Next Article in Journal
A Novel Hemocyte-Specific Small Protein Participates in White Spot Syndrome Virus Infection via Binding to Viral Envelope Protein
Next Article in Special Issue
Viral Diseases in Dermatology
Previous Article in Journal
West Nile Virus in Culex Mosquitoes in Central Macedonia, Greece, 2022
Previous Article in Special Issue
An Improved Protocol for Comprehensive Etiological Characterization of Skin Warts and Determining Causative Human Papillomavirus Types in 128 Histologically Confirmed Common Warts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Comprehensive Overview of Epidemiology, Pathogenesis and the Management of Herpes Labialis

1
Basic Medical and Dental Sciences Department, Ajman University, Ajman P.O. Box 346, United Arab Emirates
2
Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman P.O. Box 346, United Arab Emirates
3
School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia
4
School of Pharmacy, University of Nottingham Malaysia, Semenyih 43500, Malaysia
5
Department of Oral Pathology and Microbiology, Institute of Dental Sciences, Siksha‘O’Anusandhan Deemed to be University, Bhubaneswar 751030, India
*
Author to whom correspondence should be addressed.
Viruses 2023, 15(1), 225; https://doi.org/10.3390/v15010225
Submission received: 29 November 2022 / Revised: 3 January 2023 / Accepted: 8 January 2023 / Published: 13 January 2023
(This article belongs to the Special Issue Viral Diseases in Dermatology)

Abstract

:
Herpes labialis remains exceedingly prevalent and is one of the most common human viral infections throughout the world. Recurrent herpes labialis evolves from the initial viral infection by herpes simplex virus type 1 (HSV-1) which subsequently presents with or without symptoms. Reactivation of this virus is triggered by psychosocial factors such as stress, febrile environment, ultraviolet light susceptibility, or specific dietary inadequacy. This virus infection is also characterized by uninterrupted transitions between chronic-latent and acute-recurrent phases, allowing the virus to opportunistically avoid immunity and warrant the transmission to other vulnerable hosts simultaneously. This review comprehensively evaluates the current evidence on epidemiology, pathogenesis, transmission modes, clinical manifestations, and current management options of herpes labialis infections.

1. Introduction

Herpes labialis is an infection with herpes simplex virus type 1 (HSV-1) with initial episodes presenting as asymptomatic or symptomatic small blisters or sores on the skin near the site of infection. When the initial infection heals, the virus spreads to sensory nerve cells, where it remains dormant until reactivation occurs [1]. Recurrence of labial HSV is typically triggered by factors such as stress, trauma from surgery, menstruation or hormonal shifts in women, hyperthermia or infectious febrile ailments, sunlight or ultraviolet rays, and certain drugs, such as corticosteroids, with substantial psychosocial consequences to patients enduring persistent outbreaks [2].
The regularity of these recurrent outbreaks varies from one individual to another where the infection is generally contagious during times of active replication, but can also spread when symptoms are absent [1]. The disease progression in recurrent herpes simplex labialis (HSL) typically incorporates multiple stages. During the precursor stage, perceptions of pain, tingling, or burning may transpire in the affected area, followed by the development of vesicles. Rupturing of the vesicle leads to soft scab formation which is subsequently replaced by a hard scab. Periodically, the scab abates and falls off, allowing the lesion to completely heal without scarring. Throughout this healing process, symptoms such as pain and discomfort ensue, and complete healing may require seven to ten days [1,2,3,4].

2. Epidemiology

Oral HSV-1 infection continually remains exceedingly prevalent and is one of the most common human viral infections throughout the world. According to WHO, an estimated 3.7 billion people under age 50 (67%) have HSV-1 infection globally [5]. In the United States of America (USA), HSV-1 affects 57% to 80% of adults [6], while in Asia, the numbers are essentially high for adults (75%) especially those from low socioeconomic standings, and children (50%) due to epidemiological shifts, showing decreased seroprevalence in the young cohorts [7]. A reported estimate in 2016 revealed that around 3.583 billion were infected with oral HSV totaling a prevalence of 63.6% (95% uncertainty interval (UI): UI: 59.0–66.0) and the majority come from Southeast Asia region followed by the Western Pacific region [8]. It has been reported that recurrent herpes labialis (RHL) affects about one-third of the population in the USA and presents as inflamed/painful oral lesions with conditional distress. Some patients usually encounter up to six episodes per year which can be troublesome for patients and their families [6,9]. Overall, the average incidence of RHL is about 1.6 per 1000 patients each year with a prevalence of 2.5 per 1000 patients but varies significantly between countries and different communities, including more frequency in females where an estimated one-third of all affected patients experience at least one relapse per year [9]. In a similar study in Sweden, the lifetime encounters with symptomatic herpes labialis among 3597 participants were about 40% [10]. A cross-sectional survey among American college students reported HSV-1 antibodies’ prevalence of 37.2% in fresh graduates and 46.1% in final-year students, implicating a reduction in seroprevalence of HSV-1 from 62.0% (1988 to 1994) to 57.7% (1999 to 2004) [11] due to awareness.

3. Viral Pathogenesis

3.1. Pathogenetic or Symbiotic?

The natural history of oral herpes is described as human–virus symbiosis which converts reversibly from mutualism to parasitism in a balanced way and this co-existence occurs between superiorly co-evolved microorganisms with humans [12,13,14,15]. While symbiosis implies close acquaintance between two species that mutually provide benefits to both, the sophisticated nature of the virus to invade the host and evade the immune-responsive defense thus inflicting chronic infections suggests a “war-metaphor” which connotes more to a parasitic association where only the microorganism is provided with the benefit. Depending on the risk factors involved in the recurrence of HSV-1 oral infections, it is crucial to understand that there are no pure parasitic or mutualistic relationships between humans and HSV-1 [12].

3.2. Transmission & Infection

The majority of the primary infections result from direct exposure to bodily fluids such as saliva or exudates of progressive lesions, and proximate contact with lesions of infected individuals [16]. In addition, the transmission of the virus can also occur via kissing or sharing of towels/utensils. Usually, the primary infection emerges between 2 to 20 days following contact [5]. Approximately 2–10% of these infections also involve asymptomatic patients who periodically undergo HSV shedding in their saliva which is profound in immunocompromised individuals with an estimate of 38–40% [4,5,7,16]. Extensively, the asymptomatic HSV shedding is primarily reflected throughout the prodrome phase after infection, and 60% of these patients may not proceed to vesicle development even with the common symptoms of fever and pain [17].
At the contact site, HSV-1 primary infections are initiated after penetration of the tissue by the viruses and fusion of the viral envelope with the epidermal and dermal skin membranes (cellular and mucosal) [16]. Infection of the skin or the mucosa is accompanied by inflammation and tissue damage, causing the characteristic herpes blisters. Following efficient replication in epithelial cells, HSV reaches nerve endings of peripheral neurons and undergoes retrograde transport to the neuronal cell body [18]. Due to the breach of the immune system, the host cell’s retaliation to this viral suppression is disrupted, consequently evading immune responses with the complex reciprocation of the virion protein also known as “virion host shutoff” [16].

3.3. Latency and Reactivation

HSV often results in either lytic or latent infection. There is minimal gene expression and no viral particle generation during latency. The viral genome is capable of reactivation, which results in the creation of infectious virions in response to the right stimuli [19,20,21]. Sensory neurons harboring the virus contain nuclear transcripts known as latency-associated transcripts (LATs), responsible for modifying viral genome chromatins which generate a stable but reversible silencing of the HSV genome [21,22,23]. LATs are approximately 8.3 kb long noncoding RNA that expresses in the nucleus of latently infected cells [21,22,23,24,25]. Under latent conditions, LATs remain highly expressed and silence the immediate early (IE) gene expression, which are major genes in the lytic cycle [24,25,26]. LATs have a role in protecting the latently infected neurons from undergoing apoptosis in the epigenetic modification of histones to regulate the expression of the latent genes [27].
The HSV infection in the body of neurons usually results in infectious viral particles, while infection at the axons fails to produce infectious particles, especially when the infectious titer is low. However, it has been shown that axonal infection can lead to the production of an infectious virus if the cell body area is added with a helper virus with VP16, a tegument protein [28,29]. Moreover, it has been demonstrated that low levels of tegument proteins VP16 and ICP0 might lead to incompetent expression of genes leading to latency in neurons [29]. ICP0 also affects modifications of histone as well as expression of several viral genes and thus may play a significant role in latency [30,31]. Another interesting concept states that dividing the HSV stages of infection into latent and lytic cycles is not completely true, as intermediate stages with variable levels of gene expression have been recorded by few studies [29,32].
Epigenetic modifications have also been shown to regulate the latency and reactivation. The naïve HSV DNA lacks histones and once it enters the host cell, histone modifications occur on the HSV genome, thus restricting important gene expressions [33]. During replication, some viral proteins have been shown to decrease heterochromatin levels and increase euchromatin to help in gene expression, whereas, in latency, the HSV genome shows constitutive heterochromatin marks [34,35].
Although silenced, this reservoir can be stimulated from the trigeminal sensory ganglia and cause subsequent recurrent clinical outbreaks as reactivation occurs via common stimuli such as stress, ultraviolet (UV) radiation, temperature fluctuations, surgical trauma or dental procedures, immunotherapy, light/laser therapy, or even hormonal changes [20]. New infectious virus particles are created during reactivation in these neurons. These particles move anterogradely to the skin or mucosa, generating classic herpes sores. The virus may potentially enter the CNS, including the brain, and produce encephalitis or meningitis in immunosuppressive patients [20,21,22,23,24]. Recent studies have demonstrated that HSV-2 reactivation is very common in the vaginal mucosa of asymptomatic patients [36,37]. Many investigations have supported the idea that the HSV 1 regularly reactivates in the trigeminal ganglia after events such as immunosuppression, peripheral infection, and even stress. An increasing body of research suggests that asymptomatic infections occur often, followed by latency in neurons, and such recurring “mild” HSV-1 infections in the brain are associated with persistent neuroinflammation [38,39]. Microglia play an important role in the immune response to neuronal infection in the brain. Microglial cells endure an abortive infection after HSV-1 infection, proceeded by a burst of pro-inflammatory cytokine and chemokine release, which activates other glial cells within the CNS and recruits immune cells including NK cells, DC cells, and T lymphocytes to the brain [38,39,40]. HSV-1-infected microglial cells have also been demonstrated to produce reactive oxygen species which are directly responsible for damage to neurons [38,39,40]. Furthermore, it has been demonstrated that microglia continued in an active state in the brain of HSV1-infected mice until thirty days after infection, despite the absence of active replication [38,39,40]. However, whether the microglia have any role in the reactivation of herpes labialis is not yet known.

3.4. Immune Response to HSV-1

The intrinsic and innate immune responses are quite effective at controlling HSV reactivation after latency and primary infection [41,42,43,44]. Additionally, they provide a powerful immune system response against HSV. Reactivation nonetheless happens and is extremely common in some people which is most likely as a result of a combination of viral evasion mechanisms and insufficient immunological control. IFNs, one of the primary cytokine families that inhibit HSV, are expressed as a result of the majority of the signaling cascades that toll-like receptors (TLRs) initiate upon recognizing HSV [41,42,43,44,45]. Mammals have 13 TLRs that have been identified, and the main ones that sense HSV are TLR2, 3, and 9 [41,42,43,44,45]. When HSV binds or fuses, TLR2 detects viral glycoproteins, TLR3 senses dsRNA produced as a consequence of HSV replication, and TLR9 detects HSV DNA [41,42,43,44,45]. Other cytoplasmic and nuclear sensors also find intermediates of HSV DNA and RNA. To control the virus, it is essential to detect HSV nucleic acids. Severe HSE is more common in those with defects in DNA sensors, the RNA sensor TLR3, or downstream signaling pathways [41,42,43,44,45].
During the early stages of infection, adaptive immune responses are also crucial [45,46,47,48,49,50,51]. Since HSV-specific T cells are seen in both active and healed lesions in patients as well as in infected sensory human ganglia, the role of T cells is very important. Immunocompetent people have a low level of HSV-specific T cells in their blood after the acute infection has cleared up [46,47,48,49,50]. When exposed to viral antigens, blood CD8 T lymphocytes specific for HSV produce plenty of cytolytic molecules and release IFNs [46,47,48,49,50]. Around 22 HSV-1 proteins, including enzymes and structural proteins, are recognized by human CD4 and CD8 T cells, respectively [47]. HSV-specific CD4 T cells express cytokines that are similar to T helper type 1 (Th1) and Th0 and are capable of cytolysis [48,49,50]. The adaptive immune response also controls latent HSV and the depletion of CD 8 T results in higher reactivation [45].

4. Clinical Manifestations

Clinical manifestations appear as cold sores and oral lesions are distinctly characterized [52]. The initial stage of RHL generally commences with prodromal symptoms such as stinging pain, tingling or burning sensations, chilling tenderness of the skin, and itchiness at reactivation sites [53]. This occurs up to 6 h in 46–60% of patients caused by abrupt viral duplication at sensory neuron terminals of the epidermis or mucosal layers [17]. Hence, antiviral treatment is favorably given at this early stage before lesions develop to a greater extent, due to the narrow therapeutic window of most antiviral drugs [53,54,55,56]. Vesicle ruptures form scalloped patterned ulcers with surrounding erythema [55,56]. Around 25% of facial recurrences stay at the papular stage with common prodromal symptoms [17]. The disease usually resolves within 10 to 14 days and occurs naturally [17]. If untreated, further complications and morbidity ensue, such as the development of ocular blemishes, encephalitis, and subsequent severity may lead to mortality [53,53].

5. Triggers and Risk Factors

One of the foremost common triggers for RHL is fever, and this led RHL to be named herpes febrilis due to its characteristics. RHL occurs three times more frequently in patients with febrile conditions compared to those without so. Feverish patients with herpes labialis developed blisters with triple effect [16,57]. In addition, the frequency of the recurrence could also coincide with the expression of numerous candidate genes, namely human chromosome-21 (C21) or the Cold Sore Susceptibility Gene-1 (CSSG-1) [58]. Previous studies revealed a total of 6 candidate genes influencing herpes susceptibility and highlighted C21 or CSSG-1 to actively link to symptoms such as fever and the development of blisters via an unknown mechanism [56,59].
Sunlight or UV light in particular also presents a major and common trigger factor for RHL. Prone individuals such as swimmers, fishermen, farmers or skiers have prolonged exposure to sunlight and typically present with lesions and vesicles at the vermillion border of lips after 3 to 5 days [16]. Spruance et al. demonstrated that intentional exposure to controlled dosages of UV radiation resulted in the development of lower lip lesions in 60% of HSV-1 seropositive [60]. Moreover, administration of prophylactic acyclovir into HSV-1 seropositive skiers eventually prevented reactivation of latent HSV-1 compared to those on placebo [61]. Ultraviolet-B-susceptible individuals have also been more prone to experience recurrent infections of herpes labialis [62].
The lack of healthy zinc levels in HSV-1 individuals was also found to cause prolonged periods of lesion episodes, as this particular element has consequential effects in aiding recovery and healing of viral lesions [63]. As an essential mineral component in driving the natural processes of cells, tissues, and organs, zinc plays a major role in wound healing by coordinating the regenerative processes of the human body such as membrane reparation, inflammation reduction, immunity-boosting, oxidative stress decrement, cell epithelialization, and the formation of fibroids or scars [64]. The defense system in boosting immunity involves the T-cell lymphocyte response, providing a stimulated but controlled cellular immunity to viruses, fungal, or pathogenic infections, and certain autoimmune or immunocompromised diseases. The presence of zinc drastically improves the number of effector and helper T-cells, subsequently forming antibody precursors and contributing to the suppressive functions of responsible cells [65,66]. Kamakura et al. observed that the upregulation of the zinc finger transcription cell protein called insulinoma-associated-1 factor showed an increase in immunity-driven phagocytic activities to halt viral duplication and strengthen the local immunity system [67]. As such, zinc supplementation is proven to show benefits to individuals suffering from herpes and impaired T-cell functioning due to zinc deficiency [65]. Topical treatments of zinc oxide creams combined with glycine can produce considerable facial and circumoral appearances of herpes infections with or without inversed side effects [64]. Additional crucial element is vitamin D which exerts similar immunomodulatory properties to zinc and low serum levels have been shown to be coincidental with the recurrence of cold sores; additional research is needed to discover the actual mechanism [68].
Another major contributor to triggering virus reactivation is the elevated levels of psychosocial stress [69,70]. A meta-analysis covering multiple psychosocial and emotional assessments reported reliable positive correlations between chronic stress and recurrence of HSV-1 infections with clinical symptoms [71]. It is believed that immune system suppression, which enables viruses to evade immune monitoring, is the mechanism through which these stress factors affect the severity and recurrences of HSV disease [71,72,73,74]. However, the many HSV-infected neuronal types, including sensory and autonomic neurons, express the receptors for the two main stress hormones, adrenaline, and cortisol, in a selective manner [73,74]. It is possible that autonomic neurons play a unique role in the stress-induced modulation of productive HSV infection because the stress hormones epinephrine and corticosterone have different effects on HSV DNA replication and the release of infectious virus progeny [73,74]. However, the exact mechanism is not yet known. CD8+ T cells have been shown to play an integral role in the suppression of modulated immune functions caused by psychological stress and in preventing latent virus reactivation [72].
Immunocompromised hosts have a longer duration of episodes and are prone to contagious spread and comorbidities at higher rates, especially those undergoing bone marrow or organ transplants, chemotherapy, or dialysis. Although herpetic keratitis or encephalitis occurs rarely, these complications result from severe or untreated HSV-1 infections [56]. The white blood cell (WBC) differential variable is particularly used to predict the severity of immunosuppression although a direct association with herpes virus reactivation is yet to be confirmed. In one study of Caucasian ethnicity, the decreased lymphocyte count was found to have an association with herpes labialis, suggesting that modifications in WBC differential values account for risks of HSV-1 reactivation [75]. In addition, HSV-1 infection is considered “atypical” in immunocompromised individuals as they exhibit a larger extent of damage and aggression compared to immunocompetent patients, slower recovery (median time to healing up to 28 days), and a higher degree of pain. Moreover, co-infections with CMV and other viruses are common in immunocompromised patients [76]. Very little is known about the existence of viruses responsible for infections in the oral cavity and their interactions with each other.
In terms of the significance of HSV-1 trigger factors instrumental to herpes symptoms, the patient’s age, sex, the environment of residency, or inappropriate lifestyle affecting health such as smoking, consuming alcohol, or taking illicit drugs were less likely to contribute when compared to perceived elevated levels of stress. There were unclear occurrences in multivariate study models and cross-sectional studies, with no conclusive evidence of the apparent association between these putative risk factors and RHL [69]. The commonly reported trigger factors are listed as Table 1.

6. Management Strategies

6.1. Conventional Therapies

One of the earliest “home remedy” and long-standing treatments effectively used by RHL patients is cryotherapy, where self-treatment using ice cubes provides instant relief from prodromal symptoms, including burning/tingling sensations, itchiness, and pain [77]. Lip moisturizers and balms were introduced to counter humidity, and rough environments with fluctuating temperature changes which affect sufficient lip hydration [78] as these trigger factors are very likely experienced in most populations exposed to extreme weather and long exposure under the sun. Sunscreens, especially for lips, were also popularized not only for moisturizing but to block UV radiation from sunlight in seasonal weather [78,79,80,81,82].
Natural remedies, including the use of aloe vera leaf extract to produce a topical therapeutic effect with anti-inflammatory, anti-bacterial, and antiviral properties, have been tried as well. Rezazadeh et al. showed that aloe vera in gel form inhibited significant growth of HSV-1 at infection sites without any toxic side effects, indicating a convenient and safe choice to treat itchy and painful HSV-1 blisters and lesions [81]. Furthermore, the use of silica gel as an alternative gained approval as it was effective in the treatment of RHL with fewer side effects and had faster onset of action [82].
Another early drug used was levamisole due to its parasitic and antiviral properties. Although few studies showed mild effectiveness, it was proven to be lacking substantial evidence in a double-blind controlled trial by Russell et al., which did not find any significant differences in reducing frequency or severity between groups taking levamisole and the control group [83]. When it comes to targeting the pain barrier and feverish symptoms, anti-inflammatory drugs, analgesics, and antipyretics, such as paracetamol, ibuprofen, as well as mefenamic acid, were considered as there was an apparent connection between anti-inflammatory mechanisms and viral inhibitory effects of the drugs. Experimental studies found inhibition of viral duplications, in addition to antiphlogistic and anti-rheumatic properties of these drugs, but to a lesser degree [84]. One potent non-steroidal anti-inflammatory drug (NSAID) studied was piroxicam which exerts antipyretic effects as well as exerts a directly active antiviral response on the HSV-1 infection due to its lipophilicity, thus making this agent suitable as a topical form of treatment for herpes virus infections [85]. Even local anesthetics, such as tetracaine and lignocaine, have been prescribed to relieve the pain and itchiness arising from cold sores and fever blisters [86].
Internal supplementations were also believed to have some prophylactic effects in RHL individuals. Some of the earliest studies included L-lysine monohydrochloride in 1000 mg daily doses. A double-blinded, placebo-controlled crossover study by Milman et al. that evaluated 65 patients with RHL showed no apparent effect on the healing rate in the lysine group of patients but showed significant improvements in being recurrence-free [87]. However, several other studies provided data suggesting lysine monohydrochloride salts were largely ineffective in treating acute cutaneous HSV-1 infections [88]. On the other hand, Thein and Hurt’s study evaluating the long-term success and prophylactic efficacy of lysine showed a significant reduction in recurrence rates at high serum lysine levels (more than 165 nmol/mL) [89]. Additionally, high doses of lysine monohydrochloride, 1000 to 3000 mg/day, were found to reduce the recurrence rate in a study by Szapary and Cirigliano, which also specified the cost-effectiveness and safety when compared to oral antiviral drugs [90]. A combination of lysine–arginine supplementations also showed good control in managing RHL even when the lysine content is increased without affecting the balance of the original compound, suggesting the safety of supplementation even at higher doses [91].
Zinc ions have been reported to disrupt viral multiplication, with oral concentrations of zinc gluconate providing approximately ten-fold zinc serum levels in immunocompromised individuals [65,92]. Contrastingly, a recent meta-analysis by Cunningham et al. suggested inconclusive and limited evidence of zinc oxide creams in the treatment of RHL but found it was still reliable in lowering pain threshold and symptom periods [93]. This is specifically the case in immunocompromised hosts and critically ill patients as they are unable to consume oral concentrations of zinc [65].
Although NSAIDs, analgesics, zinc-based creams, and aloe vera gels were recognized as over the counter (OTC) medications, topical Docosanol (Abreva) is another OTC approved by the Food and Drug Administration in treating cold sores topically. It is a long-chain 22-carbon, saturated primary alcohol which inhibits broad spectrum lipid-enfolded viruses such as HSV-1 [88]. Experimental studies initially stated that docosanol interferes with nucleus localization and viral multiplications, but further experiments highlighted that it disrupts viral coalition with host cells and treatment should begin as early as possible for maximal effect [94]. The safety profile is also well-established without risk of resistance or side effects, making it one of the choices in managing most herpesvirus infections with adequate efficacy [95]. The conventional therapies available are listed as Table 2.

6.2. Antivirals

6.2.1. Acyclovir, Valacyclovir, Penciclovir, and Famciclovir

Therapeutics involving antivirals for recurrent HSV-1 infections include systemic and topical agents called virustatics (Table 3). The first virustatic, idoxuridine, existed in the mid-1950s as a topical application for non-specific herpesvirus, while vidarabine was licensed in 1978 as the systemic compound for treating encephalitis [96,97,98]. Towards the late 1970s and early 1980s, nucleoside virustatics were introduced and have been widely recognized as the most prescribed antiviral drugs for RHL, due to their safety and efficacy benefits [99]. The first of these landmark nucleoside inhibitors is the potent and selective Acyclovir (Zovirax) which disrupts DNA polymerase of herpes virus, followed by its generic alternative, prodrugs Valacyclovir and Famciclovir, and a resembling analog Penciclovir [96]. While there were more efficacy data available for oral antivirals in treating acute infections [90], expansive contributions continue to appear for topical antivirals such as 5% acyclovir as the first-line therapy for RHL and 1% penciclovir [82]. Although over the years numerous clinical trials conducted globally resulted in a new generation of antivirals for herpes infections, acyclovir remains the “gold standard” [96].
Another cream formulation commonly used is foscarnet (trisodium phosphonoformate) which selectively disrupts HSV-1 DNA polymerase activity, showing clinical efficacy in decreasing the size and development time of recurrent lesions [100]. Although foscarnet is employed exclusively to treat infections with cytomegalovirus (CMV), acyclovir-resistant herpes simplex virus (HSV) also tends to respond to this drug.
Structural analysis of acyclovir revealed that the molecule is almost identical to the acyclic DNA-base guanosine [82]. Like all nucleoside analogs, they are favorably cleaved by infected viral cells, where they undertake initial phosphorylation at 3000 times faster than normal unaffected cells via Tyrosine Kinases (TKs), the viral encoded enzymes [101,102]. With this attribute, acyclovir is effortlessly phosphorylated by these viral TKs compared to readily available cellular TKs, with maximum concentrations of the monophosphorylated drug in the infected cells compared to the normal uninfected cells [101,102]. Then it is converted to the active triphosphate configuration by cellular kinases to be incorporated into the elongated viral chain as opposed to normal cellular polymerase, leading to viral multiplication remission and chain termination [101]. This shows that acyclovir acts not only as a chain terminator but also as a competitive inhibitor for viral TK, while the acyclovir–triphosphate complex molecule becomes an imperious suicide inhibitor for the viral DNA polymerase, ceasing the activity of the exonuclease. As this cycle is repeated, HSV-1 DNA polymerase continues to add more nucleotides to the growing DNA chain until the excised nucleotides reach acyclovir again. The viral DNA polymerase is thus inactivated, and viral genomes are unable to reach full replication or form mature strains of HSV-1 [10].

6.2.2. Problems of Bioavailability and Resistance Leading to Novel Approaches for Newer Antivirals

To overcome the inferior hydrophilic nature and poor oral bioavailability of acyclovir, the prodrug valacyclovir was introduced via valine esterification of acyclovir. With higher bioavailability, there was increased absorption, approximately 54% in the intestinal region, thus preventing wastage when oral doses are given [102,103]. Valacyclovir is converted back into active acyclovir and valine via metabolism in the liver and kidney by the protein-like enzyme biphenyl hydrolase where the esterified amino acid is cleaved from the molecule [103,104,105]. Another prodrug developed with the same objective of more bioavailability and higher water solubility was penciclovir with a swifter phosphorylation process and a longer half-life in triphosphate form than acyclovir [103]. However, penciclovir contains the 30-hydroxyl group on the acyclic side chain which limits the extension of the nucleoside chain, thus removing the chain termination ability as seen in acyclovir although still significantly inhibiting DNA polymerase [101]. As a result, oral penciclovir has a significantly lower bioavailability than acyclovir (1.5% vs. 5%) and a shorter half-life (2–2.5 h vs. 4–5 h) and due to this, penciclovir is preferably formulated as topical creams [104]. To rectify the poor oral bioavailability and short half-life of penciclovir, the newer prodrug famciclovir, the diacetyl 6-deoxy ester of penciclovir with improved oral uptake of around 73%, was introduced [101,104]. The new orally administered prodrug can be converted completely to penciclovir via oxidation at the 6-position by the hepatic enzyme cytosolic aldehyde oxidase following first-pass metabolism and initial deacetylation by intestinal esterases, leading to the same antiviral and pharmacokinetic properties as penciclovir [106]. A single daily dose of famciclovir is rapidly absorbed in the gastrointestinal tract, yielding approximately 77% bioavailability even without the presence of food [107,108].
The resistance toward antiviral nucleoside analogs arises from two mechanisms: mutated TK genes in viral cells and mutations in DNA polymerase, which inhibits DNA synthesis and interferes with antiviral activity. This tends to occur more in immunosuppressed individuals as compared to normal immunocompetent patients with frequent RHL attacks as reported by Mubareka et al. in their comprehensive review of 11 clinical trials [106]. Further studies on acyclovir showed that viral resistance resulted mainly from mutant TKs [103]. As viral TKs are essential for the conversion of acyclovir to the monophosphate form, their absence subsequently blocks host enzyme phosphorylation resulting from single and multiple mutations of these TKs [104]. This viral TK deficiency demonstrates an identical cross-resistance pattern among all the nucleoside analogs from acyclovir, valacyclovir, penciclovir, and famciclovir among immunocompetent individuals, but the higher prevalence among immunocompromised patients with severity depending on underlying diseases such as hematologic transplants, stem cell replacements or solid organ surgeries [107,108]. Thus, immunocompromised patients with a higher magnitude of suppression to their immune system encounter more frequent recurrences, longer periods of clinical symptoms, lesion sizes, and in more severe cases, become life-threatening [109].
Even to this day, there have been minimal clinical trials that directly compare oral or topical therapeutics for RHL outbreaks. Antivirals moderately reduce the time to healing and pain subsiding, with valacyclovir showing efficacy in these two preferred outcomes, famciclovir only at a specific dosage, while acyclovir was effective in decreasing only pain duration [110]. Essentially, treatment with antivirals should start as early as possible at the prodromal phase of herpes labialis [109].
A novel approach included the introduction of newer drugs, amenamevir (ASP2151) and pritelivir (BAY 57–1293), belonging to the helicase–primase inhibitor class, with the heterotrimer complex comprising accessory proteins UL5 helicase, UL52 primase, and UL8 subunit. As the absence of a eukaryotic homolog is mitigated by this helicase–primase complex, viral replication is blocked [109]. Early phase II clinical trials only involved animal models and HSV-2 infections, but further trials are required for understanding the clinical potential of these two promising drugs on HSV1 infections [111].
Another novel approach focused on new delivery systems which can provide more efficient drug administration to enhance skin penetration with unique vesicular carriers called ethosomes. They mainly contain phospholipids along with high content of ethanol and water. Ethanol mainly allows the fluid formation of lipids in ethosomal molecules and allows compact lipid arrangement in the skin barrier. Hence, to enhance skin penetration, ethosomes use these double effects of ethanol on lipid bilayers of the stratum corneum and the vesicle, permitting soft vesicles to penetrate the modified stratum corneum structure and gradually release the active drug in extended skin layers [112]. Studies by Horwitz et al. and Nainwal that focused on ethosomal delivery of acyclovir, showed significant improvement to all relevant clinical measurements compared to the pioneer Zovirax cream, thus formulated as the new Supra-Vir acyclovir cream using this ethosome technology [113,114].
Similarly, nanomedicine involving nanoparticles was proven effective for medicinal applications using particulate components with approximately 100 nm in size or lower. It was found to have lower toxicity and adverse effects while maintaining effectiveness in targeted treatments, where efficacy was enhanced with nanoparticle surface adjustments and preservation of structural properties among other biological, antiviral, antimicrobial, and pathogenic characteristics [115]. Hence, this approach led to an experimental polyethylene glycol-coated zinc oxide nanoparticle with acyclovir to potentiate systemic and local therapies to fight against HSV-1 infections. Initial findings showed some antiviral potential for HSV-1 infections in a cell culture model with low cytotoxicity but more clinical trials are required [116].

6.3. Emergence of Bioactive Natural Products

Although non-pharmaceutical approaches beginning with nutrient supplementation such as zinc, vitamin C, vitamin D, lysine, and low arginine diets have some potential benefits in the management of HSV-1 infections, results were mostly inconsistent and unreliable. Although the emergence of other natural remedies showed encouraging results in some clinical trials, most did not provide complete safety profiles or were assumed to be safe due to their natural identity. A comprehensive review by Münstedt provided essential evidence of numerous natural medicaments, such as L-lysine, lemon balm (Melissa officinalis), aloe vera extract, astragalus, bee propolis, elderberry, echinacea, cat’s claw, sage-rhubarb, sandalwood, tea tree oil, and witch hazel, in preventing illnesses and alleviating diseases [117]. Bee propolis as lip balms was proposed as an alternative to topical antiviral applications as one study confirmed its superiority and safety when used in cases where standard antivirals were not profoundly tolerated [118]. An open-label RCT using medical-grade kanuka honey with superior antiviral effects towards HSV labial occurrence highlighted that botanical compounds can be an alternative to acyclovir with a better safety profile [103,119].
Some of the established zinc-based creams synergistically enhanced in combination with herbal-based ingredients, including echinacea and goldenseal, resolved RHL outbreaks within three days in 40% of affected individuals [120]. With stronger antiviral activities and a simple mechanism of action potentiating prophylactic benefits on viral growth stages, these natural products were favorable because of the lower chance of developing resistance, lower toxicity, and price compared to antivirals [120]. Additional natural compounds, which have been tested at basic trial levels, include neem-based creams, beeswax, jojoba oil, shea butter, sesame oil, peppermint oil, and also coconut oil [119].

6.4. The Modern, Non-medicinal Future: Laser Therapy Immunotherapies and Probiotics

As the world moves forward in technological advancements and global opinion favoring a non-medicinal future, laser treatments, electrical stimulation, immunotherapies, and vaccinations carry a huge expectation in solving RHL. Based on the literature, there were no confirmed laser therapy modalities that can fully decimate virus replication and prevent recurrence with 100% success, but clinical trials did show a reduction in the pain and frequency of recurrences in almost all patients suffering from primary and secondary infections of herpes labialis [121]. A recent review concluded that although low-level laser therapy is superior to most antivirals in RHL therapy, the inconsistent study designs and unparallel clinical parameters hinder robust conclusions and hence, more standardization is warranted [122]. Another novel method is the transdermal electrical stimulator, especially in patients intolerant to antiviral agents [88]. Additionally, the topical immuno-sensitizer squaric acid dibutyl ester (SADBE) may help in the prevention of RHL outbreaks and reduce frequency rates by delaying hypersensitivity reactions [123]. A promising risk–benefit analysis of topical 2% SADBE by Chang et al. suggested that this approach may decrease the recurrence rate and severity of outbreaks, although it may take up to 3 months for the treatment effect [124]. Finally dendritic cell vaccines could be a ray of hope when other treatments seem to give no efficacious impact [125]. With the incorporation of HSV-1 recombinant glycoprotein D vaccines in activating HSV-1-specific immune responses, mononuclear cells will receive a decreased mitogenic reaction which allows normalization of the immune system, providing a balance in viral replication breakdown [126].
Altering the commensal microbiome indigenous to a specific body site has been tried and tested for the management of several diseases. It has been reported that the commensal microbiome primes the immune system and the microbiota can promote antiviral immunity [127,128]. A recent study suggested that gut microbiome can play a role in preventing HSV encephalitis via the gut–brain axis [129] and hence, probiotics are being explored for the management of encephalitis. The bacteriocin subtilosin generated by Bacillus species affects the late infectious phases of both HSV-1 and HSV-2 invitro [130]. The subtilosin, another enterocin from Enterococcus faecium, also suppresses the early infectious cycle of HSV-1 in vitro [127,130]. Whether the manipulation of the gut and oral microbiome prevents herpes labialis infection is yet to be explored.

7. Impacts on Quality of Life

7.1. Physical and Psychological Implications

Unintentionally, patients and treatment providers often resort to quick treatment for RHL without a complete assessment of viral infection severity, dissemination of risk factors, or even internal immunocompetency checks [2]. Hence, problems with antiviral resistance, absorption, distribution, metabolism, and elimination add to dispensable aspects in the quality of life of the patients which can be impactful depending on the severity and frequency of the infection. While most symptoms are often mild enough, the prolonged duration of the illness may become uncomfortable and traumatizing up to the brink of emotional impairment. This psychological impact may especially affect the young and self-conscious where facial appearances are a priority [56].
In combining both psychosocial methods for the prevention of recurrence and reducing psychological stress, outcomes may be favorable to both the patients and also healthcare professionals [110]. It is interesting to note considerable data connecting emotional distress or negative emotions to HSV epidemiology, where RHL can frequently occur due to triggering factors, including disgust and misery [131]. Constant sleep deprivation may cause lasting effects on the body, especially on skin functionality and integrity. With prolonged sleep deficit, stress can be elevated without incitement and affect the modulation of the hypothalamic–pituitary–adrenal axis in the body, thus increasing excessive glucocorticoid production to put the body in an immunocompromised state and triggering psychological effects to reactivate HSV-1 infections [132].

7.2. Lifestyle Modifications (Habits, Hygienic Considerations, Dietary)

Practical methods in lifestyle modifications can be convenient alternatives compared to substantially expensive treatments or risky therapies and these include prevention of prolonged exposure to sunlight, use of sunscreen, exercise, and relaxation methods to reduce emotional stress, healthy hygiene habits, and healthy and sufficient sleep [133]. All these adjustable lifestyle factors were further explored in a review of holistic dermatology techniques by Hu et al. who highlighted the interrelationship between healthy habits and dermatological health in mitigating the progressive nature of most viral skin infections [132]. Dietary adjustments can also include L-lysine [134], zinc, and vitamin C supplementation as they have been linked to the healing process. A combination of alcohol drinking and nutritional decline possess a serious influence on a weakened immune defense and thereby RHL recurrence, and hence, could be tired to be controlled by lifestyle modifications [132].

8. Conclusions

Seroprevalence data show that oral HSV-1 infection regularly transmits among vulnerable humans as the virus transition constantly happens from latent to recurrent stages. A variety of management regimens ranging from natural sources to potent oral/topical medications establish sufficient relief, although still few available agents offer only temporary relief and are unable to overcome bioavailability/resistance problems. In addition, the combination of psychosocial techniques to relieve stress, and adjusting lifestyle modifications reasonably may also have positive implications in quality-of-life aspects. More substantial research and consolidated evidence are needed to further associate viral infection/reactivation with exact triggers and development of prediction models for reactivation/recurrence.

Author Contributions

Conceptualization, D.G. and K.H.K.; methodology, D.G.; validation, M.K.M. and D.G.; formal analysis, K.H.K.; investigation, D.G. and K.H.K.; resources, D.G.; data curation, D.G. and S.P.; writing—original draft preparation, K.H.K.; writing—review and editing, D.G. and S.P.; visualization and supervision, D.G. and M.K.M.; project administration, D.G. and M.K.M.; funding acquisition, D.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Herpes & Shingles: Symptoms & Treatments—Life Extension. Available online: https://www.lifeextension.com/protocols/infections/herpes-and-shingles (accessed on 19 September 2021).
  2. Birek, C.; Ficarra, G. The diagnosis and management of oral herpes simplex infection. Curr. Infect. Dis. Rep. 2006, 8, 181–188. [Google Scholar] [CrossRef] [PubMed]
  3. Boes, H.; Goulioumis, V.; Wechsler, A.; Zimmer, S.; Bizhang, M. Clinical Study on the Effectiveness of Three Products in the Treatment of Herpes Simplex Labialis. Sci. Rep. 2020, 10, 6465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Petti, S.; Lodi, G. The controversial natural history of oral herpes simplex virus type 1 infection. Oral Dis. 2019, 25, 1850–1865. [Google Scholar] [CrossRef] [PubMed]
  5. Billions Worldwide Living with Herpes. Available online: https://www.who.int/news/item/01-05-2020-billions-worldwide-living-with-herpes (accessed on 9 May 2022).
  6. Cernik, C.; Gallina, K.; Brodell, R.T. The treatment of herpes simplex infections: An evidence-based review. Arch. Intern. Med. 2008, 168, 1137–1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Khadr, L.; Harfouche, M.; Omori, R.; Schwarzer, G.; Chemaitelly, H.; Abu-Raddad, L.J. The Epidemiology of Herpes Simplex Virus Type 1 in Asia: Systematic Review, Meta-analyses, and Meta-regressions. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2019, 68, 757–772. [Google Scholar] [CrossRef] [PubMed]
  8. James, C.; Harfouche, M.; Welton, N.J.; Turner, K.M.; Abu-Raddad, L.J.; Gottlieb, S.L.; Looker, K.J. Herpes simplex virus: Global infection prevalence and incidence estimates, 2016. Bull. World Health Organ. 2020, 98, 315–329. [Google Scholar] [CrossRef]
  9. Rani, S.A.; Suragimath, G.; Rajmane, V.; Rajmane, Y. Prevalence of recurrent herpes labialis in Western Maharashtr. J. Oral Maxillofac. Pathol. 2021, 25, 51. [Google Scholar]
  10. Löwhagen, G.B.; Bonde, E.; Eriksson, B.; Nordin, P.; Tunbäck, P.; Krantz, I. Self-reported Herpes labialis in a Swedish Population. Scand. J. Infect. Dis. 2002, 34, 664–667. [Google Scholar] [CrossRef]
  11. Gibson, J.J.; Hornung, C.A.; Alexander, G.R.; Lee, F.K.; Potts, W.A.; Nahmias, A.J. A Cross-Sectional Study of Herpes Simplex Virus Types 1 and 2 in College Students: Occurrence and Determinants of Infection. J. Infect. Dis. 1990, 162, 306–312. [Google Scholar] [CrossRef]
  12. Barton, E.S.; White, D.W.; Cathelyn, J.S.; Brett-McClellan, K.A.; Engle, M.; Diamond, M.S.; Miller, V.L.; Virgin, H.W. Herpesvirus latency confers symbiotic protection from bacterial infection. Nature 2007, 447, 326–329. [Google Scholar] [CrossRef]
  13. Rajčáni, J.; Bánáti, F.; Szenthe, K.; Szathmary, S. The potential of currently unavailable herpes virus vaccines. Expert Rev. Vaccines 2018, 17, 239–248. [Google Scholar] [CrossRef]
  14. Higgins, C.R.; Schofield, J.K.; Tatnall, F.M.; Leigh, I.M. Natural history, management and complications of herpes labialis. J. Med. Virol. 1993, 41 (Suppl. 1), 22–26. [Google Scholar] [CrossRef]
  15. Artzy-Randrup, Y.; Pascual, M. Composite temporal strategies in pathogen evolution: Balancing invasion and persistence. Theor. Ecol. 2014, 7, 325–334. [Google Scholar] [CrossRef]
  16. Roizman, B.; Whitley, R.J. An Inquiry into the Molecular Basis of HSV Latency and Reactivation. Annu. Rev. Microbiol. 2013, 67, 355–374. [Google Scholar] [CrossRef] [Green Version]
  17. Arduino, P.G.; Porter, S.R. Herpes Simplex Virus Type 1 infection: Overview on relevant clinico-pathological features. J. Oral Pathol. Med. Off. Publ. Int. Assoc. Oral Pathol. Am. Acad. Oral Pathol. 2008, 37, 107–121. [Google Scholar] [CrossRef]
  18. Whitley, R.; Kimberlin, D.W.; Prober, C.G. Pathogenesis and disease. In Human Herpesviruses: Biology, Therapy, and Immunoprophylaxis; Arvin, A., Campadelli-Fiume, G., Mocarski, E., Moore, P.S., Roizman, B., Whitley, R., et al., Eds.; Cambridge University Press: Cambridge, UK, 2007. Available online: http://www.ncbi.nlm.nih.gov/books/NBK47449/ (accessed on 28 July 2021).
  19. Nicoll, M.P.; Proenca, J.T.; Efstathiou, S. The molecular basis of herpes simplex virus latency. FEMS Microbiol. Rev. 2012, 36, 684–705. [Google Scholar] [CrossRef]
  20. Nicoll, M.P.; Hann, W.; Shivkumar, M.; Harman, L.E.; Connor, V.; Coleman, H.M.; Proença, J.T.; Efstathiou, S. The HSV-1 latency-associated transcript functions to repress latent phase lytic gene expression and suppress virus reactivation from latently infected neurons. PLoS Pathog. 2016, 12, e1005539. [Google Scholar] [CrossRef]
  21. Umbach, J.L.; Kramer, M.F.; Jurak, I.; Karnowski, H.W.; Coen, D.M.; Cullen, B.R. MicroRNAs expressed by herpes simplex virus 1 during latent infection regulate viral mRNAs. Nature 2008, 454, 780–783. [Google Scholar] [CrossRef] [Green Version]
  22. Pan, D.; Flores, O.; Umbach, J.L.; Pesola, J.M.; Bentley, P.; Rosato, P.C.; Leib, D.A.; Cullen, B.R.; Coen, D.M. A neuron-specific host microRNA targets herpes simplex virus-1 ICP0 expression and promotes latency. Cell Host Microbe 2014, 15, 446–456. [Google Scholar] [CrossRef] [Green Version]
  23. Vanni, E.A.; Foley, J.W.; Davison, A.J.; Sommer, M.; Liu, D.; Sung, P.; Moffat, J.; Zerboni, L.; Arvin, A.M. The latency-associated transcript locus of herpes simplex virus 1 is a virulence determinant in human skin. PLoS Pathog. 2020, 16, e1009166. [Google Scholar] [CrossRef]
  24. Lafferty, W.E.; Coombs, R.W.; Benedetti, J.; Critchlow, C.; Corey, L. Recurrences after Oral and Genital Herpes Simplex Virus Infection. N. Engl. J. Med. 1987, 316, 1444–1449. [Google Scholar] [CrossRef] [PubMed]
  25. Cabrera, J.R.; Charron, A.J.; Leib, D.A. Neuronal Subtype Determines Herpes Simplex Virus 1 Latency-Associated-Transcript Promoter Activity during Latency. J. Virol. 2018, 92, e00430-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Cohen, J.I. Herpesvirus latency. J. Clin. Investig. 2020, 130, 3361–3369. [Google Scholar] [CrossRef] [PubMed]
  27. Perng, G.C.; Jones, C.; Ciacci-Zanella, J.; Stone, M.; Henderson, G.; Yukht, A.; Slanina, S.M.; Hofman, F.M.; Ghiasi, H.; Nesburn, A.B.; et al. Virus-induced neuronal apoptosis blocked by the herpes simplex virus latency-associated transcript. Science 2000, 287, 1500–1503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Hafezi, W.; Lorentzen, E.U.; Eing, B.R.; Müller, M.; King, N.J.; Klupp, B.; Mettenleiter, T.C.; Kühn, J.E. Entry of herpes simplex virus type 1 (HSV-1) into the distal axons of trigeminal neurons favors the onset of nonproductive, silent infection. PLoS Pathog. 2012, 8, e1002679. [Google Scholar] [CrossRef] [Green Version]
  29. Zhu, S.; Viejo-Borbolla, A. Pathogenesis and virulence of herpes simplex virus. Virulence 2021, 12, 2670–2702. [Google Scholar] [CrossRef]
  30. Boutell, C.; Everett, R.D. Regulation of alpha herpes virus infections by the ICP0 family of proteins. J. Gen. Virol. 2013, 94 Pt 3, 465–481. [Google Scholar]
  31. Lanfranca, M.P.; Mostafa, H.H.; Davido, D.J. HSV-1 ICP0: An E3 ubiquitin ligase that counteracts host intrinsic and innate immunity. Cells 2014, 3, 438–454. [Google Scholar] [CrossRef] [Green Version]
  32. Singh, N.; Tscharke, D.C. Herpes simplex virus latency is noisier the closer we look. J. Virol. 2020, 94, e01701-19. [Google Scholar] [CrossRef] [Green Version]
  33. Knipe, D.M.; Cliffe, A. Chromatin control of herpes simplex virus lytic and latent infection. Nat. Rev. Microbiol. 2008, 6, 211–222. [Google Scholar] [CrossRef]
  34. Cliffe, A.R.; Garber, D.A.; Knipe, D.M. Transcription of the herpes simplex virus latency-associated transcript pro-motes the formation of facultative heterochromatin on lytic promoters. J. Virol. 2009, 83, 8182–8190. [Google Scholar] [CrossRef] [Green Version]
  35. Wang, Q.Y.; Zhou, C.; Johnson, K.E.; Colgrove, R.C.; Coen, D.M.; Knipe, D.M. Herpesviral latency-associated transcript gene promotes assembly of heterochromatin on viral lytic-gene promoters in latent infection. Proc. Natl. Acad. Sci. USA 2005, 102, 16055–16059. [Google Scholar] [CrossRef]
  36. Miller, C.S.; Danaher, R.J. Asymptomatic shedding of herpes simplex virus (HSV) in the oral cavity. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 2008, 105, 43–50. [Google Scholar] [CrossRef]
  37. Ramchandani, M.S.; Jing, L.; Russell, R.M.; Tran, T.; Laing, K.J.; Magaret, A.S.; Selke, S.; Cheng, A.; Huang, M.L.; Xie, H.; et al. Viral genetics modulate orolabial herpes simplex virus type 1 shedding in humans. J. Infect. Dis. 2019, 219, 1058–1066. [Google Scholar] [CrossRef]
  38. Patrycy, M.; Chodkowski, M.; Krzyzowska, M. Role of Microglia in Herpesvirus-Related Neuroinflammation and Neurodegeneration. Pathogens 2022, 11, 809. [Google Scholar] [CrossRef]
  39. Chen, Z.; Zhong, D.; Li, G. The role of microglia in viral encephalitis: A review. J. Neuroinflamm. 2019, 16, 76. [Google Scholar] [CrossRef]
  40. Marques, C.P.; Cheeran, M.C.; Palmquist, J.M.; Hu, S.; Urban, S.L.; Lokensgard, J.R. Prolonged microglial cell activation and lymphocyte infiltration following experimental herpes encephalitis. J. Immunol. 2008, 181, 6417–6426. [Google Scholar] [CrossRef] [Green Version]
  41. Schoggins, J.W. Interferon-stimulated genes: What do they all do? Annu. Rev. Virol. 2019, 6, 567–584. [Google Scholar] [CrossRef]
  42. Vandevenne, P.; Sadzot-Delvaux, C.; Piette, J. Innate immune response and viral interference strategies developed by human herpesviruses. Biochem. Pharmacol. 2010, 80, 1955–1972. [Google Scholar] [CrossRef] [Green Version]
  43. Zhang, S.Y.; Casanova, J.L. Inborn errors underlying herpes simplex encephalitis: From TLR3 to IRF3. J. Exp. Med. 2015, 212, 1342–1343. [Google Scholar] [CrossRef] [Green Version]
  44. Pires de Mello, C.P.; Bloom, D.C.; Paixão, I.C. Herpes simplex virus type-1: Replication, latency, reactivation and its antiviral targets. Antivir. Ther. 2016, 21, 277–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Moss, N.J.; Magaret, A.; Laing, K.J.; Kask, A.S.; Wang, M.; Mark, K.E.; Schiffer, J.T.; Wald, A.; Koelle, D.M. Peripheral blood CD4 T-cell and plasmacytoid dendritic cell (pDC) reactivity to herpes simplex virus 2 and pDC number do not correlate with the clinical or virologic severity of recurrent genital herpes. J. Virol. 2012, 86, 9952–9963. [Google Scholar] [CrossRef] [PubMed]
  46. Zenner, H.L.; Mauricio, R.; Banting, G.; Crump, C.M. Herpes simplex virus 1 counteracts tetherin restriction via its virion host shutoff activity. J. Virol. 2013, 87, 13115–13123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zhu, J.; Koelle, D.M.; Cao, J.; Vazquez, J.; Huang, M.L.; Hladik, F.; Wald, A.; Corey, L. Virus-specific CD8+ T cells accumulate near sensory nerve endings in genital skin during subclinical HSV-2 reactivation. J. Exp. Med. 2007, 204, 595–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Zhu, J.; Peng, T.; Johnston, C.; Phasouk, K.; Kask, A.S.; Klock, A.; Jin, L.; Diem, K.; Koelle, D.M.; Wald, A.; et al. Immune surveillance by CD8alphaalpha+ skin-resident T cells in human herpes virus infection. Nature 2013, 497, 494–497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Posavad, C.M.; Zhao, L.; Dong, L.; Jin, L.; Stevens, C.E.; Magaret, A.S.; Johnston, C.; Wald, A.; Zhu, J.; Corey, L.; et al. Enrichment of herpes simplex virus type 2 (HSV-2) reactive mucosal T cells in the human female genital tract. Mucosal Immunol. 2017, 10, 1259–1269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Jing, L.; Haas, J.; Chong, T.M.; Bruckner, J.J.; Dann, G.C.; Dong, L.; Marshak, J.O.; McClurkan, C.L.; Yamamoto, T.N.; Bailer, S.M.; et al. Cross-presentation and genome-wide screening reveal candidate T cells antigens for a herpes simplex virus type 1 vaccine. J. Clin. Investig. 2012, 122, 654–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Paludan, S.R.; Bowie, A.G.; Horan, K.A.; Fitzgerald, K.A. Recognition of herpesviruses by the innate immune system. Nat. Rev. Immunol. 2011, 11, 143–154. [Google Scholar] [CrossRef]
  52. Zhang, H.; Li, Z.; Li, C.; Chen, R.; Liu, T.; Jiang, Y. Antiviral Effect of Polyphenolic Substances in Geranium wilfordii Maxim against HSV-2 Infection Using in vitro and in silico Approaches. Evid. Based Complement. Altern. Med. 2022, 18, 7953728. [Google Scholar] [CrossRef]
  53. Simmons, A. Clinical Manifestations and Treatment Considerations of Herpes Simplex Virus Infection. J. Infect. Dis. 2002, 186 (Suppl. S1), S71–S77. [Google Scholar] [CrossRef] [Green Version]
  54. Ramchandani, M.; Kong, M.; Tronstein, E.; Selke, S.; Mikhaylova, A.; Magaret, A.; Huang, M.L.; Johnston, C.; Corey, L.; Wald, A. Herpes Simplex Virus Type 1 Shedding in Tears, and Nasal and Oral Mucosa of Healthy Adults. Sex. Transm. Dis. 2016, 43, 756–760. [Google Scholar] [CrossRef] [Green Version]
  55. Kumar, S.P.; Chandy, M.L.; Shanavas, M.; Khan, S.; Suresh, K.V. Pathogenesis and life cycle of herpes simplex virus infection-stages of primary, latency and recurrence. J. Oral Maxillofac. Surg. Med. Pathol. 2016, 28, 350–353. [Google Scholar] [CrossRef]
  56. Hobbs, M.R.; Jones, B.B.; Otterud, B.E.; Leppert, M.; Kriesel, J.D. Identification of a Herpes Simplex Labialis Susceptibility Region on Human Chromosome 21. J. Infect. Dis. 2008, 197, 340–346. [Google Scholar] [CrossRef] [Green Version]
  57. Fatahzadeh, M.; Schwartz, R.A. Human herpes simplex virus infections: Epidemiology, pathogenesis, symptomatology, diagnosis, and management. J. Am. Acad. Dermatol. 2007, 57, 737–763. [Google Scholar] [CrossRef]
  58. Kriesel, J.D.; Bhatia, A.; Thomas, A. Cold sore susceptibility gene-1 genotypes affect the expression of herpes labialis in unrelated human subjects. Hum. Genome Var. 2014, 1, 14024. [Google Scholar] [CrossRef] [Green Version]
  59. Grinde, B. Herpesviruses: Latency and reactivation—Viral strategies and host response. J. Oral Microbiol. 2013, 5, 22766. [Google Scholar] [CrossRef] [Green Version]
  60. Spruance, S.L.; Freeman, D.J.; Stewart, J.C.; McKeough, M.B.; Wenerstrom, L.G.; Krueger, G.G.; Piepkorn, M.W.; Stroop, W.G.; Rowe, N.H. The natural history of ultraviolet radiation-induced herpes simplex labialis and response to therapy with peroral and topical formulations of acyclovir. J. Infect. Dis. 1991, 163, 728–734. [Google Scholar] [CrossRef] [Green Version]
  61. Spruance, S.L.; Hamill, M.L.; Hoge, W.S.; Davis, L.G.; Mills, J. Acyclovir Prevents Reactivation of Herpes Simplex Labialis in Skiers. JAMA 1988, 260, 1597–1599. [Google Scholar] [CrossRef]
  62. Taylor, J.R.; Schmieder, G.J.; Shimizu, T.; Tie, C.; Streilein, J.W. Interrelationship between ultraviolet light and recurrent herpes simplex infections in man. J. Dermatol. Sci. 1994, 8, 224–232. [Google Scholar] [CrossRef]
  63. Ranjbar, Z.; Zahed, M.; Ranjbar, M.A.; Shirmardan, Z. Comparative study of serum zinc concentration in recurrent herpes labialis patients and healthy individuals. BMC Oral Health 2020, 20, 296. [Google Scholar] [CrossRef]
  64. Godfrey, H.R.; Godfrey, N.J.; Godfrey, J.C.; Riley, D. A randomized clinical trial on the treatment of oral herpes with topical zinc oxide/glycine. Altern. Ther. Health Med. 2001, 7, 49–56. [Google Scholar] [PubMed]
  65. Eby, G.A.; Halcomb, W.W. Use of topical zinc to prevent recurrent herpes simplex infection: Review of literature and suggested protocols. Med. Hypotheses 1985, 17, 157–165. [Google Scholar] [CrossRef]
  66. Barman, N.; Haque, M.A.; Uddin, M.N.; Ghosh, D.; Rahman, M.W.; Islam, M.T.; Rahman, M.Q.; Rob, M.A.; Hossain, M.A. Status of Serum Zinc in Multidrug Resistant Tuberculosis. Mymensingh Med. J. 2016, 25, 27–30. [Google Scholar] [PubMed]
  67. Kamakura, M.; Goshima, F.; Luo, C.; Kimura, H.; Nishiyama, Y. Herpes simplex virus induces the marked up-regulation of the zinc finger transcriptional factor INSM1, which modulates the expression and localization of the immediate early protein ICP0. Virol. J. 2011, 8, 257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Öztekin, A.; Öztekin, C. Vitamin D Levels in Patients with Recurrent Herpes Labialis. Viral Immunol. 2019, 32, 258–262. [Google Scholar] [CrossRef]
  69. Stock, C.; Guillén-Grima, F.; de Mendoza, J.H.; Marin-Fernandez, B.; Aguinaga-Ontoso, I.; Krämer, A. Risk factors of herpes simplex type 1 (HSV-1) infection and lifestyle factors associated with HSV-1 manifestations. Eur. J. Epidemiol. 2001, 17, 885–890. [Google Scholar] [CrossRef]
  70. Glaser, R.; Kiecolt-Glaser, J.K. Chronic stress modulates the virus-specific immune response to latent herpes simplex virus Type 1. Ann. Behav. Med. 1997, 19, 78–82. [Google Scholar] [CrossRef]
  71. Chida, Y.; Mao, X. Does psychosocial stress predict symptomatic herpes simplex virus recurrence? A meta-analytic investigation on prospective studies. Brain Behav. Immun. 2009, 23, 917–925. [Google Scholar] [CrossRef]
  72. Nassaji, M.; Ghorbani, R.; Taheri, R.; Azizzadeh, M.; Abbassyan, F. Epidemiologic, clinical findings and risk factors of recurrent herpes labialis in healthy adult patients: A case-control study. Eur. Res. J. 2018, 5, 545–549. Available online: http://dergipark.gov.tr/doi/10.18621/eurj.384806 (accessed on 24 March 2021). [CrossRef] [Green Version]
  73. Logan, H.; Lutgendorf, S.K.; Hartwig, A.; Lilly, J.; Berberich, S.L. Immune, stress, and mood markers related to recurrent oral herpes outbreaks. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 1998, 86, 48–54. [Google Scholar] [CrossRef]
  74. Ives, A.M.; Bertke, A.S. Stress Hormones Epinephrine and Corticosterone Selectively Modulate Herpes Simplex Virus 1 (HSV-1) and HSV-2 Productive Infections in Adult Sympathetic, but Not Sensory, Neurons. J. Virol. 2017, 91, e00582-17. [Google Scholar] [CrossRef] [Green Version]
  75. Oakley, C.; Epstein, J.B.; Sherlock, C.H. Reactivation of oral herpes simplex virus: Implications for clinical management of herpes simplex virus recurrence during radiotherapy. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 1997, 84, 272–278. [Google Scholar] [CrossRef]
  76. Gangemi, A.C.; Choi, S.H.; Yin, Z.; Feurdean, M. Cytomegalovirus and Herpes Simplex Virus Co-Infection in an HIV-Negative Patient: A Case Report. Cureus 2021, 13, e13214. [Google Scholar] [CrossRef]
  77. Danziger, S. Ice-packs for cold-sores. Lancet 1978, 1, 103. [Google Scholar] [CrossRef]
  78. De Simone, I.I.E. Preventing lip problems. J. Mod. Pharm. 1997, 4, 24. [Google Scholar]
  79. Shulman, J.D.; Lewis, D.L.; Carpenter, W.M. The prevalence of chapped lips during an army hot weather exercise. Mil. Med. 1997, 162, 817–819. [Google Scholar] [CrossRef] [Green Version]
  80. Lundeen, R.C.; Langlais, R.P.; Terezhalmy, G.T. Sunscreen protection for lip mucosa: A review and update. J. Am. Dent. Assoc. 1985, 111, 617–621. [Google Scholar] [CrossRef]
  81. Rezazadeh, F.; Moshaverinia, M.; Motamedifar, M.; Alyaseri, M. Assessment of Anti HSV-1 Activity of Aloe Vera Gel Extract: An In Vitro Study. J. Dent. 2016, 17, 49–54. [Google Scholar]
  82. Zschocke, I.; Reich, C.; Zielke, A.; Reitmeier, N.; Reich, K. Silica gel is as effective as acyclovir cream in patients with recurrent herpes labialis: Results of a randomized, open-label trial. J. Dermatol. Treat. 2008, 19, 176–181. [Google Scholar] [CrossRef]
  83. Russell, A.S.; Brisson, E.; Grace, M. A Double-Blind, Controlled Trial of Levamisole in the Treatment of Recurrent Herpes Labialis. J. Infect. Dis. 1978, 137, 597–600. [Google Scholar] [CrossRef]
  84. Inglot, D. Comparisons of the antiviral activity in vitro of some non-steroidal anti-inflammatory drugs. J. Gen. Virol. 1969, 4, 203–214. [Google Scholar] [CrossRef] [PubMed]
  85. Astani, A.; Albrecht, U.; Schnitzler, P. Piroxicam inhibits herpes simplex virus type 1 infection in vitro. Pharmazie 2015, 70, 331–336. [Google Scholar] [PubMed]
  86. Kaminester, L.H.; Pariser, R.J.; Pariser, D.M.; Weiss, J.S.; Shavin, J.S.; Landsman, L.; Haines, H.G.; Osborne, D.W. A double-blind, placebo-controlled study of topical tetracaine in the treatment of herpes labialis. J. Am. Acad. Dermatol. 1999, 41, 996–1001. [Google Scholar] [CrossRef] [PubMed]
  87. Milman, N.; Scheibel, J.; Jessen, O. Prevention of recurrent herpes simplex labialis with lysine. Ugeskr. Laeger 1980, 142, 1202–1203. [Google Scholar] [PubMed]
  88. Crimi, S.; Fiorillo, L.; Bianchi, A.; D’Amico, C.; Amoroso, G.; Gorassini, F.; Mastroieni, R.; Marino, S.; Scoglio, C.; Catalano, F.; et al. Herpes Virus, Oral Clinical Signs and QoL: Systematic Review of Recent Data. Viruses 2019, 11, 463. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6563194/ (accessed on 20 March 2021). [CrossRef] [Green Version]
  89. Thein, D.J.; Hurt, W.C. Lysine as a prophylactic agent in the treatment of recurrent herpes simplex labialis. Oral Surg. Oral Med. Oral Pathol. 1984, 58, 659–666. [Google Scholar] [CrossRef]
  90. Szapary, P.; Cirigliano, M. Lysine for the Prevention and Treatment of Cutaneous Herpes Simplex Infections. Available online: https://www.reliasmedia.com/articles/42145-lysine-for-the-prevention-and-treatment-of-cutaneous-herpes-simplex-infections?v=preview (accessed on 26 August 2021).
  91. Pedrazini, M.C.; Araújo, V.C.; Montalli, V.A.M. The effect of L-Lysine in recurrent herpes labialis: Pilot study with a 8-year follow up. RGO-Rev. Gaúcha Odontol. 2018, 66, 245–249. [Google Scholar] [CrossRef] [Green Version]
  92. Femiano, F.; Gombos, F.; Scully, C. Recurrent herpes labialis: A pilot study of the efficacy of zinc therapy. J. Oral Pathol. Med. 2005, 34, 423–425. [Google Scholar] [CrossRef]
  93. Cunningham, A.; Griffiths, P.; Leone, P.; Mindel, A.; Patel, R.; Stanberry, L.; Whitley, R. Current management and recommendations for access to antiviral therapy of herpes labialis. J. Clin. Virol. Off. Publ. Pan Am. Soc. Clin. Virol. 2012, 53, 6–11. [Google Scholar] [CrossRef] [Green Version]
  94. Sacks, S.L.; Thisted, R.A.; Jones, T.M.; Barbarash, R.A.; Mikolich, D.J.; Ruoff, G.E.; Jorizzo, J.L.; Gunnilla, L.B.; Katz, D.H.; Khalil, M.H.; et al. Clinical efficacy of topical docosanol 10% cream for herpes simplex labialis: A multicenter, randomized, placebo-controlled trial. J. Am. Acad. Dermatol. 2001, 45, 222–230. [Google Scholar] [CrossRef]
  95. Treister, N.S.; Woo, S.B. Topical n-docosanol for management of recurrent herpes labialis. Expert Opin. Pharmacother. 2010, 11, 853–860. [Google Scholar] [CrossRef]
  96. Kleymann, G. Novel agents and strategies to treat herpes simplex virus infections. Expert Opin. Investig. Drugs 2003, 12, 165–183. [Google Scholar] [CrossRef]
  97. Nath, A.; Thappa, D. Newer trends in the management of genital herpes. Indian J. Dermatol. Venereol. Leprol. 2009, 75, 566–574. [Google Scholar]
  98. Trevor, A.J.; Katzung, B.G.; Kruidering-Hall, M.M.; Masters, S.B. Chapter 49. Antiviral Chemotherapy & Prophylaxis|Katzung & Trevor’s Pharmacology: Examination & Board Review, 10e|AccessPharmacy|McGraw Hill Medical. Available online: https://accesspharmacy.mhmedical.com/Content.aspx?bookid=514&sectionid=41817568 (accessed on 19 September 2021).
  99. Chen, F.; Xu, H.; Liu, J.; Cui, Y.; Luo, X.; Zhou, Y.; Chen, Q.; Jiang, L. Efficacy and safety of nucleoside antiviral drugs for treatment of recurrent herpes labialis: A systematic review and meta-analysis. J. Oral Pathol. Med. 2017, 46, 561–568. [Google Scholar] [CrossRef]
  100. Lawee, D.; Rosenthal, D.; Aoki, F.Y.; Portnoy, J. Efficacy and safety of foscarnet for recurrent orolabial herpes: A multicentre randomized double-blind study. CMAJ Can. Med. Assoc. J. 1988, 138, 329–333. [Google Scholar]
  101. Poole, C.L.; James, S.H. Antiviral Therapies for Herpesviruses: Current Agents and New Directions. Clin. Ther. 2018, 40, 1282–1298. [Google Scholar] [CrossRef]
  102. Seley-Radtke, K.L.; Yates, M.K. The evolution of nucleoside analogue antivirals: A review for chemists and non-chemists. Part 1: Early structural modifications to the nucleoside scaffold. Antivir. Res. 2018, 154, 66–86. [Google Scholar] [CrossRef]
  103. Álvarez, D.M.; Castillo, E.; Duarte, L.F.; Arriagada, J.; Corrales, N.; Farías, M.A.; Henríquez, A.; Agurto-Muñoz, C.; González, P.A. Current Antivirals and Novel Botanical Molecules Interfering with Herpes Simplex Virus Infection. Front. Microbiol. 2020, 11, 139. Available online: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7026011/ (accessed on 24 March 2021). [CrossRef]
  104. Sadowski, L.; Upadhyay, R.; Greeley, Z.; Margulies, B. Current Drugs to Treat Infections with Herpes Simplex Viruses-1 and -2. Viruses 2021, 13, 1228. [Google Scholar] [CrossRef]
  105. Wong, Y.J.; Veitz-Keenan, A. Systemic nucleoside antiviral agents may be effective in prevention of recurrent herpes labialis. Evid. Based Dent. 2013, 14, 54. [Google Scholar] [CrossRef]
  106. Mubareka, S.; Leung, V.; Aoki, F.Y.; Vinh, D.C. Famciclovir: A focus on efficacy and safety. Expert Opin. Drug Saf. 2010, 9, 643–658. [Google Scholar] [CrossRef] [PubMed]
  107. Aribi Al-Zoobaee, F.W.; Yee Shen, L.; Veettil, S.K.; Gopinath, D.; Maharajan, M.K.; Kunnath Menon, R. Antiviral Agents for the Prevention and Treatment of Herpes Simplex Virus Type-1 Infection in Clinical Oncology: A Network Meta-Analysis. Int. J. Environ. Res. Public Health 2020, 17, 8891. [Google Scholar] [CrossRef] [PubMed]
  108. Crumpacker, C. The pharmacological profile of famciclovir. Semin. Dermatol. 1996, 15 (Suppl. 1), 14–26. [Google Scholar] [PubMed]
  109. Birkmann, A.; Zimmermann, H. HSV antivirals—Current and future treatment options. Curr. Opin. Virol. 2016, 18, 9–13. [Google Scholar] [CrossRef] [PubMed]
  110. Rahimi, H.; Mara, T.; Costella, J.; Speechley, M.; Bohay, R. Effectiveness of antiviral agents for the prevention of recurrent herpes labialis: A systematic review and meta-analysis. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2012, 113, 618–627. [Google Scholar] [CrossRef]
  111. Bermek, O.; Williams, R.S. The three-component helicase/primase complex of herpes simplex virus-1. Open Biol. 2021, 11, 210011. [Google Scholar] [CrossRef]
  112. Ainbinder, D.; Paolino, D.; Fresta, M.; Touitou, E. Drug Delivery Applications with Ethosomes. J. Biomed. Nanotechnol. 2010, 6, 558–568. [Google Scholar] [CrossRef]
  113. Horwitz, E.; Pisanty, S.; Czerninski, R.; Helser, M.; Eliav, E.; Touitou, E. A clinical evaluation of a novel liposomal carrier for acyclovir in the topical treatment of recurrent herpes labialis. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod. 1999, 87, 700–705. [Google Scholar] [CrossRef]
  114. Nainwal, N.; Jawla, S.; Singh, R.; Saharan, V.A. Transdermal applications of ethosomes—A detailed review. J. Liposome Res. 2019, 29, 103–113. [Google Scholar] [CrossRef]
  115. Wang, L.; Hu, C.; Shao, L. The antimicrobial activity of nanoparticles: Present situation and prospects for the future. Int. J. Nanomed. 2017, 12, 1227–1249. [Google Scholar] [CrossRef] [Green Version]
  116. Tavakoli, A.; Ataei-Pirkooh, A.; Mm Sadeghi, G.; Bokharaei-Salim, F.; Sahrapour, P.; Kiani, S.J.; Moghoofei, M.; Farahmand, M.; Javanmard, D.; Monavari, S.H. Polyethylene glycol-coated zinc oxide nanoparticle: An efficient nanoweapon to fight against herpes simplex virus type 1. Nanomedicine 2018, 13, 2675–2690. [Google Scholar] [CrossRef]
  117. Münstedt, K. Bee products and the treatment of blister-like lesions around the mouth, skin and genitalia caused by herpes viruses—A systematic review. Complement. Ther. Med. 2019, 43, 81–84. [Google Scholar] [CrossRef]
  118. Jautová, J.; Zelenková, H.; Drotarová, K.; Nejdková, A.; Grünwaldová, B.; Hladiková, M. Lip creams with propolis special extract GH 2002 0.5% versus aciclovir 5.0% for herpes labialis (vesicular stage): Randomized, controlled double-blind study. Wien. Med. Wochenschr. 2019, 169, 193–201. [Google Scholar] [CrossRef] [Green Version]
  119. Semprini, A.; Singer, J.; Braithwaite, I.; Shortt, N.; Thayabaran, D.; McConnell, M.; Weatherall, M.; Beasley, R. Kanuka honey versus aciclovir for the topical treatment of herpes simplex labialis: A randomised controlled trial. BMJ Open 2019, 9, e026201. [Google Scholar] [CrossRef] [Green Version]
  120. Garber, A.; Barnard, L.; Pickrell, C. Review of Whole Plant Extracts with Activity Against Herpes Simplex Viruses In Vitro and In Vivo. J. Evid.-Based Integr. Med. 2021, 26, 2515690X20978394. [Google Scholar] [CrossRef]
  121. de Paula Eduardo, C.; Aranha, A.C.C.; Simões, A.; Bello-Silva, M.S.; Ramalho, K.M.; Esteves-Oliveira, M.; de Freitas, P.M.; Marotti, J.; Tunér, J. Laser treatment of recurrent herpes labialis: A literature review. Lasers Med. Sci. 2013, 29, 1517–1529. [Google Scholar] [CrossRef]
  122. Al-Maweri, S.A.; Kalakonda, B.; AlAizari, N.A.; Al-Soneidar, W.A.; Ashraf, S.; Abdulrab, S.; Al-Mawri, E.S. Efficacy of low-level laser therapy in management of recurrent herpes labialis: A systematic review. Lasers Med. Sci. 2018, 33, 1423–1430. [Google Scholar] [CrossRef]
  123. Palli, M.A.; McTavish, H.; Kimball, A.; Horn, T.D. Immunotherapy of Recurrent Herpes Labialis with Squaric Acid. JAMA Dermatol. 2017, 153, 828. [Google Scholar] [CrossRef]
  124. Chang, A.L.S.; Honari, G.; Guan, L.; Zhao, L.; Palli, M.A.; Horn, T.D.; Dudek, A.Z.; McTavish, H. A phase 2, multicenter, placebo-controlled study of single-dose squaric acid dibutyl ester to reduce frequency of outbreaks in patients with recurrent herpes labialis. J. Am. Acad. Dermatol. 2020, 83, 1807–1809. [Google Scholar] [CrossRef]
  125. Leplina, O.; Starostina, N.; Zheltova, O.; Ostanin, A.; Shevela, E.; Chernykh, E. Dendritic cell-based vaccines in treating recurrent herpes labialis: Results of pilot clinical study. Hum. Vaccines Immunother. 2016, 12, 3029–3035. [Google Scholar] [CrossRef] [Green Version]
  126. Janicki, M.P. Recurrent Herpes Labialis and Recurrent Aphthous Ulcerations: Psychological Components. Psychother. Psychosom. 1971, 19, 288–294. [Google Scholar] [CrossRef]
  127. Harper, A.; Vijayakumar, V.; Ouwehand, A.C.; Ter Haar, J.; Obis, D.; Espadaler, J.; Binda, S.; Desiraju, S.; Day, R. Viral Infections, the Microbiome, and Probiotics. Front. Cell Infect. Microbiol. 2021, 10, 596166. [Google Scholar] [CrossRef] [PubMed]
  128. Abt, M.C.; Osborne, L.C.; Monticelli, L.A.; Doering, T.A.; Alenghat, T.; Sonnenberg, G.F.; Paley, M.A.; Antenus, M.; Williams, K.L.; Erikson, J.; et al. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity 2012, 37, 158–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Erttmann, S.F.; Swacha, P.; Aung, K.M.; Brindefalk, B.; Jiang, H.; Härtlova, A.; Uhlin, B.E.; Wai, S.N.; Gekara, N.O. The gut microbiota prime systemic antiviral immunity via the cGAS-STING-IFN-I axis. Immunity 2022, 55, 847–861. [Google Scholar] [CrossRef]
  130. Torres, N.I.; Noll, K.S.; Xu, S.; Li, J.; Huang, Q.; Sinko, P.J.; Wachsman, M.B.; Chikindas, M.L. Safety, Formulation and In Vitro Antiviral Activity of the Antimicrobial Peptide Subtilosin Against Herpes Simplex Virus Type 1. Probiotics Antimicrob. Proteins 2013, 5, 26–35. [Google Scholar] [CrossRef] [Green Version]
  131. Buske-Kirschbaum, A.; Geiben, A.; Wermke, C.; Pirke, K.M.; Hellhammer, D. Preliminary Evidence for Herpes labialis Recurrence following Experimentally Induced Disgust. Psychother. Psychosom. 2001, 70, 86–91. [Google Scholar] [CrossRef]
  132. Hu, S.; Anand, P.; Laughter, M.; Maymone, M.B.C.; Dellavalle, R.P. Holistic dermatology: An evidence-based review of modifiable lifestyle factor associations with dermatologic disorders. J. Am. Acad. Dermatol. 2020, 86, 868–877. Available online: https://www.sciencedirect.com/science/article/pii/S0190962220307246 (accessed on 29 August 2021). [CrossRef]
  133. Gompf, S.G. Cold Sores (Oral Herpes) Treatment, Remedies, Causes, Medicines, Symptoms. MedicineNet. Available online: https://www.medicinenet.com/herpes_simplex_infections_non-genital/article.htm (accessed on 20 March 2021).
  134. Jamgochian, M.; Alamgir, M.; Rao, B. Diet in Dermatology: Review of Diet’s Influence on the Conditions of Rosacea, Hidradenitis Suppurativa, Herpes Labialis, and Vitiligo. Am. J. Lifestyle Med. 2021, 17, 15598276211026592. [Google Scholar] [CrossRef]
Table 1. Commonly reported trigger/risk factors for recurrent herpes labialis.
Table 1. Commonly reported trigger/risk factors for recurrent herpes labialis.
• Fever• Orofacial fracture
• Flu• Emotional/psychosocial stress
• Exposure to sun/UV light• Upper respiratory issues
• Seasonal weather• Immunosuppressive treatments
• Chapped lips/trauma• Chemotherapy
• Fatigue• Organ transplantations
• Lack of sleep• Additional viral infections
Table 2. List of early preventive/management interventions [96].
Table 2. List of early preventive/management interventions [96].
• Ice cubes• Anesthetics
• Lip balms/moisturizers• Lysine
• Lip sunscreens• Zinc and iron supplementations
• Aloe vera gel• Vitamin D supplementations
• Silica gel• Zinc-based creams
• Levamisole• Docosanol
• Antipyretics• Foscarnet
• Painkillers
Table 3. Categorization of nucleoside analogs of antiviral drugs for various herpesvirus indications and targeted mechanisms [96,97].
Table 3. Categorization of nucleoside analogs of antiviral drugs for various herpesvirus indications and targeted mechanisms [96,97].
Product
(Brands
Available)
CategoryTargeted
Mechanisms
Recommended
Dosage/Routes
Indication/
Viral Spectrum
Acyclovir
(Zovirax)
Purine analogTK, DNA polymerase, chain termination with competitive dGTPOral: 5 × 200–800 mg (5–14 days)
Topical: 5% ointment/cream
HSV, VZV, HCMV
Valacyclovir (Valtrex)Purine analog, prodrug for ACVTK, DNA polymeraseOral: 2 × 500 mg−1 g (5–10 days)HSV, VZV, HCMV
Famciclovir (Famvir)Purine analog, prodrug for PCVTK, DNA polymeraseOral: 2 × 125–250 mg (5–10 days)HSV, VZV
Penciclovir
(Denavir/Vectavir)
Purine analogTK, DNA polymerase, no chain termination but with competitive dGTPTopical: 1% creamHSV, VZV
Abbreviations: ACV = Acyclovir; VCV = Valacyclovir; FCV = Famciclovir; PCV = Penciclovir; TK = thymidine kinase; DNA = deoxyribonucleic acid; dGTP = deoxyguanosine triphosphate; HSV = herpes simplex virus; VZV = Varicella-zoster virus; HCMV = human cytomegalovirus.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gopinath, D.; Koe, K.H.; Maharajan, M.K.; Panda, S. A Comprehensive Overview of Epidemiology, Pathogenesis and the Management of Herpes Labialis. Viruses 2023, 15, 225. https://doi.org/10.3390/v15010225

AMA Style

Gopinath D, Koe KH, Maharajan MK, Panda S. A Comprehensive Overview of Epidemiology, Pathogenesis and the Management of Herpes Labialis. Viruses. 2023; 15(1):225. https://doi.org/10.3390/v15010225

Chicago/Turabian Style

Gopinath, Divya, Kim Hoe Koe, Mari Kannan Maharajan, and Swagatika Panda. 2023. "A Comprehensive Overview of Epidemiology, Pathogenesis and the Management of Herpes Labialis" Viruses 15, no. 1: 225. https://doi.org/10.3390/v15010225

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