of Oral Mucositis Guidelines created by the Multinational Association for Supportive Care in Cancer and the International Society of Oral Oncology.
InterventionAimMASCC/ISOO Guideline CategoryResultsReferences
Oral carePreventionSuggestionIncreases patient’s awareness and enhances their compliance with treatment[5,37,46,60,63,67,68,69]
Oral cryotherapyPreventionRecommendationLocal vasoconstriction that minimizes drug absorption[11,46,70,71,72,73]
Photobiomodulation therapyPreventionRecommendationPromotes wound healing and has an anti-inflammatory effect[23,26,39,40,41,46]
Benzydamine mouthwashPreventionRecommendationAnti-inflammatory properties by inhibiting the production of pro-inflammatory cytokines[46,53,54,74,75,76]
Keratinocyte growth factor-1 (palifermin)PreventionRecommendationProliferation and restoration of epithelial cells[26,27,46]
GlutaminePreventionSuggestionIt is used by cells of the immune system[28,30,34,46]
HoneyPreventionSuggestionInhibits bacterial growth and enhances healing rate[32,35,37,38,46,77]
Patient-controlled analgesia (e.g., 0.2% morphine mouthwash)TreatmentRecommendationPain management[46,78,79]
Zinc supplementsPreventionSuggestionPrevents lipids peroxidation and replaces redox-reactive metals[28,30,46]
Doxepin mouthwashTreatmentSuggestionIn topical application, it has analgesic and anesthetic properties[46,78,80]
Vitamin EPreventionSuggestionAntioxidant that may protect tissue damage from free oxygen radicals[28,30,31,46,73]
AmifostinePreventionSuggestionReduces DNA strand breaks, recruits ROS scavengers, and preserves salivary glands, endothelium, and connective tissue integrity[4,33,46]
Table 4. Summary of studies with natural products for prevention/treatment of oral mucositis.
Table 4. Summary of studies with natural products for prevention/treatment of oral mucositis.
NameProperties/MechanismsApplicationExperimental
Setting/Model
References
Manuka (Leptospermum scoparium) essential oilAnti-inflammatory, analgesic, antimycotic, and antibacterialMouthwashRandomized placebo-controlled trial[235]
Kanuka (Kunzea ericoides) essential oilAnti-inflammatory, analgesic, antimycotic, and antibacterialMouthwashRandomized placebo-controlled trial[235]
Indigo root
(Isatis indigotica)
Anti-inflammatory and antiviralMouthwashRandomized clinical trial[236]
Rhodiola algidaImmunomodulatory effectsMouthwashRandomized clinical trial[237]
Thymus spp. LAntiseptic, anti-inflammatory, antimicrobial, and antimycoticMouthwashRandomized pilot study[238]
EucalyptusAntibacterial, antiviral, antifungal, anti-inflammatory, analgesic, and antioxidantTopical gelHamsters[239]
Zizyphus jujubaAnti-inflammatory, analgesic, and wound healingTopical gel and dietary Hamsters[240]
Zataria multifloraCarminative, stimulant, diaphoretic, diuretic, antiseptic, anesthetic, antispasmodic, anti-hermitic, antidiarrheal, and analgesicMouthwashRandomized clinical trial[241]
Carapa guianensis oilAnti-inflammatory, analgesic, and antimicrobialTopical gel/swabControlled and randomized clinical trial/ hamsters[242,243]
Plantago ovataAntioxidant, anti-inflammatory, and antibacterialMouthwashRandomized cross-over clinical trial[244]
Achillea millefoliumAntimicrobial and anti-inflammatoryMouthwashDouble-blind, randomized, controlled trial[82]
Vaccinium myrtillusAntioxidant, cardioprotective, neuroprotective, anti-inflammatory, and anticarcinogenicTopical application, gavage administration, mouthwashClinical trials, Hamsters[245,246,247]
Carum carviAntioxidant, antidiabetic, antifungal, and antimicrobialTopical gelHamsters[248]
Pistacia atlanticaAntioxidant and anti-inflammatoryTopical gelHamsters[249,250]
Hypericum perforatumAntioxidant and anti-inflammatoryTopical gelHamsters[251]
Elaeagnus angustifoliaAnti-inflammatory, analgesic, and wound healingTopical gelHamster[252]
Trachyspermum ammiAnti-inflammatory, antiviral, antifungal, antioxidant, and analgesicTopical gelHamsters[250]
Hippophae rhamnoidesAntioxidant, anti-inflammatory, antimicrobial, and anti-ulcerogenicGavage administrationRats[253,254]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ferreira, A.S.; Macedo, C.; Silva, A.M.; Delerue-Matos, C.; Costa, P.; Rodrigues, F. Natural Products for the Prevention and Treatment of Oral Mucositis—A Review. Int. J. Mol. Sci. 2022, 23, 4385. https://doi.org/10.3390/ijms23084385

AMA Style

Ferreira AS, Macedo C, Silva AM, Delerue-Matos C, Costa P, Rodrigues F. Natural Products for the Prevention and Treatment of Oral Mucositis—A Review. International Journal of Molecular Sciences. 2022; 23(8):4385. https://doi.org/10.3390/ijms23084385

Chicago/Turabian Style

Ferreira, Ana Sofia, Catarina Macedo, Ana Margarida Silva, Cristina Delerue-Matos, Paulo Costa, and Francisca Rodrigues. 2022. "Natural Products for the Prevention and Treatment of Oral Mucositis—A Review" International Journal of Molecular Sciences 23, no. 8: 4385. https://doi.org/10.3390/ijms23084385

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