Open access peer-reviewed chapter

Salivary Gland Radio-Protection, Regeneration and Repair: Innovative Strategies

Written By

Ziyad S. Haidar

Submitted: 25 September 2020 Reviewed: 04 November 2020 Published: 11 January 2021

DOI: 10.5772/intechopen.94898

From the Edited Volume

Biomechanics and Functional Tissue Engineering

Edited by Ziyad S. Haidar, Ibrokhim Y. Abdurakhmonov and Abdelwahed Barkaoui

Chapter metrics overview

606 Chapter Downloads

View Full Metrics

Abstract

Saliva has a critical role in the maintenance of oral, dental and general health and well-being. Alteration(s) in the amount/quantity and/or quality of secreted saliva may induce the development of several oro-dental variations, thereby negatively-impacting overall quality of life. Diverse factors may affect the process of saliva production and quantity/quality of secretion, including medications, systemic or local pathologies and/or reversible/irreversible damage. Indeed, chemo- and/or radio-therapy, particularly, in cases of head and neck cancer, for example, are well-documented to induce serious damage and dysfunction to the radio-sensitive salivary gland tissue, resulting in hypo-salivation, xerostomia (dry mouth) as well as numerous other adverse intra−/extra-oral, medical and quality-of-life issues. Although a single governing mechanism of radiation-induced salivary gland tissue damage and dysfunction has not been yet elucidated, the potential for a synergy in radio-protection (mainly, and possible -reparation) via a combinatorial approach of mechanistically distinct strategies, has been suggested and explored over the years. This is, undoubtfully, in parallel to the ongoing efforts in improving the precision, safety and efficacy of radiotherapy protocols/outcomes, as well as in developing new technological and pharmaceutical alternatives, topics covered in this chapter.

Keywords

  • radioprotection
  • salivary gland
  • xerostomia
  • head and neck cancer
  • oro-dental health

1. Introduction

It is well recognized that the incidence of cancer, the second leading cause of death, globally, is increasing, an ongoing major burden of disease and public health burden, World-wide. While there were 14.1 million cancer cases reported in 2012, the World Health Organization (WHO) estimated about 1 in 6 deaths is due to cancer, with 9.6 million such deaths reported in 2018. In the United States, today, cancer is the second leading, after heart disease, cause of death amongst men and women, with over 1 million new cases diagnosed, annually [1].

Despite a reduction in tobacco consumption and the significant modern advancements in medicine, the number of new cancer cases, per year, is projected to rise to 22.2 million by 2030 [1]. Cancers, often squamous cell carcinomas/neoplasms, that involve the oral cavity, nostrils, paranasal sinuses, naso−/oro−/hypo-pharynx, larynx, and the salivary glands, are commonly/collectively (despite their heterogeneity) termed head and neck cancers (HNC), which, together are responsible for nearly 200,000 deaths, a year, World-wide [2]. In the United States alone, HNC represent 4–5% of all cancers, and in Europe, HNC are the sixth most common group of cancers [3].

Besides the alarming incidence and mortality rates, HNC suffer a relatively poor prognosis, overall, whether due to delays in diagnosis, staging, treatment, particulars of the tumor site, onset, type of symptoms and/or efficacy of therapies, to mention a few. Such factors further contribute to permitting the progress and upstaging of the malignant tumor(s) which eventually result in enfeebled survival, despite the application of novel or advanced intensive therapeutic regimens. Briefly, treatment, often a multi-disciplinary case-specific approach, can employ chemo−/radio−/immune-therapy, surgery, or combinatorial strategies [4].

Herein, radiotherapy (RT), whether radical or prophylactic, remains a mainstay of HNC treatment, especially in light of modern improvements in precisely targeting and delivering the required radiation doses to the tumor, thereby allowing additional sparing of normal/healthy surrounding tissue(s), greatly reducing side or adverse effects of radiation, and consequently improving the quality of life (QoL) of patients as well as their families [5, 6, 7, 8]. IMRT (intensity-modulated radiotherapy), VMAT (volumetric modulated arc therapy) and particle (ion-based) therapy are perhaps fine examples of modern high-precision RT [7].

RT, in general, aims to realize localized destruction and control of the target tumor (−cells) and halt of the reproductive potential, while minimizing toxicity onset. Specifically, high-energy radiation is deposited, causing DNA strands to break thereby damaging the cell genome either directly or indirectly (via free-radical production) and subsequently resulting in apoptosis, mitotic cell death, and tissue hypoxia, through different cascades and processes [5, 7]. Depending on the radiation dose and tissue turnover, amongst other factors, RT can almost always be expected to result in a range of side effects, of which some are reversible and others are irreversible (Figure 1). Indeed, HNC and oral squamous cell carcinoma (OSCC) patients receiving RT often experience pain, taste disturbances, difficulties in mastication and deglutition (swallowing) and suffer from mucositis, fungal infections, dental decay, alterations in speech, all of which are mainly due to or linked to salivary gland dysfunction which in turn results in hyposalivation and xerostomia [9, 10, 11, 12].

Figure 1.

Head and neck cancers regions and irradiation intensity risk during HNC radiotherapy.

Herein, xerostomia, a dry mouth sensation, is one of the main complications and complaints for HNC patients receiving RT, mainly as a sequela of the un-avoidable damage to the parotid and sub-mandibular glands (both produce over 80% of saliva) anatomically located with the radiation zone [8, 12]. Inflammation, fibrosis, atrophy and the reduced wound healing response, i.e. reparative and regenerative capacity of the glands, mainly due to lack of functional salivary gland stem/progenitor cells post-irradiation, render the inevitable radiotherapy-induced salivary gland damage and dysfunction, whether occurring early or late, a significant impediment to the QoL and survival of HNC and OSCC patients [10, 13, 14].

Therefore, besides modern advancements in radiation engineering technologies, ample pharmacological and pharmaceutical solutions have been explored [14]. Accumulating knowledge in understanding underlying signaling pathways, cellular and tissue responses, spatio-temporally, fuel the continuing efforts aimed to explore, develop and translate novel solutions to support in the prevention (and treatment of) radiation-induced side-effects and damage of salivary glands, a main focus of this chapter, designed to provide the clinical reader with a summary of relevant literature and recent innovative developments in salivary gland radioprotection and potential salivary gland repair, post-RT.

Advertisement

2. Saliva and salivary glands: pre-, during- and post-RT

Briefly, exocrine salivary glands are classified as either major (parotid, sub-mandibular and sub-lingual) or minor (labial and buccal gland, glosso-palatine gland, and palatine and lingual) glands. Anatomically, all three major glands are highly vascularized, innervated and are architecturally similar featuring a ductal structure with a secretory/excretory (saliva-producing acini surrounded by myo-epithelial cells, myo-fibroblasts, immune cells, stromal cells, endothelial cells and nerve fibers) opening into the oral cavity/mouth [15]. The glands differ in their type of acinar cells and as a result, in the type of produced saliva. While the parotid is composed of only serous acini thereby producing watery saliva, the sub-mandibular and sub-lingual glands contain a mix of serous and mucous (glycoprotein- rich) acini, thereby producing saliva of a different composition, a seromucous secretion. Secretion of saliva is stimulated by the sympathetic (proteins) and parasympathetic (serous/ions) branches of the autonomic nervous system [15, 16].

Saliva is basically an oral lubricant fluid with multiple digestive functions critical for oro-dental health, QoL and general well-being. It is composed of a complex mixture of water (99%), electrolytes (sodium, potassium, calcium, magnesium, etc. …), mucins, proteins, white blood cells, epithelial cells, immunoglobulins, anti-microbials/−bacterials and enzymes (1%) [16, 17, 18]. Hence, saliva is essential for moistening, chewing, swallowing and chemically-digesting foods. It also facilitates speaking, aids the tongue in taste sensing, helps protect the oral mucosa (localized immunity/mucosal resistance) and plays a role in tissue re-mineralization. A healthy adult produces/secretes a daily average of 0.5–1.5 L, at differential rates over the day, and at a near neutral (buffer) pH of 6.7 [15, 16, 17, 19, 20, 21, 22].

Therefore, alterations in quantity (↓: hypo- or ↑: hyper-salivation) or quality of the secreted/produced saliva are associated to a variety of conditions and diseases and have been associated with some medications and therapies [23]. For instance, sialorrhea is a general term used for hyper-salivation (or drooling), often as a result of medication, systemic diseases, psychiatric disorders and/or oral pathologies, amongst others [14]. It is also often linked to conditions such as Parkinson’s, epilepsy, amyotrophic lateral sclerosis or ALS, cerebral palsy, developmental disabilities, pregnancy and/or drugs including clozapine [16, 24] Common treatments for sialorrhea include surgical intervention, radiation of the salivary glands (to halt and diminish its function) and the use of oral anti-cholinergic drugs (to inhibit saliva production), however with known side or adverse effects. In recent years, numerous studies investigated the use of neuro-toxins, mainly botulinum neurotoxins or BoNTs, which basically are bacterial exotoxins that interfere and block the exocytotic release of vesicular neuro-transmitters cholinergic neuromuscular activity in the target tissue, including commercially-available RimabotulinumtoxinB (RimaBoNT-B, FDA approval in 2000) and IncobotulinumtoxinA (IncoBoNT-A, FDA approval in 2010) in patients suffering sialorrhea, with attractively promising results [24, 25].

On the other hand, salivary gland hypofunction (progressive loss of gland function) is commonly described or associated with the reduction of salivary flow and production, quantitatively. Frydrych [26], discussed salivary gland hypofunction etiology and classified causes into seven major areas, developmental, autoimmune/chronic inflammatory, endocrine, neurological/psychiatric, metabolic, infectious and iatrogenic [26]. In a healthy individual, un-stimulated “whole” salivary flow rate is averaged at 0.35 mL saliva per minute, with abnormalities indicated if the rate drops. For example, one of the most prevalent and studied diseases or disorders of the salivary gland is Sjögren’s syndrome (SS), a chronic auto-immune inflammatory reaction characterized by lymphocytic infiltration of the exocrine glands (mostly to the salivary or lacrimal glands), which generates a significant reduction in salivary flow rate - to below 0.1 mL whole saliva per minute secreted, un-stimulated [27]. It is perhaps noteworthy herein that whole saliva indicates the collection of saliva (secreted from all salivary glands) present in the mouth. Other quantification techniques require direct collection from the specific gland. Moreover, often is reported in diagnosing SS that only un-stimulated whole saliva flow rates are used.

Hypo-salivation, therefore, is salivary flow rate reduction, quantified, clinically via sialometry. Xerostomia, on the other hand, is the reported perception or sensation, subjectively, of oral dryness. Hypo-salivation may or may not be accompanied by xerostomia, and vice versa. Dryness in the mouth can be a side-effect of medications or due to diseases such as HIV/AIDS, diabetes, hypertension and/or other factors including smoking, dehydration, mouth breathing, aging and/or head and neck irradiation [14, 16, 23, 28, 29]. Indeed, xerostomia is one of the most commonly reported (and expected) complications of RT (during and after RT) for HNC, and as mentioned earlier, mainly as a predictable consequence to the significant damage (and generated inflammatory immune response) caused to the salivary glands which are located and included within the RT-zone or field [30, 31, 32].

RT, besides impairing salivary gland function and salivary flow rate, impacts the quality of the secreted saliva, given the loss or atrophy of acinar and ductal cells and granules (and stem/stromal and progenitor cells) and the consequential morphological changes to salivary fluid quality (including pH and buffering capacity), thereby affecting the essential protective, functional and overall physiologic processes (Figure 2). Such damage [32] and impact can appear as soon as one week after the first radiation therapy session (acute RT-induced damage is due to a disturbance in the involved signal transduction pathways on the cell membrane). Progressive decrease in salivary gland function is evident with more RT sessions (delayed or late RT-induced damage is due to apoptosis-driven parenchymal cell loss, inflammation, blood vessel dilation and function loss, nerve injury and reduced parasympathetic nervous function, and fibrosis) rendering rescue, repair and regeneration rather challenging [33, 34, 35].

Figure 2.

Progression of RT-induced salivary gland damage and dysfunction in HNC patients.

As a result, the QoL of a large proportion of patients receiving RT is severely compromised [36, 37], with thicker or more viscous saliva and xerostomia leading in reported complaints [38]. Indeed, RT-related biochemical and proteomic alterations where several key glycoproteins, proteins and other molecules are affected have been identified [31, 39]. For example, Jehmlich et al. [40], discussed such variations post-RT, detected significant alterations in 48 proteins and highlighted the development of oral mucositis as a result of salivary gland dysfunction. Psycho-social and emotional impact on QoL of HNC patients, especially the elderly [41], where they experience and suffer from a compromised ability to and taste, chew, and swallow foods extended to their forced switching of dietary preferences to soft and carbohydrate-rich foods, thereby resulting in serious nutritional deficiencies [38, 40, 41]. Hyposalivation and sequential xerostomia also affect speaking and communication abilities, and patients experience nocturnal oral discomfort, hence, causing additional stress leading to withdrawal from everyday or day-to-day societal and emotional interactions [42, 43, 44].

Furthermore, with the prolonged oral clearance of sugars, the oral mucosa becomes painfully-dry, sticky and more susceptible to infection, the progression of dental caries (tooth decay), gingival and periodontal disease and trauma, accentuating the importance of oro-dental hygiene and care, especially in the elderly patients [41]. Other sequelae include erosion and ulceration of mucosal tissues, oral candidiasis, dysgeusia and dysphagia. Therefore, it is common for HNC patients to suffer from depression, feelings of anguish and anxiety after receipt of the RT protocol [14, 37, 45, 46, 47, 48, 49]. While the recovery of irradiated salivary glands at the cellular and molecular has been thus far shown to be limited, salivary recovery post-RT, from our clinical exposure and expertise, is possible, yet a lengthy (> 3 years), dire and capricious process, with underlying mechanisms not yet fully understood.

Advertisement

3. Radiation-induced damage prevention and potential regeneration of salivary glands

Understanding the underlying mechanisms governing cellular and molecular control of salivary gland function is highly pertinent, during- and post-RT, to aid in developing suitable and effective therapies, whether preventive or reparative. To date, it is safe to state that available therapies continue to be symptomatic and no definitive solution or approach has been shown to compensate and/or recover the impairment of salivary glands and function. Life-style modifications, synthetic saliva and/or use of salivary stimulants and sialagogues, suffer shortcomings and are not satisfactory to our patients, as they either only provide temporary (short-term) relief or might have other disquieting side-effects. Hence, global attention has been diverted to seek and develop alternative novel methods, tools and therapies, to offer to HNC patients undergoing RT, that can provide superior long-term efficacy. Herein, tissue engineering, regenerative medicine, pharmaceutics and nanotechnology may contribute.

Advertisement

4. Tissue engineering and reparative/regenerative medicine: current regimens and strategies

Several tissues and organs are highly sensitive to irradiation, such as the skin, esophagus and bone marrow. However, the salivary glands are intricately radiosensitive, given their highly-differentiated cell content marked with a very low or slow proliferative rate [50]. This can help explain why the salivary glands, in specific, are somewhat unique in their early- and delayed-effects post-RT, when compared to other tissues and organs. Nonetheless, salivary gland dysfunction and/or hypofunction has been shown, in some cases, to be reversible. Such treatment intervention is multi-factorial and highly-dependent on original causality, for example, in cases of alcohol abuse and dehydration or hypothyroidism. RT-induced salivary gland damage and dysfunction is a far more challenging scenario. Auto-immune/chronic inflammatory diseases, such as SS or systemic lupus erythematosus also result in irreversible damage to the salivary glands [26].

Today, as mentioned earlier, only palliative and efficacy-limited regimens are commercially-available [47]. Tables 14 highlight a selection of various radio-protection strategies, at different stages of development, pre-clinically (in vitro and in in vivo testing) and clinical (human clinical trials). Briefly, database search was performed in PubMed-indexed articles using a multi-search of the following keywords: “Salivary Glands AND Radioprotection [Title/Abstract]”, “Salivary Glands AND Radioprotection [MeSH]”, “Salivary AND Glands AND Radioprotection [Title/Abstract]”, “Salivary Gland AND Radioprotection [Title/Abstract]”, “Salivary AND Gland AND Radioprotection [ALL FIELDS]”, “Salivary Glands AND Radioprotection [ALL FIELDS] and “Salivary Gland AND Radioprotection [ALL FIELDS]”. Eligibility and inclusion criteria included English articles reporting radio-protection data from in vitro, in vivo and/or clinical setting/trials. Articles dated back to 1978 up to the search end-date of December 31st of 2019 were analyzed. Reviews, communications or articles with preliminary results were not included in our analysis (Figure 3). Herein, our purpose is to screen the available literature and assess the level of development of new strategies, regimens and/or innovative solutions, to provide a usable prior-Art formatted report. Hence, not all included articles, which are tabulated for the reader, were aimed to be presented and dissected to be discussed in detail. This review attempts to provide an overview of the current understanding, status and prospect of salivary gland radioprotection systems, with a look onto potential reparative and regenerative keys, where we, amongst other clinicians and researchers, do aspire for a superior, safe, efficacious and long-term innovative solution that reverses RT-induced damage to the salivary glands of our HNC patients. Moreover, we opted to avoid concluding our overview with calls for additional research or validation, given that vital tissue engineering strategies employing the design, characterization and optimization of novel biomaterials (and 3D printing), that can also be housing/incorporating release-controlled nanoparticles or nanocapsules that also are designed to encapsulate distinct mesenchymal stem cells, induced pluripotent stem cells (iPSCs), growth factors or cytokines and/or pharmaceutical agents or drugs, currently investigated at different levels of development are limitless in distinctions and details.

AgentMain FindingsRef
bFGF-PLGA microspheresAdministration of basic Fibroblast Growth Factor (bFGF) prior to and immediately after irradiation, partially protected (44%) the rat parotid gland.[51]
pH-responsive nanoparticles for active siRNAs deliveryIntroduction of siRNAs specifically targeting the Pkcδ or Bax genes significantly blocked the induction of these pro-apoptotic proteins that normally occurs post-irradiation in cultured salivary gland cells. Level of cell death from subsequent irradiation was significantly decreased.[52]
rhHGFTreatment of irradiated hPTS with recombinant human Hepatocyte Growth Factor (rhHGF) restored salivary marker expression and secretory function of hPTS. Changes in the phosphorylation levels of apoptosis-related proteins through HGF-MET axis inhibited irradiation-induced apoptosis.[53]
TIGAR over-expressionTIGAR (a p53-inducible regulator of glycolysis and apoptosis) over-expression could diminish the radio-sensitivity of Hs 917.T cells, and decrease the autophagy level induced by ionizing irradiation.[54]

Table 1.

Radioprotection of salivary glands, in vitro.

MICE
AgentMain FindingsRef
Keratinocyte Growth Factor-1 (KGF-1)Local delivery of keratinocyte growth factor-1 into irradiated salivary glands protected RT-induced salivary cell damage, suppressed p53-mediated apoptosis and prevented salivary hypofunction.[55]
pH-responsive nanoparticles complexed with siRNAsKnockdown of Pkcδ reduced the number of apoptotic cells during the acute phase of irradiation damage and also markedly improved salivary secretion at 3 months.[52]
Dasatinib / ImatinibDelivery of dasatinib or imatinib resulted in >75% protection/rescue of salivary gland function at 60 days end-point. Continuous dosing with dasatinib extended protection to at least 5 months and was correlated with histologic evidence of regenerated salivary gland acinar cells.[56]
Human Adipose tissue-derived Mesenchymal Stem Cells (AdMSCs)Local transplantation of AdMSCs improved tissue remodeling following irradiation-induced damage in salivary gland tissue. The use of a carrier enhanced the effects of AdMSC-mediated cellular protection against irradiation via paracrine secretion.[57]
Botulinum Toxins (BTX)Irradiated mice showed a 50% reduction in salivary flow after 3 days, whereas mice pre-injected with BTX had 25% reduction in salivary flow rate (ƿ < 0.05). BTX pre-treatment ameliorates RT-induced salivary gland dysfunction.[58]
AdMSCs secretomeSecretome modulated by hypoxic conditions to contain therapeutic factors contributed to salivary gland tissue re-modeling and demonstrated a potential to improve consequences of RT-induced salivary hypofunction.[59]
Resveratrol (RES)Administration of RES reversed the reduction of saliva secretion induced by irradiation and restored salivary amylase and superoxide dismutase activity. RES can protect salivary glands against the negative effects of irradiation.[60]
MICE
AgentMain FindingsRef
AmifostineAmifostine alleviated the effects of irradiation on the bio-functions of cells, such as organelles, highly-involved in the secretory process. Amifostine can alleviate xerostomia caused by the late or delayed effects of irradiation.[61]
Serotype 5 Adenoviral (Ad5) vector-mediated transfer of basic Fibroblast Growth Factor (AdbFGF) or Vascular Endothelial Growth Factor (AdVEGF) complementary DNAsSingle local administration of a modest dose (5 × 109 particles/gland) of a serotype 5 adenovirus (Ad5) vector encoding either bFGF or VEGF prior to irradiartion, prevents rapid micro-vessel density loss in salivary glands and reduces the loss in salivary flow rate (as measured 8 weeks post-RT).[62]
Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl)Tempol treatment was found to protect salivary glands significantly against radiation damage (approximately 60% improvement), with no tumor protection observed.[63]
Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl)Tempol treatment pre-irradiation significantly reduced RT-induced salivary hypofunction (approximately 50–60%). Tempol (I.V. or S.C.) administration also showed significant radio-protection. Topical use of tempol, either as a mouthwash or gel, was also reported to be radioprotective.[64]
Isoproterenol (IPR)IPR, stimulates adenylate cyclase/cyclic AMP (AC/cAMP) to increase the level of cAMP,25 and then increases cellular membrane ion permeability, ion active transport, and protein bio-synthesis. These events, together with the release of heavy metals, appear to reduce irradiation injury.[65]
Tempol (4-hydroxy-2,2,6,6-tetramethylpiperidine-N-oxyl)Irradiation resulted in a dose-dependent reduction of salivary flow rate in this mouse model.[66]
WR-2721
WR-3689
WR779 13
Tumors examined take up less WR-3689 than the other two protectors. In RIF-1 tumor, WR-3689 is taken up most avidly, but the three drugs tend to be equally protective.[67]
WR-2721There is potential for protecting dose-limiting, late-responding normal tissue in the RT of human tumors with both neutrons and conventional radiotherapy.[68]
WR-1065Localized delivery to salivary glands markedly improved radioprotection at the cellular level. Also, mitigated the adverse side-effects associated with systemic administration.[69]
Hypoxia pre-conditioned human Adipose tissue-derived Mesenchymal Stem Cells (hAdMSCs-HPX)Results suggest that hAdMSCs-HPX protect salivary glands from RT-induced apoptosis, and preserve acinar structure and functions via the activation of FGFR-PI3K signaling by actions of hAdMSC- secreted factors, including FGF-10.[70]
EntolimodAt days 8 and 15, entolimod treatment led to noticeable mitigation of damage in salivary gland tissue. Treatment 1 hr. post-RT irradiation seems more effective than 30 min pre-RT.[71]
MICE
AgentMain FindingsRef
Statins (Simvastatin)Administration of Simvastatin could delay and reduce the extent of elevation/over-expression of TGF-β1, which in turn protects the submandibular glands from RT-induced injury.[72]
RAT
AgentMain FindingsRef
Se, Zn and Mn + Lachesis muta venom (O-LM)O-LM prevented permanent submandibular gland alterations demonstrating promising results in radioprotection and recovery from RT-induced injury.[73]
Pilocarpine, Methacholine, Reserpine and Methacholine + ReserpinePre-treatment with pilocarpine or methacholine improved all measured glandular functions. Pre-treatment with a combination of reserpine and methacholine showed additive protective effects on submandibular gland function, signifying cooperation of muscarinic and alpha-adrenergic receptors.[74]
Phenylephrine
Isoproterenol
Methacholine or Methacholine + Phenylephrine
Pre-treatment with phenylephrine, isoproterenol and methacholine combined with phenylephrine resulted in less irradiation damage to parotid gland functions as indicated by quantified lag phase and flow rate.[75]
WR-2721WR-2721 provided a significant degree of protection for all glandular functional parameters including gland weight.[76]
cAMPThe demonstrated substantial protective effect of exogenously-administered cAMP on the parotid gland supports the previously-suggested radioprotection mechanism by the beta-adrenergic agonist isoproterenol, which is known to elevate endogenous intracellular cAMP.[77]
WR-2721
Isoproterenol
The aminothiol WR-2721 and beta-adrenergic agonist isoproterenol both conferred considerable radioprotection to the rat parotid gland. Isoproterenol acts on the beta-receptor, and its specific antagonist, propranolol, eliminated the protective effect of isoproterenol, thereby implicating the beta-receptor and cAMP in the radioprotection mechanism.[78]
WR-2721While non-protected glands suffered a drastic reduction in the amount of acinar tissue, ducts and blood vessels exhibited only minor morphological changes. Herein, WR-2721 protected the glands with similar signs of damage yet to a much lesser degree, in comparison.[79]
WR-2721WR-2721 protected against the acute phase of irradiation damage manifested during the first week post-RT. The drug also protected against chronic damage, appearing later.[80]
ThymolThymol at a dose of 50 mg/Kg significantly impacted (positively) salivary gland dysfunction caused by ionizing irradiation. Short- and late- side effects of RT on the salivary glands were considered reduced by Thymol in those rats.[81]
Rat
AgentMain FindingsRef
TLK1BAfter a single fraction of 16 Gy, the decline in salivary function at 8 weeks was less pronounced in TLK1B-treated animals (40%) when compared to saline-treated controls (67%).[82]
TLK1B associated with rAAV9AAV2/ 9-TLK1B groups showed no decline in salivary flow post-irradiation (121% increase) and salivary flow was not significantly different in irradiated and non-irradiated animals treated similarly with TLK1B.[83]

Table 2.

Radioprotection of salivary glands, in vivo using murine models.

RABBIT
AgentMain FindingsRef
Lidocaine HydroChloridePre-treatment with lidocaine improved irradiation tolerance of both, parotid and submandibular glands. Ultra-structure was largely preserved.[84]
Lidocaine
Amifostine
Pilocarpin
Only animals pre-treated with lidocaine or amifostine (alone or combined with pilocarpin) showed a slight non-significant reduction in the salivary ejection fraction. Lidocaine and amifostine could largely preserve the glandular ultra-structure.[85]
mini-PIG
AgentMain FindingsRef
Orciprenaline
Carbachol
Acinar cells of both glands were significantly more numerous in the pre-treatment group. Also, cells seemed better preserved. Yet, such effects were more pronounced in the parotid gland (appearing almost normal) than in the submandibular gland.[86]
Adenoviral vector encoding FGF2 (AdLTR2EF1a-FGF2)A single pre-administration of a hybrid serotype 5 adenoviral vector encoding FGF2 (AdLTR2EF1a-FGF2) resulted in the protection of parotid microvascular endothelial cells from irradiation damage and significantly limited the decline of parotid salivary flow.[87]

Table 3.

Radioprotection of salivary glands, in vivo using non-murine models.

AgentMain FindingsRef
WR-2721Administration of WR-2721 prior to each dose of irradiation was feasible and without significant toxicity at 100 mg/m2. Salivary gland function improved over time after completion of RT, particularly in the parotid gland.[88]
Botulinum Toxin A (BTX-A)The SUVmean of the 225Ac-labeled PSMA radio-ligand in the injected parotid gland (right) showed a highly significant decrease of up to 60% when compared with the left side in the 63 years old patient with advanced metastatic castration-resistant prostate cancer (suffering from sialorrhoea) receiving 80 units of BTX-A.[89]
AmifostineAmifostine reduces acute xerostomia and mucositis.[90]
SMGT+IMRTSurgical submandibular gland transfer (SMGT) was combined with intensity-modulated radiotherapy (IMRT) in a prospective phase II feasibility trial, in a single institution, including 40 HNC patients. At 12 months post-RT, the rate of absent or only mild xerostomia was 89%, and salivary flow rates were approximately 75% of pre-RT levels. Hence, patients reported decreased xerostomia and improved QoL.[91]
Helical Tomotherapy (HT)HT is described as an innovate, more precise and less toxic RT technique using a continuously rotating gantry to integrate 3D image guidance (a linear accelerator with computerized tomography) and deliver IMRT in a helical pattern. In 175 HNC patients, followed for up to 36 months, HT was used to deliver irradiation doses to bi-lateral parotid glands (PG-T), contra-lateral submandibular gland (cSMG), and accessory salivary glands in the oral cavity. Xerostomia was significantly decreased when the mean doses of PG-T, cSMG, and OC were kept below 29.12Gy, 29.29Gy, and 31.44Gy, respectively.[92]

Table 4.

Radioprotection of salivary glands, clinically in human subjects.

Figure 3.

PRISMA flow diagram for the bibliographic electronic search on PubMed central.

Palliative care for RT-induced salivary gland dysfunction- current and commercially-available palliative options for HNC patients undergoing RT include chewing gum (sugar-free), saliva substitutes, oral and topical lubricants, malic and ascorbic acid, saliva stimulants and sialogogue such as pilocarpine (Salagen, for example) and cevimeline (Evoxac, for example). As mentioned above, none have proved to restore normal QoL and patient satisfaction, mainly due to their limited efficacy and effectiveness [30, 42]. On top, adverse side effects are common, and such options are often costly to patients, requiring multiple daily use over long periods of time. In parallel, patients, especially the elderly, institutionalized and frail, need to go through education and training to acquire new eating and life-style habits, learn to prevent or avoid impaired swallowing and potential choking, and improve their oral and dental hygiene practices and tools to prevent (or halt the progression of) dental and oral mucosal diseases, infections and tooth loss. Other palliative care options including acupuncture and electro-stimulation (enhancement of salivary reflexes) are currently undergoing investigation [30, 93].

The only Food and Drug Administration–approved radioprotective and anti-xerostomia drug for clinical use (adjuvant setting) is Amifostine, an organic thiophosphate, cryoprotective agent and free radical scavenger administered subcutaneously or most often intravenously upon reconstitution with normal saline prior to or simultaneously with RT to then accumulate within the salivary glands, has been extensively-studied since its development, initially under the nuclear warfare program [14, 94]. Today, while it continues to benefit some patients, prophylactically, via minimizing the effects of xerostomia and taste loss, it is often associated with severe side effects including a rapid decrease in blood pressure (hypotension), nausea and emesis or vomiting. Recent analysis of several clinical trials associated Amifostine to low-quality and mixed evidence in preventing dry mouth complaints in patients receiving RT to the head and neck region, in the short- to medium-terms (up to three months post-RT) and have questioned its potential in tumor cell protection, thereby further narrowing its clinical safety and efficacy window, especially in light of its high cost [94, 95]. Essentially, its use in radiation-induced xerostomia has already been cautioned in the year 2008 by the American Society of Clinical Oncology [96], and so, its controversial and debatable safety and use in all cancer cases lingers.

Preventive and interventional care for RT-induced salivary gland dysfunction- the main objective of any planned and/or prescribed option should be the relief of symptoms and complications associated with hypo-salivation and xerostomia in HNC patients scheduled to receive RT, in order to prevent deteriorations in their QoL thereby enhancing their battle with cancer, its treatment and consequences [97]. As discussed earlier, despite advancements in irradiation techniques and regimens including IMRT, only palliative and prophylactic options are available, all of which do suffer substantial short-comings [98, 99]. One might even consider IMPT or intensity modulated proton therapy, used to deliver a much-reduced irradiation dose and subsequently less toxic than IMRT, thereby alleviating much of the typical side effects of RT, however, IMPT is known to be more expensive and lacks accessibility and availability [43, 98, 99].

  1. Surgical Intervention alternative- to prevent RT-induced hyposalivation, sub-mandibular gland preservation and protection from irradiation via surgical relocation to the sub-mental space, thereby away or out of irradiation zone, has been explored, with positive results. It is perhaps worth mentioning herein that sub-mandibular salivary gland supplies up to 90% of the un-stimulated saliva formation/secretion. However, such highly-invasive interventional procedures are peculiar and require exquisite surgical manipulation skills and settings. Further, surgical transfer of salivary glands is not indicated or possible for cancers of the oral cavity or patients undergoing (systemic) chemotherapy. In addition, for the gland to either retain or restore functionality, the connection of the gland to the main duct must be maintained or restored, respectively [100], altogether render it a very limit-ed/−ing option.

    In terms of innovative approaches, Rao et al. [101] recently described the use of a synthetic hydrogel (TraceIT, composed of water and iodinated cross-linked polyethylene glycol), injected via an 18-gauge needle, to serve as a minimally-invasive “spacer” (previously demonstrated in the treatment of prostate cancer), and displace or relocate the sub-mandibular gland in order to protect it from irradiation toxicity and be able to deliver a reduced irradiation dose, however the experimental model used comprised of four refrigerated cadaveric specimens and no further in vivo or clinical studies evaluated usability, malleability, safety and efficacy, amongst other factors, in clinical organ spacing.

  2. Tissue Engineering and Regenerative Medicine alternative- clearly, better approaches need to be explored and developed, driving the search elsewhere, into the multi-disciplinary areas of tissue engineering and regenerative medicine, in order to combine with and improve current options or to innovate and translate new alternative solutions, for wound healing. This is especially true, in light of accumulating knowledge and understanding of the underlying mechanisms governing radiation-induced salivary gland damage and dysfunction [47]. Indeed, from inducing DNA damage (via: a. the generation of ROS/reactive oxygen species or b. the breakage of the DNA double strand), to mutations to cell death (by apoptosis or necrosis, depending on cell type, injury and cellular responses), to the loss of salivary progenitors, to the accruing evidence regarding the regenerative capacity (slow yet existent) of salivary glands following RT-induced injury, more evidently upon the administration of a stimuli (exogenous delivery of stem cells and/or growth factors, for example), altogether re-emphasize the potential of such complex yet innovative approaches in finding a better clinical alternative solution.

    In a recent clinical study, Ho et al. [102] evaluated the effects of a commercially-available slowly-dissolving adhering disc/tablet formulation (OraCoat XyliMelts) on the oro-dental health, enamel remineralization, bio-film formation, saliva presence, pH and buffering in 5 patients diagnosed with xerostomia (criteria: un-stimulated whole saliva flow rate below 0.2 mL per minute and a stimulated saliva flow rate of less than 0.5 mL in 5 minutes). They also assessed patient self-reported comfort with the mint-flavored, xylitol-releasing tablets. Subjects were instructed to use the disc as often as needed for dry mouth symptoms relief. At the end, a mean of 4 + 1 discs each day and 2 discs each night, were used. Overall, desirable effects of the product on symptomatic alleviation and management of xerostomia were reported. The authors reported effective local palliation, reduced dental sensitivity, improved salivary production and buffering capacity, reduced plaque formation and alleviated xerostomia symptoms, without the need to use any systemic sialagogue medications throughout the 21 days of the study [102]. Yet, this is a pilot study, limited for involving a small of number of participants.

Biomaterials and Cell Therapy- one of the fundamental roles for the maintenance of the body of any living organism is regeneration, which enables the repair and restoration of lost or damaged tissue [47, 103]. Adult stem/stromal and progenitor cells have been identified in many tissues, and are known to have a key role in the regeneration and repair, initiated or activated either by the excessive loss of differentiated cells (pool) or via (niche) environmental cues. In the presence of functional biomaterials such as the previously-described injectable hydrogel spacer [101] and a feasible agent-delivery tablet or disc [102], would loading, encapsulating or incorporating putative salivary progenitor or stem/stromal cells, for example, a distinct type of stimuli, yield better results? Supplying salivary gland progenitor and stem/stromal cells, via a proper release-controlled dose-responsive carrier, might be able to re-establish the disrupted salivary stem/progenitor cell pool and niche, restore glandular tissue homeostasis, reverse hypo-salivation, and perhaps control xerostomia, a hypothesis we are currently examining in our laboratory, employing natural and synthetic polymers, liposomes, solid lipid nanoparticles and core-shell nanocapsules, and further supplementing by other pharmaceutical agents.

Modern medicine and biomedical research aim to control and enhance radioprotective as well as regenerative and reparative capabilities through the utilization of cells (cell lineages or primary cells), growing surface control using bio-scaffolds and/or manipulating growth factor/cytokine concentrations [47, 104], strategies designed to stimulate residual cells to regenerate acini and other parenchymal elements (ductal ligation) and infiltrate growth factor doses to boost salivary gland repair post-RT [105].

Growth Factor Therapy- somatomedin C is a hormone, similar to insulin in molecular structure, and actually is better known as IGF-1 or insulin-like growth factor 1 [106]. While a statement as “increased insulin-like growth factor signaling induces cell proliferation, survival and cancer progression” is true, it is traditional and partial, to a great extent. Today we understand that the issue is much more complex. For instance, IGF regulates cellular senescence which is known to halt proliferation of aged and stressed cells and do play a key role against cancer development. Actually, there is accruing evidence that, over time, IGF not only regulates but also induces pre-mature cellular senescence (tumor suppressor protein p53-dependant, in terms of acetylation, stabilization and activation) [107]. Hence, despite the understandably-alarming, at first and for some, suggestion to exogenously administer/supply cytokines and growth factors to sites of cancer, the recent years have indeed witnessed a note-worthy increase in the study of growth factors as cytoprotectants including their use as radioprotectors for salivary glands, and to reduce RT-induced symptoms, such as oral mucositis. To date, various growth factors have emerged as potential radioprotectors, including neurotrophic factors [108, 109], epidermal growth factor (EGF), fibroblast growth factor (FGF) [51, 110], keratinocyte growth factor (KGF) [111, 112] and the afore-mentioned insulin-like growth factor-1 or IGF-1 [55, 113, 114]. Meyer et al., [113], for example, investigated and determined the radioprotectant and therapeutic effect of IGF-1, in a murine model. They found that IGF-1 is mediated by the activation and maintenance of a histone deacetylase, specifically the Sirtuin 1 (SirT-1). Pre-treatment with IGF-1 enabled the repair of double-stranded breaks in the DNA of parotid salivary gland cells within the first hours post-irradiation, thereby allowing for optimal DNA repair (i.e. IGF-1 promotes DNA repair in irradiated parotid salivary glands via the maintenance and activation of SirT-1) to fulfill the cell cycle checkpoints. However, hours later and as early as 8 h, RT-induced apoptotic cells were detected [113]. Such observations lead to further study the signaling cross-talk between IGF-1 and SirT-1, thereby identifying several activators, stabilizers and inhibitors, including the afore-mentioned inhibition of the p53-mediated apoptosis and the phosphoinositide 3-kinase (PI3K) – protein kinase B (Akt) pathway [107], in-depth study-worthy topics, beyond the scope of this concise review. To date, studies, collectively indicate that cytokines can be radioprotective, anti-apoptotic and suggest/promote that the exogenous and localized (via a release-controlled delivery system, preferably directly injectable) utilization of growth factors do stimulate endogenous stem cell populations/niche and will eventually contribute to the desired and/or pursued clinical solution suitable for preventing RT-induced damage, diminishing salivary hypofunction, as well as restoring salivary gland function in irradiated HNC cases.

Gene Transfer Therapy- the utilization of gene transfer, DNA transmission and cell transduction to produce high levels of transgenic protein in order to correct cellular dysfunction and/or induce a new cellular function, post-RT, is a wide area of investigation and development. Baum et al. [115], utilized an adenoviral technique to transfer the Aquaporin-1 (AQP1) gene into the sub-mandibular gland, reporting an increase in salivary flow when compared to control viruses into rat or mini-pig models [115, 116]. Yet, key shortcomings continue to exist for non-viral as well as viral vectors [103], rendering translation for routine clinical use difficult. Likewise, the therapeutic potential of genetic modification and application of small-interfering RNAs or siRNA for the purpose of target gene silencing are intensively investigated, progressing from pre-clinical testing in animal models to ongoing clinical trials for cancer, lung disease and liver damage in human subjects. Thus far, highly limited in salivary gland tissues and accompanied with significant safety concerns [50]. For example, AQP-1 gene transfer into the salivary glands via adeno-viral vectors to treat disorders such as SS, yielded strong immune responses, mainly due to the limited or low efficiency of intra-cellular siRNA delivery [117, 118]. Herein, similar to growth factors, cell therapy and pharmaceutical agent administration, the availability of a reproducible, scalable, safe and effective, release-controlled carrier/vehicle suitable for therapeutic siRNA delivery, directly into the salivary gland, ensuring sufficient residency/retention, is a challenge.

Advertisement

5. Closing remarks

5.1 Wnt/β-catenin pathway: radio-protective role and effect in RT-induced salivary gland damage

In irradiation studies and radioprotection literature, numerous cellular signaling pathways and cell-cycle alteration mechanisms have been explored. Of those, the Wnt/β-catenin signaling pathway seems to receive the utmost attention, recently, towards preventing the damage caused by irradiation [119]. Briefly, this canonical Wingless–Int (Wnt) pathway leads to the accumulation and translocation of co-activator β-catenin, a multi-functional protein involved in cell–cell adhesion, gene transcription and physiologic homeostasis (adullt), into the nucleus, via a series of molecular events initiated through the binding of specific Wnt proteins to the frizzled receptors on the cell surface. The pathway plays a critical role in cell regulating cell migration and determining cell fate, and mutations have been linked to human birth defects, cancer and other disorders and diseases [120, 121, 122, 123].

Activating the canonical Wnt/β-catenin signaling pathway is complex. It depends on a family of glyco-proteins involved in cell-to-cell communication. To simplify, the interaction of ß-catenin with the cell adhesion molecule, e-cadherin, is involved in phenotypes: adhesion, mobility and proliferation [121, 122]. In absence of a Wnt ligand, β-catenin is degraded by the “destruction complex”. Several proteins are involved within this complex whereby Axin acts as a scaffold protein facilitating the interaction of Glycogen Synthase Kinase 3β (GSK-3β), Adenomatous Polyposis Coli (APC) and Casein Kinase 1α (CK1α), for β-catenin phosphorylation [123, 124]. Then, phosphorylated β-catenin is recognized by the β-transducin-repeat-containing protein (β-TrCP) and goes through the ubiquitin-proteasome degradation pathway. When the Wnt ligand activates Wnt signaling through the plasmatic membrane receptor frizzled with other lipoprotein receptors, the cytoplasmic protein disheveled (Dvl) is recruited and thereby activated. Herein, the activation of Dvl disrupts the “destruction complex” by dissociation of the GSK-3β from the Axin and inhibits the GSK-3β. As a result, β-catenin phosphorylation is also inhibited, allowing stabilization and translocation of β-catenin into the nucleus. Nuclear β-catenin then binds to a transcription factor-T cell factor and a lymphoid-enhancing factor (Tcf/Lef) and finally activates a response, i.e. changes in gene expression [120, 125, 126].

The Wnt signaling pathway cross-talks with other signaling pathways, and can be modulated by several activators and inhibitors. For example, the utilization of growth factors, to activate or inhibit, has been extensively studied, further adding to the complexity given the wide range of involved genes [119]. Cross-talk between signaling pathways is possible via the common regulatory protein GSK-3β. For example, when the epidermal growth factor (EGF) is recognized by its native receptor (EGF-R), this complex activates the afore-mentioned phosphoinositide 3-kinase (PI3K) which facilitates the activation of AKT kinase regulator. Herein, the activation of AKT results in the inhibition of GSK-3β by phosphorylation [127, 128, 129] and ultimately leads to the translocation of β-catenin into the nucleus. On the other hand, the fibroblast growth factor (FGF) is also able to cross-talk with GSK-3β (common pathway with EGF) and the activation of its native receptor (FGF-R) is followed by PI3K which then results in the inhibition of GSK-3β via AKT activation [125, 130]. Herein, FGF-R activation also involves MapK activation which inhibits GSK-3β through the p90 ribosomal protein s6 kinase (p90rsk) in an AKT-independent manner [131, 132, 133]. Therefore, activating the Wnt signaling pathway (Figure 4) through the utilization of cytoplasmic regulatory proteins (from other signaling pathways) is potentially able to promote β-catenin stabilization, its translocation to the nucleus and the activation of survival genes [134]. Such understanding and revelations can lead to produce a plausible and innovative alternative strategy for the activation of native repair systems that may allow and promote the survival of the cells during and after RT. Possibly, can be even extended to explore plausibility for prevention.

Figure 4.

EGF and FGF pathway(s) interaction with ß-catenin and canonical Wnt signaling pathway.

To the best of knowledge, Hakim et al. [135] conducted one of the first/earliest clinical studies connecting signaling pathways (Wnt/β-catenin and TGF-β) with salivary gland irradiation damage. They reported an alteration in the expression pattern of Wnt1 in viable irradiated acinar cells of xerostomic patients, suggesting a possible therapeutic effect of the Wnt pathway in controlling RT-induced salivary gland damage and dysfunction [135], in accordance with previous in vitro studies [120]. Following this line of research, Hai et al. [136] carried out a study analyzing the transient activation of the Wnt/β-catenin signaling pathway to prevent irradiation damage to the salivary glands. They reported, using a murine model, that activating the Wnt/β-catenin pathway through the transient activation of Wnt1 in the basal epithelium helped to prevent chronic salivary dysfunction generated by local irradiation, specifically via suppressing apoptosis and preserving or rescuing the life of salivary stem/progenitor cells. Salivation in experimental mice when compared to controls (animals receiving only RT) was increased/higher [120, 136]. However, the radioprotective effect of Wnt/β-catenin activation seems, thus far, to only occur within a limited time lapse. Activating the signaling path 3 days before or 3 days after irradiation yielded dissimilar effects on the tissues [136].

Indeed, in another approach, the activation and modulation of cell signaling pathway(s) using a cocktail (more than one) of activators has been suggested, with the Wnt signaling pathway (and its components) as therapeutic target(s). Thula et al. [51] evaluated the effect of EGF and bFGF (basic FGF) in salivary gland explants, reporting promising results regarding gland radioprotection [51]. Overall, taking the studied findings into account, it can be proposed that a Wnt/β-catenin signaling pathway activator might be a good candidate to be developed as a potential preventive and therapeutic strategy against the RT-induced salivary gland damage. Herein, as was and is the present scenario with cells, proteins, genes, growth factors and drugs, a suitable delivery vehicle is once more, deemed vital.

Technology Promise in Translational Tissue Engineering and NanoMedicine- the interplay between tissue engineering, regenerative medicine, biomaterials, bio-nanotechnology and nanomedicine continues to be the hallmark of current scientific research World-wide, promising to change every aspect of human life via creating revolutionary materials of biological origin for use in the diagnosis and treatment of devastating human diseases, a multi-disciplinary approach to innovative and translational solutions, suitable for scale-up, safe, efficacious and cost-effective routine clinical use [137, 138, 139]. Whether conventional small-molecule agents or emerging protein and/or peptide-based macromolecular biopharmaceutics, therapeutic effect is of vital significance. Controlled or at least predictable delivery is also substantially necessary. An intense effort is invested into engineering such complex bio-systems capable to achieve optimum cell-material interactions, while keeping intact the materials bulk properties. One of the core interests of nanobiotechnology, for example, this decade has been drug/gene/cell bio-functional delivery, driving the design and development of bio-inspired, intelligent or “smart” nano-systems [137, 138, 140]. It can be stated that a competitive and superiorly successful delivery system should offer: therapeutic outcome enhancement, patient compliance improvement and overall cost reduction of therapy. For HNC cases suffering RT-induced salivary gland damage and dysfunction, an attractive delivery system, for clinical ease-of-use, can perhaps entail a directly injectable formulation, sterilizable, capable to efficiently-hold a dose-responsive bio-load, maintain its bio-activity over time, and “predictably” control its pharmaco-kinetic release profile.

Advertisement

Funding and acknowledgments

This work was supported by generous funding and operating grants provided to the BioMAT’X R&D&I Group, part of CIIB (Centro de Investigación e Innovación Biomédica at UAndes), through the Faculty of Dentistry and Fondo de Ayuda a la Investigacion FAI - No. INV-IN-2015-101 (2015–2019), Department for Research, Development and Innovation, Universidad de los Andes, Santiago de Chile. The authors wish to acknowledge supplementary funding provided under the awarded national grants from CORFO-CTecnológicos para la Innovación #18COTE-89695 (the bioFLOSS project, 2018–2021) and CONICYT-FONDEF Chile #ID16I10366 (the maxSALIVA project, 2016/17–2020).

Advertisement

Conflict of interest

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. 1. Cancer, Fact Sheet, World Health Organization, available from https://www.who.int/news-room/fact-sheets/detail/cancer. Accessed Oct 23, 2020
  2. 2. International Agency for Research on Cancer (IARC). (2019). GLOBOCAN 2018, Cancer Incidence and Mortality Worldwide. Lyon: International Agency for Research on Cancer. Avaliable from http://gco.iarc.fr/today/fact-sheets-cancers. Accessed Aug 25, 2020
  3. 3. Yan, K., Agrawal, N., Gooi, Z. Head and Neck Masses. Med Clin North Am 2018,102,1013-1025. doi: 10.1016/j.mcna.2018.06.012
  4. 4. Cognetti, D. M., Weber, R. S., Lai, S. Y. Head and neck cancer: an evolving treatment paradigm. Cancer 2008, 113, 1911-1932, doi:10.1002/cncr.23654
  5. 5. Jaffray, D. A., Gospodarowicz, M. K. Radiation Therapy for Cancer, 2015, ISBN 9781464803499
  6. 6. Shetty, A. V, Wong, D. J. Systemic Treatment for Squamous Cell Carcinoma of the Head and Neck. Otolaryngol. Clin. North Am. 2017, 50, 775-782, doi:10.1016/j.otc.2017.03.013
  7. 7. Barazzuol, L., Coppes, R. P., van Luijk, P. Prevention and treatment of radiotherapy-induced side effects. Molecular oncology. 2020, 14, 1538-1554. https://doi.org/10.1002/1878-0261.12750
  8. 8. Gil, Z., Fliss, D. M. Contemporary management of head and neck cancers. Isr. Med. Assoc. J. 2009, 11, 296-300
  9. 9. Deloch, L., Derer, A., Hartmann, J., Frey, B., Fietkau, R., Gaipl, U. S. Modern Radiotherapy Concepts and the Impact of Radiation on Immune Activation. Front. Oncol. 2016, 6, 141, doi:10.3389/fonc.2016.00141
  10. 10. Baskar, R., Dai, J., Wenlong, N., Yeo, R., Yeoh, K.W. Biological response of cancer cells to radiation treatment. Front Mol Biosci 2014,1,24. doi:10.3389/fmolb.2014.00024
  11. 11. Manukian, G., Bar-Ad, V., Lu, B., Argiris, A., Johnson, J.M. Combining Radiation and Immune Checkpoint Blockade in the Treatment of Head and Neck Squamous Cell Carcinoma. Front Oncol 2019. doi:10.3389/fonc.2019.00122
  12. 12. Jensen, SB., Vissink, A., Limesand, K.H., Reyland, M.E. Salivary Gland Hypofunction and Xerostomia in Head and Neck Radiation Patients. J Natl Cancer Inst Monogr 2019, 53. doi: 10.1093/jncimonographs/lgz016
  13. 13. Wu, V. W. C., Leung, K. Y. A Review on the Assessment of Radiation Induced Salivary Gland Damage After Radiotherapy. Front Oncol 2019. doi:10.3389/fonc.2019.01090
  14. 14. Miranda-Rius, J., Brunet-Llobet, L., Lahor-Soler, E., Farré, M. Salivary Secretory Disorders, Inducing Drugs, and Clinical Management. Int. J. Med. Sci. 2015, 12, 811-824, doi:10.7150/ijms.12912
  15. 15. Ghannam, M. G., Singh, P. Anatomy, Head and Neck, Salivary Glands. StatPearls2019 Available from https://www.ncbi.nlm.nih.gov/books/NBK538325/. Accessed Jan 29, 2020
  16. 16. Punj, A. Secretions of Human Salivary Gland. Secretions of Human Salivary Gland, Salivary Glands - New Approaches in Diagnostics and Treatment, Işıl Adadan Güvenç, IntechOpen. 2018. doi: 10.5772/intechopen.75538. Available from: https://www.intechopen.com/books/salivary-glands-new-approaches-in-diagnostics-and-treatment/secretions-of-human-salivary-gland
  17. 17. Benn, A. M., Thomson, W. M. Saliva: an overview. N. Z. Dent. J. 2014, 110, 92-96
  18. 18. Tiwari, M. Science Behind Human Saliva. J Nat Sci Biol Med 2011, 2, 53-58. doi: 10.4103/0976-9668.82322
  19. 19. Proctor, G. B. The physiology of salivary secretion. Periodontol. 20002016, 70, 11-25, doi:10.1111/prd.12116
  20. 20. Qin, R., Steel, A., Fazel, N. Oral mucosa biology and salivary biomarkers. Clin. Dermatol. 2017, 35, 477-483, doi:10.1016/j.clindermatol.2017.06.005
  21. 21. Farnaud, S. J. C., Kosti, O., Getting, S. J., Renshaw, D. Saliva: physiology and diagnostic potential in health and disease. ScientificWorldJournal. 2010, 10, 434-456, doi:10.1100/tsw.2010.38
  22. 22. Fábián, T. K., Beck, A., Fejérdy, P., Hermann, P., Fábián, G. Molecular mechanisms of taste recognition: considerations about the role of saliva. Int. J. Mol. Sci. 2015, 16, 5945-5974, doi:10.3390/ijms16035945
  23. 23. von Bültzingslöwen, I., Sollecito, T. P., Fox, P. C., Daniels, T., Jonsson, R., Lockhart, P. B., Wray, D., Brennan, M. T., Carrozzo, M., Gandera, B., Fujibayashi, T., Navazesh, M., Rhodus, N. L., Schiødt, M. Salivary dysfunction associated with systemic diseases: systematic review and clinical management recommendations. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod.2007, 103 Suppl, S57.e1-15, doi:10.1016/j.tripleo.2006.11.010
  24. 24. Dashtipour, K., Bhidayasiri, R., Chen, J. J., Jabbari, B., Lew, M., Torres-Russotto, D. RimabotulinumtoxinB in sialorrhea: systematic review of clinical trials. J. Clin. Mov. Disord. 2017, 4, 9, doi:10.1186/s40734-017-0055-1
  25. 25. Jost, W.H., Friedman, A., Michel, O., Oehlwein, C., Slawek, J., Bogucki, A., Ochudlo, S., Banach, M., Pagan, F., Flatau-Baqué, B., Dorsch, U., Csikós, J., Blitzer, A. Long-term incobotulinumtoxinA treatment for chronic sialorrhea: Efficacy and safety over 64 weeks. Parkinsonism Relat Disord 2020, 70, 23-30. doi: 10.1016/j.parkreldis.2019.11.024
  26. 26. Frydrych, A. M. Dry mouth: Xerostomia and salivary gland hypofunction. Aust. Fam. Physician 2016, 45, 488-492
  27. 27. Azuma, N., Katada, Y., Kitano, S., Sekiguchi, M., Kitano, M., Nishioka, A., Hashimoto, N., Matsui, K., Iwasaki, T., Sano, H. Correlation between salivary epidermal growth factor levels and refractory intraoral manifestations in patients with Sjögren’s syndrome. Mod. Rheumatol. 2014, 24, 626-632, doi:10.3109/14397595.2013.850766
  28. 28. Millsop, J. W., Wang, E. A., Fazel, N. Etiology, evaluation, and management of xerostomia. Clin. Dermatol. 2017, 35, 468-476, doi:10.1016/j.clindermatol.2017.06.010
  29. 29. Tan, E. C. K., Lexomboon, D., Sandborgh-Englund, G., Haasum, Y., Johnell, K. Medications That Cause Dry Mouth As an Adverse Effect in Older People: A Systematic Review and Metaanalysis. J. Am. Geriatr. Soc. 2018, 66, 76-84, doi:10.1111/jgs.15151
  30. 30. Vissink, A., Mitchell, J. B., Baum, B. J., Limesand, K. H., Jensen, S. B., Fox, P. C., Elting, L. S., Langendijk, J. A., Coppes, R. P., Reyland, M. E. Clinical management of salivary gland hypofunction and xerostomia in head-and-neck cancer patients: successes and barriers. Int. J. Radiat. Oncol. Biol. Phys. 2010, 78, 983-991, doi:10.1016/j.ijrobp.2010.06.052
  31. 31. Schaue, D., Kachikwu, E. L., McBride, W. H. Cytokines in radiobiological responses: a review. Radiat. Res. 2012, 178, 505-523, doi:10.1667/RR3031.1
  32. 32. Williams, J. P., McBride, W. H. After the bomb drops: a new look at radiation-induced multiple organ dysfunction syndrome (MODS). Int. J. Radiat. Biol. 2011, 87, 851-868, doi:10.3109/09553002.2011.560996
  33. 33. Mohammadi, N., Seyyednejhad, F., Oskoee, P.A., Oskoee, S.S., Mofidi, N. Evaluation of Radiation-induced Xerostomia in Patients with Nasopharyngeal Carcinomas. J Dent Res Dent Clin Dent Prospects 2007, 1, 65-70. doi: 10.5681/joddd.2007.011
  34. 34. Strojan, P., Hutcheson, K. A., Eisbruch, A., Beitler, J. J., Langendijk, J. A., Lee, A. W. M., Corry, J., Mendenhall, W. M., Smee, R., Rinaldo, A., Ferlito, A. Treatment of late sequelae after radiotherapy for head and neck cancer. Cancer Treat. Rev. 2017, 59, 79-92, doi:10.1016/j.ctrv.2017.07.003
  35. 35. Franzén, L., Funegård, U., Ericson, T., Henriksson, R. Parotid gland function during and following radiotherapy of malignancies in the head and neck. A consecutive study of salivary flow and patient discomfort. Eur. J. Cancer1992, 28, 457-462
  36. 36. Siddiqui, F., Movsas, B. Management of Radiation Toxicity in Head and Neck Cancers. Semin. Radiat. Oncol. 2017, 27, 340-349, doi:10.1016/j.semradonc.2017.04.008
  37. 37. Berk, L. B., Shivnani, A. T., Small, W. Jr. Pathophysiology and management of radiation-induced xerostomia. J. Support. Oncol. 2005, 3, 191-200
  38. 38. Hammerlid, E., Silander, E., Hörnestam, L., Sullivan, M. Health-related quality of life three years after diagnosis of head and neck cancer--a longitudinal study. Head Neck 2001, 23, 113-125
  39. 39. Hall, S. C., Hassis, M. E., Williams, K. E., Albertolle, M. E., Prakobphol, A., Dykstra, A. B., Laurance, M., Ona, K., Niles, R. K., Prasad, N., Gormley, M., Shiboski, C., Criswell, L. A., Witkowska, H. E., Fisher, S. J. Alterations in the Salivary Proteome and N-Glycome of Sjögren’s Syndrome Patients. J. Proteome Res. 2017, 16, 1693-1705, doi:10.1021/acs.jproteome.6b01051
  40. 40. Jehmlich, N., Stegmaier, P., Golatowski, C., Salazar, M. G., Rischke, C., Henke, M., Völker, U. Differences in the whole saliva baseline proteome profile associated with development of oral mucositis in head and neck cancer patients undergoing radiotherapy. J. Proteomics 2015, 125, 98-103, doi:10.1016/j.jprot.2015.04.030
  41. 41. Thomson, W. M. Dry mouth and older people. Aust. Dent. J.2015, 60 Suppl 1, 54-63, doi:10.1111/adj.12284
  42. 42. Cereda, E., Cappello, S., Colombo, S., Klersy, C., Imarisio, I., Turri, A., Caraccia, M., Borioli, V., Monaco, T., Benazzo, M., Pedrazzoli, P., Corbella, F., Caccialanza, R. Nutritional counseling with or without systematic use of oral nutritional supplements in head and neck cancer patients undergoing radiotherapy. Radiother. Oncol. 2018, 126, 81-88, doi:10.1016/j.radonc.2017.10.015
  43. 43. Li, Y., Taylor, J. M. G., Ten Haken, R. K., Eisbruch, A. The impact of dose on parotid salivary recovery in head and neck cancer patients treated with radiation therapy. Int. J. Radiat. Oncol. Biol. Phys. 2007, 67, 660-669, doi:10.1016/j.ijrobp.2006.09.021
  44. 44. Jiang, N., Zhao, Y., Jansson, H., Chen, X., Mårtensson, J. Experiences of xerostomia after radiotherapy in patients with head and neck cancer: A qualitative study. J. Clin. Nurs. 2018, 27, e100–e108, doi:10.1111/jocn.13879
  45. 45. Wang, W., Xiong, W., Wan, J., Sun, X., Xu, H., Yang, X. The decrease of PAMAM dendrimer-induced cytotoxicity by PEGylation via attenuation of oxidative stress. Nanotechnology 2009, 20, 105103, doi:10.1088/0957-4484/20/10/105103
  46. 46. Nadig, S. D., Ashwathappa, D. T., Manjunath, M., Krishna, S., Annaji, A. G., Shivaprakash, P. K. A relationship between salivary flow rates and Candida counts in patients with xerostomia. J. Oral Maxillofac. Pathol. 2017, 21, 316, doi:10.4103/jomfp.JOMFP_231_16
  47. 47. Kagami, H., Wang, S., Hai, B. Restoring the function of salivary glands. Oral Dis. 2008, 14, 15-24, doi:10.1111/j.1601-0825.2006.01339.x
  48. 48. Villa, A., Abati, S. Risk factors and symptoms associated with xerostomia: a cross-sectional study. Aust. Dent. J. 2011, 56, 290-295, doi:10.1111/j.1834-7819.2011.01347.x
  49. 49. Bressan, V., Bagnasco, A., Aleo, G., Catania, G., Zanini, M. P., Timmins, F., Sasso, L. The life experience of nutrition impact symptoms during treatment for head and neck cancer patients: a systematic review and meta-synthesis. Support. Care Cancer 2017, 25, 1699-1712, doi:10.1007/s00520-017-3618-7
  50. 50. Grundmann, O., Mitchell, G. C., Limesand, K. H. Sensitivity of salivary glands to radiation: from animal models to therapies. J. Dent. Res. 2009, 88, 894-903, doi:10.1177/0022034509343143
  51. 51. Thula, T. T., Schultz, G., Tran-Son-Tay, R., Batich, C. Effects of EGF and bFGF on irradiated parotid glands. Ann. Biomed. Eng. 2005, 33, 685-695
  52. 52. Arany, S., Xu, Q., Hernady, E., Benoit, D. S. W., Dewhurst, S., Ovitt, C. E. Pro-apoptotic gene knockdown mediated by nanocomplexed siRNA reduces radiation damage in primary salivary gland cultures. J. Cell. Biochem. 2012, 113, 1955-1965, doi:10.1002/jcb.24064
  53. 53. Yoon, YJ., Shin, HS., Lim, JY. A hepatocyte growth factor/MET-induced antiapoptotic pathway protects against radiation-induced salivary gland dysfunction. Radiother Oncol. 2019, 138, 9-16. doi: 10.1016/j.radonc.2019.05.012
  54. 54. Tai, G., Zhang, H., Du, J., Chen, G., Huang, J., Yu, J., Cai, J., Liu, F. TIGAR overexpression diminishes radiosensitivity of parotid gland fibroblast cells and inhibits IR-induced cell autophagy. Int J Clin Exp Pathol. 2015, 8, 4823-4829
  55. 55. Choi, J.S., Shin, H.S., An, H.Y., Kim, Y.M., Lim, J.Y. Radioprotective effects of Keratinocyte Growth Factor-1 against irradiation-induced salivary gland hypofunction. Oncotarget 2017, 8, 13496-13508, doi:10.18632/oncotarget.14583
  56. 56. Wie, S. M., Wellberg, E., Karam, S. D., Reyland, M. E. Tyrosine Kinase Inhibitors Protect the Salivary Gland from Radiation Damage by Inhibiting Activation of Protein Kinase C-δ. Mol. Cancer Ther. 2017, 16, 1989-1998, doi:10.1158/1535-7163.MCT-17-0267
  57. 57. Choi, J.S., An, H.Y., Shin, H.S., Kim, Y.M., Lim, J.Y. Enhanced tissue remodelling efficacy of adipose-derived mesenchymal stem cells using injectable matrices in radiation-damaged salivary gland model. J. Tissue Eng. Regen. Med. 2018, 12, e695–e706, doi:10.1002/term.2352
  58. 58. Zeidan, Y. H., Xiao, N., Cao, H., Kong, C., Le, Q.T., Sirjani, D. Botulinum Toxin Confers Radioprotection in Murine Salivary Glands. Int. J. Radiat. Oncol. Biol. Phys. 2016, 94, 1190-1197, doi:10.1016/j.ijrobp.2015.12.371
  59. 59. An, H.Y., Shin, H.S., Choi, J.S., Kim, H. J., Lim, J.Y., Kim, Y.M. Adipose Mesenchymal Stem Cell Secretome Modulated in Hypoxia for Remodeling of Radiation-Induced Salivary Gland Damage. PLoS One 2015, 10, e0141862, doi:10.1371/journal.pone.0141862
  60. 60. Xu, L., Yang, X., Cai, J., Ma, J., Cheng, H., Zhao, K., Yang, L., Cao, Y., Qin, Q., Zhang, C., Zhang, Q., Sun, X. Resveratrol attenuates radiation-induced salivary gland dysfunction in mice. Laryngoscope 2013, 123, E23–E29, doi:10.1002/lary.24276
  61. 61. Okumura, H., Nasu, M., Yosue, T. Effects of amifostine administration prior to irradiation to the submandibular gland in mice: autoradiographic study using 3H-leucine. Okajimas Folia Anat. Jpn. 2009, 85, 151-160
  62. 62. Cotrim, A. P., Sowers, A., Mitchell, J. B., Baum, B. J. Prevention of irradiation-induced salivary hypofunction by microvessel protection in mouse salivary glands. Mol. Ther. 2007, 15, 2101-2106, doi:10.1038/sj.mt.6300296
  63. 63. Cotrim, A. P., Hyodo, F., Matsumoto, K., Sowers, A. L., Cook, J. A., Baum, B. J., Krishna, M. C., Mitchell, J. B. Differential radiation protection of salivary glands versus tumor by Tempol with accompanying tissue assessment of Tempol by magnetic resonance imaging. Clin. Cancer Res. 2007, 13, 4928-4933, doi:10.1158/1078-0432.CCR-07-0662
  64. 64. Cotrim, A. P., Sowers, A. L., Lodde, B. M., Vitolo, J. M., Kingman, A., Russo, A., Mitchell, J. B., Baum, B. J. Kinetics of tempol for prevention of xerostomia following head and neck irradiation in a mouse model. Clin. Cancer Res. 2005, 11, 7564-7568, doi:10.1158/1078-0432.CCR-05-0958
  65. 65. Aonuma, M., Nasu, M., Iwata, H., Yosue, T. Radioprotection of the murine submandibular gland by isoproterenol: autoradiography study with 3H-leucine. Odontology 2004, 92, 14-21, doi:10.1007/s10266-004-0032-7
  66. 66. Vitolo, J. M., Cotrim, A. P., Sowers, A. L., Russo, A., Wellner, R. B., Pillemer, S. R., Mitchell, J. B., Baum, B. J. The stable nitroxide tempol facilitates salivary gland protection during head and neck irradiation in a mouse model. Clin. Cancer Res. 2004, 10, 1807-1812
  67. 67. Rasey, J. S., Krohn, K. A., Menard, T. W., Spence, A. M. Comparative biodistribution and radioprotection studies with three radioprotective drugs in mouse tumors. Int. J. Radiat. Oncol. Biol. Phys. 1986, 12, 1487-1490
  68. 68. Rasey, J. S., Nelson, N. J., Mahler, P., Anderson, K., Krohn, K. A., Menard, T. Radioprotection of normal tissues against gamma rays and cyclotron neutrons with WR-2721: LD50 studies and 35S-WR-2721 biodistribution. Radiat. Res. 1984, 97, 598-607
  69. 69. Varghese J.J., Schmale I.L., Mickelsen D., Hansen M.E., Newlands S.D., Benoit D.S.W., Korshunov V.A., Ovitt C.E. Localized Delivery of Amifostine Enhances Salivary Gland Radioprotection. J Dent Res. 2018, doi: 10.1177/0022034518767408
  70. 70. Shin H.S., Lee S., Kim Y.M., Lim J.Y. Hypoxia-Activated Adipose Mesenchymal Stem Cells Prevents Irradiation-Induced Salivary Hypofunction by Enhanced Paracrine Effect Through Fibroblast Growth Factor 10. Stem Cells. 2018, doi: 10.1002/stem.2818
  71. 71. Toshkova, IA., Gleibermana, SA., Metta, VL., Hutsonb, AS., Singhc, AK., Gudkovde, AV., Burdelyad, LG. Mitigation of Radiation-Induced Epithelial Damage by the TLR5 Agonist Entolimod in a Mouse Model of Fractionated Head and Neck Irradiation. Radiat. Res. 2017, 187, 570-580
  72. 72. Xu, L., Yang, X., Chen, J., Ge, X., Qin, Q., Zhu, H., Zhang, C., Sun, X. Simvastatin attenuates radiation-induced salivary gland dysfunction in mice. Drug Des Devel Ther. 2016, 10, 2271-2278
  73. 73. Crescenti, E. J., Medina, V. A., Croci, M., Sambuco, L. A., Prestifilippo, J. P., Elverdin, J. C., Bergoc, R. M., Rivera, E. S. Radioprotection of sensitive rat tissues by oligoelements Se, Zn, Mn plus Lachesis muta venom. J. Radiat. Res. 2011, 52, 557-567
  74. 74. Coppes, R. P., Vissink, A., Zeilstra, L. J., Konings, A. W. Muscarinic receptor stimulation increases tolerance of rat salivary gland function to radiation damage. Int. J. Radiat. Biol. 1997, 72, 615-625
  75. 75. Coppes, R. P., Zeilstra, L. J., Vissink, A., Konings, A. W. Sialogogue-related radioprotection of salivary gland function: the degranulation concept revisited. Radiat. Res. 1997, 148, 240-247
  76. 76. Menard, T. W., Izutsu, K. T., Ensign, W. Y., Keller, P. J., Morton, T. H., Truelove, E. L. Radioprotection by WR-2721 of gamma-irradiated rat parotid gland: effect on gland weight and secretion at 8-10 days post irradiation. Int. J. Radiat. Oncol. Biol. Phys. 1984, 10, 1555-1559
  77. 77. Sodicoff, M., Conger, A. D. Radioprotection of the rat parotid gland by cAMP. Radiat. Res. 1983, 96, 90-94
  78. 78. Sodicoff, M., Conger, A. D. Radioprotection of the rat parotid gland by WR-2721 and isoproterenol and its modification by propranolol. Radiat. Res. 1983, 94, 97-104
  79. 79. Pratt, N. E., Sodicoff, M., Liss, J., Davis, M., Sinesi, M. Radioprotection of the rat parotid gland by WR-2721: morphology at 60 days post-irradiation. Int. J. Radiat. Oncol. Biol. Phys. 1980, 6, 431-435
  80. 80. Sodicoff, M., Conger, A. D., Pratt, N. E., Trepper, P. Radioprotection by WR-2721 against long-term chronic damage to the rat parotid gland. Radiat. Res. 1978, 76, 172-179
  81. 81. Abedi, SM., Yarmand, F., Motallebnejad, M., Seyedmajidi, M., Moslemie, D., Bijanif, A., Hosseinimehrg, SJ. Radioprotective Effect of Thymol Against Salivary Glands Dysfunction Induced by Ionizing Radiation in Rats. Iran J Pharm Res. 2016, 15, 861-866
  82. 82. Palaniyandi, S., Odaka, Y., Green, W., Abreo, F., Caldito, G., De Benedetti, A., Sunavala-Dossabhoy, G. Adenoviral delivery of Tousled kinase for the protection of salivary glands against ionizing radiation damage. Gene Ther. 2011, 18, 275-282
  83. 83. Shanmugam, PST., Dayton, RD., Palaniyandi, S., Abreo, F., Caldito, G., Klein, RL., Sunavala-Dossabhoy. Recombinant AAV9-TLK1B Administration Ameliorates Fractionated Radiation-Induced Xerostomia. Hum Gene Ther. 2013, 24, 604-612
  84. 84. Hakim, S.G., Benedek, G. A., Su, Y.X., Jacobsen, H.C., Klinger, M., Dendorfer, A., Hemmelmann, C., Meller, B., Nadrowitz, R., Rades, D., Sieg, P. Radioprotective effect of lidocaine on function and ultrastructure of salivary glands receiving fractionated radiation. Int. J. Radiat. Oncol. Biol. Phys. 2012, 82, e623–e630, doi:10.1016/j.ijrobp.2011.09.017
  85. 85. Hakim, S. G., Kosmehl, H., Lauer, I., Nadrowitz, R., Wedel, T., Sieg, P. A comparative study on the protection profile of lidocaine, amifostine, and pilocarpin on the parotid gland during radiotherapy. Cancer Res 2005, 65, 10486-10493, doi:10.1158/0008-5472.CAN-05-0023
  86. 86. Lotz, S., Caselitz, J., Tschakert, H., Rehpenning, W., Seifert, G. Radioprotection of minipig salivary glands by orciprenaline-carbachol. An ultrastructural and semiquantitative light microscopic study. Virchows Arch. A. Pathol. Anat. Histopathol. 1990, 417, 119-128
  87. 87. Guo, L., Gao, R., Xu, J., Jin, L., Cotrim, AP., Yan, X., Zheng, C., Goldsmith, CM., Shan, Z., Hai, B., Zhou, J., Zhang, C., Baum, BJ., Wang, S. AdLTR2EF1α-FGF2-mediated prevention of fractionated irradiation-induced salivary hypofunction in swine. Gene Ther. 2014, 21, 866-873
  88. 88. McDonald, S., Meyerowitz, C., Smudzin, T., Rubin, P. Preliminary results of a pilot study using WR-2721 before fractionated irradiation of the head and neck to reduce salivary gland dysfunction. Int. J. Radiat. Oncol. Biol. Phys. 1994, 29, 747-754
  89. 89. Baum, R. P., Langbein, T., Singh, A., Shahinfar, M., Schuchardt, C., Volk, G. F., Kulkarni, H. Injection of Botulinum Toxin for Preventing Salivary Gland Toxicity after PSMA Radioligand Therapy: an Empirical Proof of a Promising Concept. Nucl. Med. Mol. Imaging 2018, 52, 80-81, doi:10.1007/s13139-017-0508-3
  90. 90. Vacha, P., Fehlauer, F., Mahlmann, B., Marx, M., Hinke, A., Sommer, K., Richter, E., Feyerabend, T. Randomized phase III trial of postoperative radiochemotherapy +/− amifostine in head and neck cancer. Is there evidence for radioprotection? Strahlenther. Onko. 2003, 179, 385-389, doi:10.1007/s00066-003-1016-1
  91. 91. Scrimger, R.A., Seikaly, H., Vos, L.J., Harris, J., O'Connell, D., Ghosh, S., Debenham, B., Jha, N. Combination of submandibular salivary gland transfer and intensity-modulated radiotherapy to reduce dryness of mouth (xerostomia) in patients with head and neck cancer. Head Neck 2018, 40:2353-2361. doi: 10.1002/hed.25339
  92. 92. Teng, F., Fan, W., Luo, Y., Ju, Z., Gong, H., Ge, R., Tong, F., Zhang, X., Ma, L. Reducing Xerostomia by Comprehensive Protection of Salivary Glands in Intensity-Modulated Radiation Therapy with Helical Tomotherapy Technique for Head-and-Neck Cancer Patients: A Prospective Observational Study. Biomed Res Int2019, 14, 2019:2401743. doi: 10.1155/2019/2401743
  93. 93. de Castro, G. Jr, Guindalini, R. S. Supportive care in head and neck oncology. Curr. Opin. Oncol. 2010, 22, 221-225, doi:10.1097/CCO.0b013e32833818ff
  94. 94. Gu, J., Zhu, S., Li, X., Wu, H., Li, Y., Hua, F. Effect of amifostine in head and neck cancer patients treated with radiotherapy: a systematic review and meta-analysis based on randomized controlled trials. PLoS One 2014, 9, e95968, doi:10.1371/journal.pone.0095968
  95. 95. Riley, P., Glenny, A.M., Hua, F., Worthington, H.V. Pharmacological interventions for preventing dry mouth and salivary gland dysfunction following radiotherapy. Cochrane database Syst. Rev. 2017, 7, CD012744, doi:10.1002/14651858.CD012744
  96. 96. The American Society of Clinical Oncology. Clinical Practice Guideline Update: Use of Chemotherapy and Radiation Therapy Protectants. J Oncol Pract2008, 4, 277-279. Written by: Hensley, M.L., Hagerty, K.L., Kewalramani, T., Green, D.M., Meropol, N. J., Wasserman, T.H., Cohen, G.I., Emami, B., Gradishar, W.J., Mitchell, R.B., Thigpen, J.T., Trotti, A., von Hoff, D., Schuchter, L. M. doi: 10.1200/JOP.0868502
  97. 97. Vissink, A., Jansma, J., Spijkervet, F. K., Burlage, F. R., Coppes, R. P. Oral sequelae of head and neck radiotherapy. Crit. Rev. Oral Biol. Med. 2003, 14, 199-212
  98. 98. Braam, P. M., Terhaard, C. H., Roesink, J. M., Raaijmakers, C. P. Intensity-modulated radiotherapy significantly reduces xerostomia compared with conventional radiotherapy. Int. J. Radiat. Oncol. Biol. Phys. 2006, 66, 975-980, doi:10.1016/j.ijrobp.2006.06.045
  99. 99. Teng, F., Fan, W., Luo, Y., Ju, Z., Gong, H., Ge, R., Tong, F., Zhang, X., Ma, L. Reducing Xerostomia by Comprehensive Protection of Salivary Glands in Intensity-Modulated Radiation Therapy with Helical Tomotherapy Technique for Head-and-Neck Cancer Patients: A Prospective Observational Study. Biomed Res Int 2019. doi: https://doi.org/10.1155/2019/2401743
  100. 100. Marzouki, H. Z., Elkhalidy, Y., Jha, N., Scrimger, R., Debenham, B. J., Harris, J. R., O’Connell, D. A., Seikaly, H. Modification of the submandibular gland transfer procedure. Laryngoscope 2016, 126, 2492-2496, doi:10.1002/lary.26029
  101. 101. Rao, A. D., Coquia, S., De Jong, R., Gourin, C., Page, B., Latronico, D., Dah, S., Su, L., Clarke, S., Schultz, J., Rosati, L. M., Fakhry, C., Wong, J., DeWeese, T. L., Quon, H., Ding, K., Kiess, A. Effects of biodegradable hydrogel spacer injection on contralateral submandibular gland sparing in radiotherapy for head and neck cancers. Radiother. Oncol. 2018, 126, 96-99, doi:10.1016/j.radonc.2017.09.017
  102. 102. Ho, J., Firmalino, M. V., Anbarani, A. G., Takesh, T., Epstein, J., Wilder-Smith, P. Effects of A Novel Disc Formulation on Dry Mouth Symptoms and Enamel Remineralization in Patients With Hyposalivation: An In Vivo Study. Dent. (Sunnyvale, Calif.)2017, 7, doi:10.4172/2161-1122.1000411
  103. 103. Ogawa, M., Oshima, M., Imamura, A., Sekine, Y., Ishida, K., Yamashita, K., Nakajima, K., Hirayama, M., Tachikawa, T., Tsuji, T. Functional salivary gland regeneration by transplantation of a bioengineered organ germ. Nat. Commun. 2013, 4, 2498, doi:10.1038/ncomms3498
  104. 104. Zhang, N.N., Huang, G.L., Han, Q.B., Hu, X., Yi, J., Yao, L., He, Y. Functional regeneration of irradiated salivary glands with human amniotic epithelial cells transplantation. Int. J. Clin. Exp. Pathol. 2013, 6, 2039-2047
  105. 105. Okazaki, Y., Kagami, H., Hattori, T., Hishida, S., Shigetomi, T., Ueda, M. Acceleration of rat salivary gland tissue repair by basic fibroblast growth factor. Arch. Oral Biol. 2000, 45, 911-919
  106. 106. Michalopoulou, F., Petraki, C., Philippou, A., Analitis, A., Msaouel, P., Koutsilieris, M. Expression of IGF-IEc Isoform in Renal Cell Carcinoma Tissues. Anticancer Res 2020, 40, :6213-6219. doi: 10.21873/anticanres.14641
  107. 107. Tran, D., Bergholz, J., Zhang, H., He, H., Wang, Y., Zhang, Y., Li, Q., Kirkland, J. L., Xiao, Z. X. Insulin-like growth factor-1 regulates the SIRT1-p53 pathway in cellular senescence. Aging cell 2014, 13, 669-678. https://doi.org/10.1111/acel.12219
  108. 108. Xiao, N., Lin, Y., Cao, H., Sirjani, D., Giaccia, A. J., Koong, A. C., Kong, C. S., Diehn, M., Le, Q.T. Neurotrophic factor GDNF promotes survival of salivary stem cells. J. Clin. Invest. 2014, 124, 3364-3377, doi:10.1172/JCI74096
  109. 109. Swick, A., Kimple, R. J. Wetting the whistle: neurotropic factor improves salivary function. J. Clin. Invest. 2014, 124, 3282-3284, doi:10.1172/JCI77194
  110. 110. Kojima, T., Kanemaru, S., Hirano, S., Tateya, I., Suehiro, A., Kitani, Y., Kishimoto, Y., Ohno, S., Nakamura, T., Ito, J. The protective efficacy of basic fibroblast growth factor in radiation-induced salivary gland dysfunction in mice. Laryngoscope 2011, 121, 1870-1875, doi:10.1002/lary.21873
  111. 111. Borges, L., Rex, K. L., Chen, J. N., Wei, P., Kaufman, S., Scully, S., Pretorius, J. K., Farrell, C. L. A protective role for keratinocyte growth factor in a murine model of chemotherapy and radiotherapy-induced mucositis. Int. J. Radiat. Oncol. Biol. Phys. 2006, 66, 254-262, doi:10.1016/j.ijrobp.2006.05.025
  112. 112. Lombaert, I. M., Brunsting, J. F., Wierenga, P. K., Kampinga, H. H., de Haan, G., Coppes, R. P. Keratinocyte growth factor prevents radiation damage to salivary glands by expansion of the stem/progenitor pool. Stem Cells 2008, 26, 2595-2601, doi:10.1634/stemcells.2007-1034
  113. 113. Meyer, S., Chibly, A. M., Burd, R., Limesand, K. H. Insulin-Like Growth Factor-1-Mediated DNA Repair in Irradiated Salivary Glands Is Sirtuin-1 Dependent. J. Dent. Res. 2017, 96, 225-232, doi:10.1177/0022034516677529
  114. 114. Grundmann, O., Fillinger, J. L., Victory, K. R., Burd, R., Limesand, K. H. Restoration of radiation therapy-induced salivary gland dysfunction in mice by post therapy IGF-1 administration. BMC Cancer 2010, 10, 417, doi:10.1186/1471-2407-10-417
  115. 115. Baum, B. J., Zheng, C., Cotrim, A. P., Goldsmith, C. M., Atkinson, J. C., Brahim, J. S., Chiorini, J. A., Voutetakis, A., Leakan, R. A., Van Waes, C., Mitchell, J. B., Delporte, C., Wang, S., Kaminsky, S. M., Illei, G. G. Transfer of the AQP1 cDNA for the correction of radiation-induced salivary hypofunction. Biochim. Biophys. Acta 2006, 1758, 1071-1077, doi:10.1016/j.bbamem.2005.11.006
  116. 116. Redman, R. S. On approaches to the functional restoration of salivary glands damaged by radiation therapy for head and neck cancer, with a review of related aspects of salivary gland morphology and development. Biotech. Histochem. 2008, 83, 103-130, doi:10.1080/10520290802374683
  117. 117. Cotrim, A. P., Sowers, A., Mitchell, J. B., Baum, B. J. Prevention of irradiation-induced salivary hypofunction by microvessel protection in mouse salivary glands. Mol. Ther. 2007, 15, 2101-2106, doi:10.1038/sj.mt.6300296
  118. 118. Guo, L., Gao, R., Xu, J., Jin, L., Cotrim, AP., Yan, X., Zheng, C., Goldsmith, CM., Shan, Z., Hai, B., Zhou, J., Zhang, C., Baum, BJ., Wang, S. AdLTR2EF1α-FGF2-mediated prevention of fractionated irradiation-induced salivary hypofunction in swine. Gene Ther. 2014, 21, 866-873
  119. 119. Song, G., Ouyang, G., Bao, S. The activation of Akt/PKB signaling pathway and cell survival. J. Cell. Mol. Med. 2005,9, 59-71
  120. 120. Wang, J.F., Liu, C., Zhang, Q., Huang, G.H. Research progress in the radioprotective effect of the canonical Wnt pathway. Cancer Biol. Med. 2013, 10, 61-71, doi:10.7497/j.issn.2095-3941.2013.02.001
  121. 121. Vidya Priyadarsini, R., Senthil Murugan, R., Nagini, S. Aberrant activation of Wnt/β-catenin signaling pathway contributes to the sequential progression of DMBA-induced HBP carcinomas. Oral Oncol. 2012, 48, 33-39, doi:10.1016/j.oraloncology.2011.08.008
  122. 122. Huang, J.;Qu, Q., Guo, Y., Xiang, Y., Feng, D. Tankyrases/β-catenin Signaling Pathway as an Anti-proliferation and Anti-metastatic Target in Hepatocarcinoma Cell Lines. J Cancer 2020, 11, 432-440. doi: 10.7150/jca.30976
  123. 123. Orme, M. H., Giannini, A. L., Vivanco, M. D., Kypta, R. M. Glycogen synthase kinase-3 and Axin function in a beta-catenin-independent pathway that regulates neurite outgrowth in neuroblastoma cells. Mol. Cell. Neurosci. 2003, 24, 673-686
  124. 124. Garan, A., Akyüz, S., Oztürk, L. K., Yarat, A. Salivary parameters and caries indices in children with black tooth stains. J. Clin. Pediatr. Dent. 2012, 36, 285-288
  125. 125. Nusse, R., Clevers, H. Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017, 169, 985-999, doi:10.1016/j.cell.2017.05.016
  126. 126. Huang, H., He, X. Wnt/beta-catenin signaling: new (and old) players and new insights. Curr. Opin. Cell Biol. 2008, 20, 119-125, doi:10.1016/j.ceb.2008.01.009
  127. 127. Doble, B. W., Woodgett, J. R. GSK-3: tricks of the trade for a multi-tasking kinase. J. Cell Sci. 2003, 116, 1175-1186
  128. 128. Cross, D. A., Alessi, D. R., Cohen, P., Andjelkovich, M., Hemmings, B. A. Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature 1995,378, 785-789, doi:10.1038/378785a0
  129. 129. Krasilnikov, M. A. Phosphatidylinositol-3 kinase dependent pathways: the role in control of cell growth, survival, and malignant transformation. Biochemistry. (Mosc). 2000, 65, 59-67
  130. 130. Huang, L., Fu, L. Mechanisms of resistance to EGFR tyrosine kinase inhibitors. Acta Pharm. Sin. B 2015, 5, 390-401, doi:10.1016/j.apsb.2015.07.001
  131. 131. Torres, M. A., Eldar-Finkelman, H., Krebs, E. G., Moon, R. T. Regulation of ribosomal S6 protein kinase-p90(rsk), glycogen synthase kinase 3, and beta-catenin in early Xenopus development. Mol. Cell. Biol. 1999, 19, 1427-1437
  132. 132. Dailey, L., Ambrosetti, D., Mansukhani, A., Basilico, C. Mechanisms underlying differential responses to FGF signaling. Cytokine Growth Factor Rev. 2005, 16, 233-247, doi:10.1016/j.cytogfr.2005.01.007
  133. 133. Alcaraz, E., Vilardell , J., Borgo, C., Sarró, E., Plana, M., Marin, O., Pinna, L.A., Bayascas, J.R., Meseguer, A., Salvi, M., Itarte, E., Ruzzene, F. Effects of CK2β subunit down-regulation on Akt signalling in HK-2 renal cells. PLoS One 2020. doi: https://doi.org/10.1371/journal.pone.0227340
  134. 134. Kennedy, S. G., Wagner, A. J., Conzen, S. D., Jordán, J., Bellacosa, A., Tsichlis, P. N., Hay, N. The PI 3-kinase/Akt signaling pathway delivers an anti-apoptotic signal. Genes Dev. 1997, 11, 701-713
  135. 135. Hakim, S. G., Ribbat, J., Berndt, A., Richter, P., Kosmehl, H., Benedek, G. A., Jacobsen, H. C., Trenkle, T., Sieg, P., Rades, D. Expression of Wnt-1, TGF-β and related cell-cell adhesion components following radiotherapy in salivary glands of patients with manifested radiogenic xerostomia. Radiother. Oncol. 2011, 101, 93-99, doi:10.1016/j.radonc.2011.07.032
  136. 136. Hai, B., Yang, Z., Shangguan, L., Zhao, Y., Boyer, A., Liu, F. Concurrent transient activation of Wnt/β-catenin pathway prevents radiation damage to salivary glands. Int. J. Radiat. Oncol. Biol. Phys. 2012, 83, e109–e116, doi:10.1016/j.ijrobp.2011.11.062
  137. 137. Haidar, Z.S. Bio-Inspired/-Functional Colloidal Core-Shell Polymeric-Based NanoSystems: Technology Promise in Tissue Engineering, Bioimaging and NanoMedicine. Polymers 2010, 2, 323-352. https://doi.org/10.3390/polym2030323
  138. 138. Riley, P., Glenny, A.M., Hua, F., Worthington, H.V. Pharmacological interventions for preventing dry mouth and salivary gland dysfunction following radiotherapy. Cochrane Database of Systematic Reviews 2017, 7. doi: 10.1002/14651858.CD012744
  139. 139. Ocampo, J., Vásquez, B., Sandoval, C., Navarrete, J., Haidar, Z.S., Olate, S. Características Morfocuantitativas de la Glándula Submandibular de Ratón (Mus musculus)/ Morphocuantitative Characteristics of the Mouse (Mus musculus) Submandibular Gland. International Journal of Morphology 2020, 38, 570-577. https://dx.doi.org/10.4067/S0717-95022020000300570
  140. 140. Ocampo, J., Olate, S., Haidar, Z.S., Vásquez, B. Hiposialia y Xerostomía Post Irradiación: Terapias Innovadoras en el Campo Biomolecular/Hyposialia and Xerostomy Post Irradiation: Innovative Therapies in the Biomolecular Field. International Journal of Morphology 2019, 37, 1564-1571. https://dx.doi.org/10.4067/S0717-95022019000401564

Written By

Ziyad S. Haidar

Submitted: 25 September 2020 Reviewed: 04 November 2020 Published: 11 January 2021