You are on page 1of 21

Biochemical Pharmacology 206 (2022) 115322

Contents lists available at ScienceDirect

Biochemical Pharmacology
journal homepage: www.elsevier.com/locate/biochempharm

Review

Acyclovir resistance in herpes simplex viruses: Prevalence and


therapeutic alternatives
Hanna H. Schalkwijk , Robert Snoeck , Graciela Andrei *
Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium

A R T I C L E I N F O A B S T R A C T

Keywords: Herpes simplex viruses (HSV), the causative agents of recurrent orofacial and anogenital infections, can cause
Immunocompromised significant morbidity and mortality in both immunocompetent and immunocompromised individuals. In
Immune-privileged sites immunocompromised patients, HSV tends to be more persistent with chance of dissemination. The nucleoside
Herpes simplex virus
analogue acyclovir has drastically improved the management of HSV infections although acyclovir resistant
Acyclovir
strains have been reported in the clinic. We performed a systematic search to summarize the prevalence data
Acyclovir resistance
Prevalence reported in both the immunocompetent and immunocompromised populations. Defining the global prevalence of
acyclovir resistance in HSV infections is hampered by the high variability in methodology, patient selection,
study design, and treatment history among the studies. Acyclovir resistant HSV is infrequent in the immuno­
competent population (generally below 1%), irrespective of treatment history. Exceptions are infections at
immune-privileged sites such as the cornea, where frequent recurrences and extensive acyclovir therapy favor
the emergence of acyclovir resistance. Higher frequencies of acyclovir resistant HSV infections are reported
among immunocompromised individuals, with the highest prevalence seen among hematopoietic stem cell
transplant recipients. All antivirals approved for the treatment of HSV infections have the same target, i.e. the
viral DNA polymerase, and cross-resistance to different antivirals has been described, complicating therapy of
acyclovir resistant strains. In this review we will discuss acyclovir mode of action, mechanisms of resistance,
prevalence of resistance, and alternative antiviral treatments for acyclovir resistant HSV infections.

1. Introduction human cytomegalovirus (CMV), HHV-6A, HHV-6B, and HHV-7. The


gammaherpesvirinae contains Epstein-Barr virus (EBV) and HHV-8 or
The Herpesviridae family includes a large group of double-stranded Kaposi’s sarcoma-associated herpesvirus (KSHV) [3]. All HHV infections
DNA viruses that can infect a wide range of animal species [1]. Her­ can cause severe disease in immunocompromised hosts such as lym­
pesviruses show narrow host-specificity indicating evolution with their phoproliferative disorders associated with EBV or KSHV and dissemi­
hosts over long periods of time [2]. A common characteristic is their nated infections (VZV, CMV, or HSV) [4,5]. HSV-1 and HSV-2 can also
ability to establish lifelong latent infections, followed by periods of cause substantial morbidity during primary and recurrent infections in
reactivation upon diverse stimuli. Nine human herpesviruses (HHV) immunocompetent individuals [6]. Infections of the central nervous
have been identified which are classified into three subfamilies based on system (CNS) with herpesviruses are an important cause of morbidity
their genome organization, cell tropism, and growth characteristics: the and mortality and can lead to long-term neurologic sequelae. The most
alphaherpesvirinae, betaherpesvirinae, and gammaherpesvirinae. The significant CNS infections are due to HSV and CMV. CNS infection with
alphaherpesvirinae comprises herpes simplex virus 1 (HSV-1) and 2 (HSV- CMV occur mainly perinatally, though it may also be seen seldom in
2), and varicella zoster virus (VZV). The betaherpesvirinae includes children and adults, particularly in the immunocompromised

Abbreviations: ACV, acyclovir; ACV-MP, acyclovir monophosphate; ACVr, acyclovir resistant; CMV, human cytomegalovirus; CNS, central nervous system; CPE,
cytopathic effect; DNA pol, DNA polymerase; dT, thymidine; dTMP, thymidine monophosphate; EC50, concentration inhibiting 50% of virus growth in cell culture;
HHV, human herpesvirus; HIV, human immunodeficiency virus; HSCT, hematopoietic stem cell transplant; HSE, herpes simplex encephalitis; HSV, herpes simplex
virus; HSV-1, herpes simplex virus 1; HSV-2, herpes simplex virus 2; SEM, skin, eye and/or mouth; TK, thymidine kinase; VACV, valacyclovir; VZV, varicella zoster
virus.
* Corresponding author at: Rega institute for Medical Research, Herestraat 49 bus 1043, Leuven, Belgium.
E-mail address: graciela.andrei@kuleuven.be (G. Andrei).

https://doi.org/10.1016/j.bcp.2022.115322
Received 31 August 2022; Received in revised form 20 October 2022; Accepted 20 October 2022
Available online 26 October 2022
0006-2952/© 2022 Elsevier Inc. All rights reserved.
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

population. CNS infection with HSV can occur in neonates as well as in 2.3. Neonatal herpes
older children and adults [7].
The discovery of the nucleoside analogue acyclovir (ACV) was a Although neonatal herpes is relatively rare, it is a potentially
milestone in the management of HSV infections. However, frequent use devastating complication of anogenital herpes during pregnancy. HSV
of ACV increased the incidence of ACV resistant (ACVr) HSV infections. transmission to the fetus commonly occurs through exposure of the
Drug resistance in herpes simplex viruses poses a major concern in the neonate to HSV-1 or HSV-2, shed in the genital tract during delivery (85
immunocompromised host as they often require long-term antiviral %). Besides perinatal infection, HSV may be acquired by intrauterine (5
therapy, and this, in combination with ongoing viral replication in­ %) or post-partum infection (10 %) [23]. HSV-2 is responsible for about
creases the risk of drug resistance emergence. The limited number of two-thirds of the cases with HSV-1 accounting for the remains [24]. Risk
antivirals available for the therapy of HSV infections, all having the of transmission is the highest when primary anogenital herpes develops
same drug target (i.e., the viral DNA polymerase), makes the manage­ late during pregnancy [22,24]. The global rate of neonatal herpes is
ment of HSV drug resistance in the clinic challenging. In this review we estimated at 10 per 100.000 live births [24].
summarize the prevalence data on ACV resistance in herpes simplex Neonatal herpes can be classified as (i) localized skin, eye and/or
viruses. We also describe clinical manifestations of HSV infections, mouth (SEM) disease accounting for 45 % of neonatal herpes, (ii) CNS
mechanisms of ACV resistance, and therapeutic alternatives. disease, accounting for 30 % of cases, and as (iii) disseminated disease
that accounts for 25 % of cases [25]. In SEM disease, lesions are confined
2. Herpes simplex virus infections to the skin, eye, and/or mouth, whereas neonates with CNS disease
present irritability, poor feeding, seizures, and/or temperature insta­
HSV infections can have a variety of clinical manifestations. HSV-1 is bility with or without SEM involvement. Disseminated disease involves
typically transmitted during childhood and predominantly causes oro­ multiple organs including the CNS, lungs, liver, skin, eye, and/or mouth
facial herpes but can also cause anogenital herpes. HSV-2, mostly [26]. SEM disease has no attributable mortality, whereas the mortality
sexually transmitted, causes anogenital herpes and infrequently causes rate due to disseminated disease is 85 % when left untreated [27,28].
orofacial herpes. In 2016, the global seroprevalence of HSV-1 was esti­ Due to antiviral treatment, mortality rates have been reduced to ~ 5 %
mated at 66.6 % in the population aged 0–49 years and seroprevalence in neonates with CNS disease and to ~ 30 % in neonates with dissemi­
of HSV-2 in 15–49-year-olds was estimated at 13.2 % [8]. Seropreva­ nated disease [29,30].
lence increases with age and the highest seroprevalence of HSV-1 and
HSV-2 is reported in Africa [8–10]. Women are more likely to become 2.4. Herpetic keratitis
infected with HSV-2, possibly due to a greater biological susceptibility
[8,10,11]. Several clinical manifestations of HSV infections will be dis­ Herpetic eye infections can have different manifestations including
cussed in more detail in the following sections. blepharitis, conjunctivitis, retinitis, and herpetic keratitis [31]. Herpetic
keratitis, mostly associated with HSV-1, manifests predominantly as
2.1. Orofacial herpes epithelial keratitis, limited to the superficial cornea, or stromal keratitis
[31,32]. In 2016, the incidence of herpetic keratitis was estimated at 24
Primary orofacial HSV infections are often asymptomatic, but cases per 100.000 person-years, giving an estimated 1.7 million cases of
symptomatic infection is characterized by fever and lesions at the buccal herpetic keratitis worldwide [33]. Herpetic keratitis is an important
and gingival mucosa. Intraoral lesions are mostly associated with pri­ cause of visual impairment due to its recurrent nature, and annually
mary infection while lip lesions (cold sores) are suggestive for re­ causes 40.000 new cases of monocular visual impairment [34,35]. HSV-
currences [12]. Recurrences with HSV-1 occur in approximately 40 % of 1 is considered the leading cause of infectious blindness in Western
infected individuals, while orofacial HSV-2 recurs in only 4 % of the countries.
cases [13]. Recurrent orofacial herpes is preceded by pain, burning,
tingling, or itching after which vesicles appear and is often self-limiting, 2.5. Herpes simplex encephalitis (HSE)
with lesions usually healing after 8 to 10 days [12].
Herpes simplex encephalitis, mostly caused by HSV-1, has an esti­
2.2. Anogenital herpes mated incidence of one case in 250.000–500.000 individuals per year
and is the most common cause of sporadic infectious encephalitis
Anogenital herpes is mostly caused by HSV-2, and less frequently by [36–39]. Common symptoms of HSE are changes in mental status,
HSV-1 although the proportion of anogenital herpes caused by HSV-1 is abnormal behavior, fever, headache, and seizures but clinical presen­
increasing, particularly among young people in the Americas and Eu­ tation differs among patients, making diagnosis difficult [40,41].
ropean region [8,14,15]. Possible explanations are the decrease in HSV- Without treatment, the mortality rate of HSE is 70 % and most survivors
1 acquisition during childhood and an increased frequency of oral sex suffer from severe neurologic sequelae [12]. Neurologic sequelae remain
[16,17]. Anogenital HSV shedding and transmission to sexual partners common despite antiviral treatment, especially when treatment is
can take place even in the absence of symptoms [12]. Anogenital herpes delayed [40].
infections with HSV-1 and HSV-2 are clinically indistinguishable. The
clinical presentation is characterized by painful, vesicular, and ulcera­ 2.6. HSV infection in the immunocompromised host
tive lesions on the external genitalia, perianal region, buttocks, or upper
thighs. Lesions cause pain, itching, burning, and dysuria. Patients with Both innate and adaptive immune responses play a pivotal role in
primary infection can additionally present with fever, headache, malaise determining the severity of HSV infections and frequency of reactivation
and myalgia, although primary infections usually remain asymptomatic [42]. Immunocompromised patients are at risk for developing severe
[18]. HSV-2 causes frequent recurrent anogenital outbreaks, especially and potentially life-threatening HSV infections, particularly when cell-
in the first year, typically lasting for 8 to 10 days, whereas HSV-1 re­ mediated immune responses are reduced, as is the case in hematopoi­
currences tend to be milder and less frequent [19–21]. In most patients, etic stem cell transplant (HSCT) recipients and in human immunodefi­
the rate of reactivation decreases with time, but some patients continue ciency virus (HIV) infected individuals [43]. Latent HSV frequently
to have recurrences for>10 years [19,22]. reactivates in immunocompromised individuals, particularly in HSCT
recipients, of whom 80 % experience HSV reactivation [43–45]. Most
HSV infections in immunocompromised hosts locate to the orofacial and
anogenital sites and may have an atypical appearance with extensive

2
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

lesions, slow healing, more invasive disease, and risk of viral dissemi­
nation [43,46,47]. Therefore, prophylactic or therapeutic antiviral
treatment is often required in immunocompromised patients to prevent
morbidity and mortality.

3. Acyclovir

3.1. Mode of action

Since its introduction in the 1980 s, the nucleoside analogue


acyclovir [9-(2-hydroxyethoxymethyl)guanine] has become the first
line antiviral for prophylaxis and treatment of HSV infections. Acyclovir
is an acyclic guanosine analogue that lacks both the 2′ - and 3′ -OH
moieties and the 2′ - and 3′ -carbons [48] (Fig. 1). Three phosphorylation
steps are required to obtain the active (triphosphate) form of ACV, the
first one being performed by the virus-encoded thymidine kinase (TK)
[49]. The two successive phosphorylation steps are carried out by the
cellular guanosine monophosphate (GMP) kinase and nucleoside
diphosphate (NDP) kinase, respectively [50–52] (Fig. 1). Acyclovir
triphosphate (ACV-TP) competes with deoxyguanosine triphosphate
(dGTP) as substrate of the viral DNA polymerase (DNA pol) and is
incorporated into the elongating DNA chain [53]. Since incorporated
ACV monophosphate (ACV-MP) lacks a 3′ -hydroxyl moiety, its incor­
poration results in chain termination [54]. ACV-MP cannot be efficiently
excised by the 3′ -5′ exonuclease activity of the viral DNA pol due to its
altered sugar structure that impedes the hydrolysis reaction [55].
Acyclovir is highly selective due to its dependence on viral TK activity
for activation and a 100-fold more efficient recognition and incorpora­
tion of ACV-TP by the HSV DNA pol in comparison to cellular DNA
polymerases [56–58]. The major limitation of ACV is its poor oral
bioavailability (10 to 20 %), likely due to inadequate absorption of the
drug. Therefore, the prodrug valacyclovir (VACV), an L-valyl-ester of
ACV, has been developed with a three- to fivefold higher bioavailability
than ACV [59,60] (Fig. 1).

3.2. Clinical use of acyclovir

Topical formulations of ACV are available for the treatment of


recurrent herpes labialis (cold sores) and herpetic keratitis [61,62]. Oral
ACV and VACV are used for treatment of primary and recurrent ano­
genital HSV infections and as chronic suppressive therapy to prevent
recurrent anogenital herpes [63,64]. Therapy with ACV or VACV
effectively reduces symptomatic recurrences of anogenital herpes and
asymptomatic shedding, although the suppression of viral replication is
incomplete [65,66]. Prophylactic therapy is frequently used in severe Fig. 1. Mode of acyclovir activation. Hydrolysis converts the prodrug valacy­
immunosuppressed patients, including HSCT recipients, and is highly clovir to acyclovir. Three phosphorylation steps are required for acyclovir
effective in preventing reactivation [67–70]. Oral (V)ACV is often activation, the first one performed by the herpes simplex virus (HSV) thymidine
indicated for therapy of mucocutaneous HSV infections in immuno­ kinase (TK), confining its action to viral-infected cells. The two subsequent
compromised individuals. Intravenous formulations of ACV are used to phosphorylations are ensured by the cellular guanosine monophosphate (GMP)
treat severe HSV infections, including neonatal herpes, encephalitis, and kinase and nucleoside diphosphate (NDP) kinase. In its triphosphate form
infections in immunocompromised patients [68]. (acyclovir-TP), acyclovir targets the viral DNA polymerase by inhibiting viral
DNA synthesis. Abbreviations: acyclovir-MP, acyclovir monophosphate;
acyclovir-DP, acyclovir diphosphate.
4. Acyclovir resistance

A lack of clinical response to ACV treatment a week after beginning 4.1. Viral thymidine kinase
of therapy can be suggestive of acquisition of drug resistance [63]. There
are several risk factors for the development of ACV resistance and those The UL23 genes of HSV-1 and HSV-2 encode for the 376 amino acid
include prolonged use of ACV, suboptimal dosing of ACV, (degree of) long TK proteins which share approximately 74 % sequence identity. Six
immunosuppression, and ongoing viral replication [4,71,72]. Acyclovir highly conserved regions (site 1 to site 6) have been identified among
resistance arises due to the acquisition of mutations in the UL23 gene, Herpesviridae thymidine kinases [75] (Fig. 2). The most important sites
encoding for the viral TK, and/or the UL30 gene, encoding for the DNA for enzyme activity are the ATP-binding site (site 1), nucleoside binding
pol. Acyclovir resistant HSV infections are most frequently associated site (site 4), and the highly conserved cysteine at codon 336 (HSV-1) and
with mutations in the TK gene [73,74]. 337 (HSV-2), which is essential to maintain the three-dimensional
conformation of the active site and contributes to the phosphorylation
efficiency of the viral enzyme [63,76].
Viral TK activity is dispensable in dividing cells but is essential for

3
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

Fig. 2. Schematic overview of the A) herpes simplex virus 1 (HSV-1) and B) herpes simplex virus 2 (HSV-2) thymidine kinase protein. Conserved regions are
represented by grey boxes. Number under the boxes indicate the codon position of these regions. Abbreviations: ATP, ATP-binding site; NBS, nucleoside-binding site.

viral replication and reactivation in non-proliferating cells, such as exonuclease, palm, fingers, and thumb domains (Fig. 3). Herpesvirus
neurons [77–79]. The HSV-1 and HSV-2 TK have a broad range of ac­ polymerases additionally possess a pre-NH2-terminal domain which is
tivity and can phosphorylate thymidine (dT), deoxycytidine (dC), required for efficient viral replication and latency establishment
deoxyguanosine (dG), and ACV, into their respective monophosphate although the precise role has not been established yet [91–94]. The palm
forms dTMP, dCMP, dGMP, and ACV-MP [77,80]. Furthermore, HSV and finger domains are involved in the catalytic activity of the enzyme
TKs exhibit thymidylate activity and can convert dTMP into its and binding of incoming nucleotides, while the thumb domain interacts
diphosphate dTDP, although HSV-2 TK exhibits a much lower rate of with the primer-template complex [95]. The 3′ -5′ -exonuclease domain
thymidylate activity than HSV-1 TK [81]. exhibits proofreading activity allowing the excision and correction of
Mutations in the TK that confer ACV resistance affect its enzymatic mismatched nucleotides during DNA replication, and is responsible for
activity and are associated with the following phenotypes: TK-negative the low mutation frequency of herpesviruses (1x10-7 to 1x10-8 muta­
mutants which completely lack TK activity (ACV- / dT-), TK-altered tions/base/infectious cycle) [96,97]. Seven regions of conserved se­
mutants (ACV- / dT 15–100 %) that retain the ability to phosphorylate quences have been identified in herpesvirus DNA pols that are numbered
dT but have an impaired ability to phosphorylate ACV, TK-low mutants I to VII based on their degree of conservation among the DNA pol, region
(ACV- / dT 1–15 %) that show decreased TK activity or production, and I being the most conserved [91] (Fig. 3). Besides, the HSV DNA pol also
TK-partial mutants (ACV 1–15 % / dT 100 %), exhibiting full dT phos­ contains the δ-region C, which is shared by polymerases having ho­
phorylation capacity and reduced ACV phosphorylation activity mology with the eukaryotic DNA polymerases δ.
[82–84]. Most ACVr TK mutants in the clinic exhibit a TK-negative or Acyclovir resistance associated mutations in the DNA pol are pri­
TK-low phenotype, but TK-partial and TK-altered mutant strains have marily located in the conserved regions of the palm, finger, and thumb
also been reported [82–85]. domains, favoring less ACV incorporation into the viral DNA [95].
Resistance associated mutations are located throughout the entire Mutations in the exonuclease domain are found less frequently and it is
UL23 gene and have been extensively described in several reviews proposed that they confer resistance by enhancing the nucleoside
[74,82,86,87]. The HSV TK gene presents several G- and C- homopoly­ analogue excision rate [96]. An overview of resistance mutations
mer stretches that are hot-spots for nucleotide insertions and deletions detected in the HSV DNA polymerases is available in several reviews
resulting in frameshifts and can potentially introduce premature stop [74,86,95]. Mutations in the DNA pol conferring ACV resistance
codons. Approximately 50 % and up to 80 % of the ACVr TK mutants in commonly show cross-resistance to the second-line treatment foscarnet
the clinic harbor insertions or deletions in these homopolymer stretches, (see section 6.2) [95,98].
while others carry nucleotide substitutions in conserved and non-
conserved regions introducing amino acid changes or stop codons (see 4.3. Diagnosis of antiviral drug-resistance
section 5.8) [73,83,88,89].
Laboratory diagnosis of antiviral resistance is essential to guide cli­
4.2. Viral DNA polymerase nicians towards different treatment options in case of therapy failure.
Both phenotypical and genotypical assays are available for the diagnosis
The UL30 genes of HSV-1 and HSV-2 encode for a DNA pol of 1235 of antiviral drug resistance. Genotypical analysis is based on the iden­
and 1240 amino acids long, respectively, that have a protein sequence tification of resistance mutations in the UL23 and/or UL30 gene and is
identity of approximately 90 % [90]. They share several functional almost exclusively performed by Sanger sequencing. Sequencing of both
domains with the type B DNA polymerase family, including the 3′ -5′ the UL23 and UL30 genes yields the most reliable results. Sanger

Fig. 3. Schematic overview of the A) herpes simplex virus 1 (HSV-1) and B) herpes simplex virus 2 (HSV-2) DNA polymerase protein. Proposed functions and
subdomains are shown. Conserved regions are represented by grey boxes. The numbers under the boxes indicate the codon position of these regions.

4
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

sequencing lacks sensitivity to detect minor resistant variants with a susceptibility. Articles published until May 2022 written in English,
frequency below 20 %. Recently an ultra-deep sequencing approach reporting prevalence data on ACVr HSV infections based on a minimum
using the Miseq Illumina platform has been implemented for HSV of 5 patients with specified immune status were selected. All included
resistance testing, enabling the detection of emerging resistant sub­ studies used phenotypical and/or genotypical assays to confirm ACV
populations at an earlier stage [99]. Although genotypic analysis is a fast resistance. Clinical trials and studies where patient selection was based
approach that does not require the growth of virus in cell culture, on suspected resistance were excluded.
interpretation can be complicated due to the high variability of the TK After deduplication, 5276 articles were screened based on title and
and DNA pol [74]. Novel mutations are continuously being identified in abstract and 174 articles were assessed for eligibility based on full-text.
the clinic, and phenotypical characterization or functional assays are Prevalence data from 43 included articles was extracted and subdivided
then required to confirm their effect on antiviral susceptibility. In the into immunocompetent individuals (21 articles), infections at immune-
study by Burrel et al. [89], 36 % of the clinical samples that were privileged sites such as the eye and CNS (see section 5.3, 6 articles), and
genotypically characterized for ACV resistance required additional the general immunocompromised population (12 articles). When spec­
phenotyping to interpret drug susceptibility due to the detection of ified, immunocompromised patients were subdivided into solid organ
formerly unreported or undefined mutations in the UL23 and/or UL30 transplant recipients (7 articles), HSCT recipients (16 articles), and HIV
genes. infected individuals (9 articles).
Phenotypical assays are used to determine the effective concentra­
tion of an antiviral inhibiting 50 % of virus growth in cell culture (EC50). 5.2. Immunocompetent population
As it is dependent on the propagation of the virus in cell culture,
phenotypical assays are often time-consuming and can only be per­ Twenty-one studies reported prevalence data on ACVr HSV infections
formed if the virus is successfully isolated in cell culture. The presence of in the immunocompetent population [71,73,108,109,111–127]
minor resistant subpopulations can be detected if the resistant virus (Table 1). The used methods and resistance cutoffs varied widely be­
exceeds 20 % and is not lost during growth in cell culture [100]. A va­ tween those studies and it is therefore difficult to define the true inci­
riety of phenotypical assays have been used to determine HSV multi­ dence of ACV resistance. The reported prevalence of ACVr HSV
plication in the presence of antivirals, including plaque reduction assays, infections in the immunocompetent population ranged from 0 % to 6.2
cytopathic effect (CPE) reduction assays, luciferase reporter assays, real- %. However, most studies, including multiple extensive screening sur­
time quantitative polymerase chain reaction (qPCR), colorimetric as­ veys, reported a prevalence under 1 %.
says, virus yield inhibition assays, and DNA hybridization assays Four studies reported frequencies of ACV resistance above 1 % with
[101–108]. The plaque reduction assay is considered the gold standard only the study from Collins et al. [109] reporting over 5 % prevalence in
for HSV drug susceptibility testing and uses the number of viral plaques the United States survey [108,109,123,125]. The studies from Collins
formed under various antiviral concentrations as readout, which is et al. and Varella et al. [125], reporting 6.2 %, and 4.2 % resistance,
somewhat subjective when counting very small plaques. The DNA hy­ respectively, used the dye uptake assay that has a high false-positive rate
bridization method quantifies the viral DNA produced in infected cells (section 4.3). Additionally, all patients included in the studies from
under ACV pressure as an assessment of antiviral susceptibility and the Collins et al. received ACV therapy, although the incidence of ACVr virus
obtained EC50 values correlate well with those obtained by plaque was similar to that observed in untreated patients. The study from Wang
reduction assays [71,104]. The dye uptake assay is a modification of the et al. [108] reported ACVr HSV strains in 3 out of 68 (4.4 %) immuno­
neutral red dye cell viability assay and measures the ability of an anti­ competent children when using the phenotypical method mVILA but
viral to inhibit cell damage induced by HSV replication [102]. The dye only one case (1.4 %) could be confirmed by Sanger sequencing.
uptake assay generally gives higher EC50 values than plaque reduction Schmidt et al. [123] reported a prevalence of 4.3 %, but scarce infor­
assays and its limitation is the high false-positive rate for resistance of 3 mation was available on clinical diagnosis, immunocompetence, and
% [109]. treatment status of the tested patients.
Many variables can influence the phenotypical results, such as the Acyclovir resistance occurred at similar frequencies in ACV treated
used assay system, cell line, viral inoculum concentration, range of and treatment naive patients (Table 1). Christophers et al. [116] re­
antiviral concentrations, and resistance breakpoint but efforts have been ported a prevalence of 0.67 % in ACV treated patients versus 0.42 % in
made to standardize the results [74,105]. The breakpoint used to define untreated patients. Fife et al. [117] reported similar frequencies of ACV
drug resistance differs between different laboratories, although an EC50 resistance before and after six years of suppressive ACV therapy in
of ≥ 2 µg/mL (8.9 µM) in plaque reduction assays is widely accepted as a immunocompetent patients with recurrent anogenital herpes. Thus,
breakpoint for resistance [110]. Other studies have used breakpoints despite the widespread availability of ACV over the past decades, ACV
between 1 and 3 µg/mL (4.4 to 13.3 µM), ≥3-fold increase in EC50 values resistance remains infrequent in the immunocompetent population.
compared to the mean EC50 of all isolates, or EC50 values > 10-fold Acyclovir resistant HSV infections in the immunocompetent popu­
higher than sensitive control strains [105]. HSV-1 is more susceptible to lation are usually self-limiting as the natural immune response can
ACV than HSV-2 and it is therefore advisable to use different resistance eliminate the virus that is not eradicated by antiviral therapy. Conse­
cutoff values for each HSV species [73,111–113]. quently, antiviral resistance often has a limited effect on disease
outcome in immunocompetent individuals, although exceptions have
5. Prevalence of acyclovir resistance been reported [111,128,129]. Cases of herpetic keratitis were included
in some studies [112,116]. As the cornea is an immune-privileged site,
5.1. Systematic search and article inclusion there is a high risk for development of drug resistance in patients
suffering from herpetic keratitis even though they do not have an
As discussed in section 4, several factors can increase the risk for impaired immunity.
emergence of ACV resistance, including long-term ACV use and immune
status. Hence, the prevalence of ACVr HSV infections varies among 5.3. Immune-privileged sites
different population groups. It is generally accepted that the prevalence
of ACVr HSV infections is higher in immunocompromised patients than Immune-privileged sites such as the cornea and CNS show locally
in immunocompetent patients. We reviewed the prevalence of ACV reduced immune responses. HSV infections at these sites are not self-
resistance reported in different populations by performing a systematic limiting and ACV susceptibility correlates with a clinical response.
search in PubMed, Embase, Web of Science, and Cochrane with several Therapy failure due to ACVr HSV infections can therefore ultimately
related keywords including herpes simplex, acyclovir, and resistance or lead to blindness in case of ocular infections and to neurologic sequelae

5
H.H. Schalkwijk et al.
Table 1
Characteristics of studies reporting on acyclovir resistance in herpes simplex virus among the immunocompetent population.
First author Publication Country HSV disease Treatment HSV N◦ patients N◦ N◦ patients N◦ N◦ HSV N◦ Method (cutoff
year status species patients with ACV samples positive resistant resistance)
tested resistant HSV isolates HSV
(%) (tested) isolates

Bacon [114] 2002 United Recurrent herpes Untreated & HSV-1 1803 1002a 2 (0.20 %) 1803 1087 (1002) 2 Plaque reduction assay
States labialis ACV treated (EC50 ≥ 2 µg/mL)
Boon [115] 2000 United Recurrent herpes Untreated or HSV-1 & 1304 924 1 (0.11 %) 1297 920 HSV-1 1 Plaque reduction assay
Kingdom labialis previously ACV HSV-2 2 HSV-2 0 (EC50 ≥ 2 µg/mL)
treated 2 HSV-1 & 0
HSV-2
Christophers 1998 United Genital herpes Untreated Untyped 708 708 3 (0.42 %) 760 760 3 Plaque reduction assay
[116] Kingdom Genital herpes ACV treated 149 149 1 (0.67 %) 162 162 1 (EC50 ≥ 3 µM)
Genital herpes, Unknown 918 918 1c (0.11 %) 944 944 1
herpetic keratitis, or
unknownb
Collins [109] 1993 Europe Unspecifiedb ACV treated Untyped 287 287 2 (0.70 %) 420 420 2 Plaque reduction assay
(EC50 ≥ 3 µM)
United 663 663 41 (6.2 %) 1448 1448 44 Dye uptake assay (EC50 ≥
States 3 µg/mL)
Danve- 2004 France Different herpetic Untreated & HSV-1 1237 1237 3 (0.24 %) 1326 1326 3 Dye uptake assay (EC50 ≥
Szatanekd infectionsb ACV treated 6.5 µM)
[111] HSV-2 618 618 3 (0.49 %) 664 664 13 Dye uptake assay (EC50 ≥
13.5 µM)
Englund [71] 1990 United Herpetic infections at Untreated & Untyped 59 59 0 (0 %) -e 61 0 DNA hybridization (EC50
States oral site or genital ACV treated ≥ 9 µM)
tract
Fife [117] 1994 United Recurrent genital History of ≥ 6 Untyped 239 4 113 4 Dye uptake assay (EC50 ≥
6

– –
States herpes years of chronic 3 µg/mL)
ACV therapy
Frobertd [73] 2014 France Different herpetic Unspecified HSV-1 & – – – 703 703 1 Dye uptake assay (EC50 ≥
infections in adultsb HSV-2 6.5 µM HSV-1, ≥13.5 µM
Different herpetic – – – 324 324 0 HSV-2) & Sanger
infections in childrenb sequencing of UL23 &
UL30
Gray [118] 1989 United Herpetic infections of Unspecified HSV-1 155 155 0 (0 %) 97 97 0 Dye uptake assay
Kingdom skin, mucous or HSV-2 66 66 0
genitals Untyped 4 4 0
Hasegawa 2001 Japan Genital herpes in Untreated & HSV-1 & 38 38 0 (0 %) 56 56 0 Plaque reduction assay
[119] women ACV treated HSV-2
Hashido [120] 1988 Japan Genital herpes in Unspecified HSV-1 & – – – 56 56 0 Plaque reduction assay
women HSV-2 (EC50 ≥ 13.32 µM)
Nugier [112] 1992 France Unspecifiedb Untreated & HSV-1 459 459 1c (0.22 %) 520 520 1 Dye uptake assay (EC50 ≥

Biochemical Pharmacology 206 (2022) 115322


ACV treated 1.5 µg/mL)
HSV-2 142 142 1 (0.70 %) 146 146 2 Dye uptake assay (EC50 ≥
3 µg/mL)
Reyes [121] 2003 United (suspected) Genital Untreated & HSV-1 – 189 0 (0 %) – 189 0 Plaque reduction assay
States herpes ACV treated HSV-2 – 1644 3 (0.18 %) – 1644 3 (EC50 ≥ 2 µg/mL)
Sangdara 2008 Thailand (suspected) Herpetic Unspecified HSV-1 & 148 – 0 155 155 (1 3 0) 0 Plaque reduction assay
[122] infections in genital or HSV-2 (EC50 ≥ 3 µg/mL)
non-genital areasb
Schmidtd 2015 Germany Different herpetic Untreated & HSV-1 276 276 12 (4.3 %) 276 276 12 Sanger sequencing of
[123] infectionsb ACV treated UL23 & UL30, Plaque
reduction assay (≥5x
EC50 control strain)
2005 368 368 1 (0.27 %) 389 389 1
(continued on next page)
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

or mortality in case of CNS infections [28,130].

ACV: acyclovir, EC50: effective concentration inhibiting 50% of virus growth in cell culture, HSV: herpes simplex virus. a includes 37 patients potentially immunocompromised based on subjects’ reported medication and
medical histories, b may include infections at immune-privileged sites, c resistant sample isolated from ocular swab, d Samples collected in laboratories from routine diagnostic services, e data not available, f only 1 out of
Six studies reporting prevalence data at immune-privileged sites

mVILA (˃3x EC50 control


control strain) & Sanger

Dye uptake assay (EC50


Plaque reduction assay

Plaque reduction assay

Plaque reduction assay


were included: five related to ocular infections [35,107,118,130,131],

Sanger sequencing of
Sequencing of UL23
ELVIRA (≥3x EC50

(EC50 ≥ 3 µg/mL)

(EC50 ≥ 2 µg/mL)
and one to CNS infections [132] (Table 2). The study of Mitterreiter et al.
Method (cutoff

[132] reported on 12 HSE patients, including 7 immunocompromised

UL23 & UL30


resistance)

˃2 µg/mL)
individuals, of which 11 received antiviral treatment. No ACV resistance
was reported but only sequencing of the UL23 (TK) gene was performed

strain)
due to limited availability of HSV DNA. Although ACVr HSV infections of
the CNS have been described in several case reports [133–135], other
HSE patient cohorts reporting on the prevalence of ACV resistance are
1 (EC50 =
resistant

2.29 µg/
lacking.
isolates
HSV

The prevalence of ACV resistance in ocular HSV infections ranged


mL)
N◦

0
from 0 % to 34.6 %. Menage et al. [107] and Gray et al. [118] reported
no resistance but included only limited number of patients (35, and 6,
260 (2 3 3)
respectively), and Gray et al. sampled patients before start of ACV
positive

(tested)
isolates
N◦ HSV

treatment. Acyclovir resistance was detected more frequently (6.4 % to

102
34.6 %) in patients with (recurrent) herpetic keratitis, most of whom
27

24

68

were receiving ACV treatment or had a history of ACV treatment


samples

[35,130]. Thus, ACV resistance seems more prevalent in the immuno­


1000

1191

competent population in case of herpetic keratitis, in particular in pa­


102
36

68
N◦

tients with recurrent disease and a history of ACV treatment.


resistant HSV
N◦ patients

5.4. General immunocompromised population


with ACV

3f (4.4 %)
1 (4.2 %)

1 (1.4 %)
0 (0 %)

0 (0 %)

0 (0 %)

In immunocompromised patients, infections with resistant HSV


(%)

strains can be associated with persistent and disseminated disease and


are potentially life-threatening [82]. It is therefore essential to test
the 3 isolates had a known drug-resistance genotype (G insertion in homopolymer stretch at positions 430–436 in the HSV-1 TK).
patients

clinically unresponsive infections in immunocompromised patients for


tested

ACV resistance to rationally adapt antiviral therapy. The prevalence of


233

102
27

24

68
N◦

ACVr HSV infections in the general immunocompromised population


ranged from 0 % to 28.8 % (Table 3), which was higher than that found
asymptomatic)

in the immunocompetent population. The study from Carrega et al. was


N◦ patients

1191 (702

the only study reporting no ACV resistance in immunocompromised


individuals, but tested only 10 patients, all children with cancer [136].
1000

102
36

68

The 11 other studies all reported a prevalence of ACV resistance above 3


%.
HSV-1 &

HSV-1 &

HSV-1 &

HSV-1 &

The immunocompromised population is a highly diverse group as


species

HSV-2

HSV-1

HSV-2

HSV-1

HSV-2

HSV-2

several conditions can lie at the base of immunosuppression, including


HSV

HIV infection, primary immunodeficiency, chemotherapy, and immu­


nosuppressive therapy following transplantation, inflammation or im­
mune diseases [71,111,118]. Patients with profound
Unspecified &
Untreated &
ACV treated

ACV treated
Unspecified

Unspecified

Unspecified

Unspecified
Treatment

immunosuppression, in particular those with suppressed cell-mediated


immunity, may have prolonged viral replication associated with more
status

extensive lesions and often require lengthy or multiple courses of ACV


treatment. Accordingly, severely immunocompromised individuals are
more likely to carry ACVr HSV than moderately immunosuppressed in­
Oral mucositis or skin

Oral HSV infection in


(asymptomatic) Oral

dividuals. Hence, the prevalence data presented in each study is largely


herpetic infections
Different herpetic

Orofacial lesionsb
(A)symptomatic
genital herpetic

defined by the underlying morbidities of the included patients. Evalu­


HSV disease

ating the prevalence of ACVr HSV infections in patients grouped ac­


infectionsb

infections
children

cording to the pathology causing immunosuppression is therefore more


lesion

informative.

5.5. Solid organ transplant recipients


Netherlands

Thailand
Country

Japan

Herpesviruses are the most common viral pathogens causing disease


China
Brazil

Iran
The

after solid organ transplantation. Infections with HSV occur most


commonly in the first month after transplantation and ACV prophylaxis
for at least 1 month after solid organ transplantation is therefore rec­
Publication

ommended [137,138]. The prevalence of ACVr HSV infections in the


1994

2012

2011

1989

2007

seven studies reporting on solid organ transplant recipients ranged from


year
Table 1 (continued )

0 % to 10 %, with only 1 study reporting a prevalence above 3.5 %


(Table 4). The highest prevalence was seen in patients receiving heart
Tateishi [124]

Varella [125]
First author

and/or lung transplantations [111,116].


Wang [108]
Stránskád

Ziyaeyan
Yoosook
[113]

[126]

[127]

7
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

5.6. Hematopoietic stem cell transplant recipients

ACV: acyclovir, EC50: effective concentration inhibiting 50% of virus growth in cell culture, HSV: herpes simplex virus, PCR: polymerase chain reaction, a study included 5 immunocompetent and 7 immunocompromised
Dye uptake assay (˃10x mean

Plaque reduction assay (EC50


˃2µM), Sanger sequencing of
Real time PCR (EC50 ˃1µM),
Method (cutoff resistance)

Plating efficiency, Real time


Sanger sequencing of UL23
CPE reduction assay (≥5x
Nowadays, most HSCT recipients receive prophylactic ACV and this

EC50 of sensitive isolates)


has considerably reduced the number of patients suffering from HSV

EC50 control strains)


infections posttransplantation [12,139]. The use of ACV prophylaxis

PCR (EC50 ˃1µM)


Dye uptake assay
reduces the risk of resistance development in comparison to acute
treatment, as less viral replication is allowed.
In the study from Wade et al. [140], patients received acute ACV

UL23
therapy and ACV resistance was reported in three out of 52 patients (5.8
%). High levels of ACV resistance were reported in HSCT recipients
receiving prophylaxis (Table 5). However, prevalence data in most
HSV isolates
N◦ resistant

studies was based on the number of patients tested (i.e., patients with
HSV infections while receiving prophylaxis), and this might over­
11

44
estimate the prevalence of HSV resistance in this population (range from
1

0
0 % to 100 %). When assessing ACV resistance in all patients receiving
HSCTs, lower but clinically significant rates of ACVr HSV infections were
positive
isolates

observed, generally within the 0–10 % range (Table 5, column preva­


N◦ HSV

lence of ACV resistant HSV) [141–143]. This shows that a substantial


173

169
40

35

15
6

portion of all patients receiving stem cell transplants, irrespective of


their HSV serostatus, experience ACVr breakthrough infections.
samples

Two studies reported high levels of ACV resistance in HSV-1 in­


173

169

fections while no ACVr HSV-2 infections were detected among the HSCT
40

35

15
N◦

recipients [111,144]. Additionally, the origin of the transplant (alloge­


neic or autologous donor) might impact the emergence of ACV resis­
resistant HSV

tance. Although HSV excretion is detected at equal rates after


N◦ patients

27 (34.6 %)
11 (6.4 %)
with ACV

1 (2.9 %)

autologous (10 %) and allogeneic (9.7 %) transplantation, ACVr HSV


0 (0 %)

0 (0 %)

0 (0 %)

infections seem to occur more frequently in allogeneic transplant re­


(%)

cipients [111,139]. Suboptimal dosing of ACV prophylaxis might also


dispose for resistance development. The study from Langston et al. [145]
demonstrated HSV reactivation in all 7 HSV seropositive patients
patients
Characteristics of studies reporting on acyclovir resistance in herpes simplex virus infections at immune-privileged sites.

tested

receiving ACV prophylaxis (5 mg/kg intravenous every 8 h), of whom 5


173
35

35

12

78

patients developed ACVr infections. Increasing the dose of prophylaxis


N◦

to 10 mg/kg was associated with a reduction in HSV reactivation and


patients

ACV resistance.
A rise in the frequency of ACVr HSV infections among HSCT re­
173

12a
35

35

78
N◦

cipients was described by several studies [73,144]. This might be


explained by an increased number of HLA-mismatched donors or
HSV-2 (1)

HSV-2 (8)
HSV-1 &

HSV-1 &
species

improved protocols for stem cell transplantation and induction chemo­


HSV-1

HSV-1

HSV-1

HSV-1
HSV

therapy regimens that induce longer and deeper immunosuppression.

5.7. HIV infected individuals


ACV treated & history

ACV treated & history


Unknown, untreated,

of ACV treatment (n
Treatment status

treatment (n = 8)
of ACV treatment
Untreated & ACV

or history of ACV

Untreated & ACV


treated (n = 10)
treated (n = 5)

HSV-2 infected patients have an increased risk of HIV acquisition


Unspecified

[146]. When left untreated, HIV infected individuals can show a marked
reduction of their CD4+ T cell count and become severely immunosup­
= 19)

pressed. This advanced state of HIV infection, often referred to as ac­


quired immunodeficiency syndrome (AIDS), is associated with
opportunistic infections and malignancies [147]. Low CD4+ T cell
Herpes simplex
Ocular herpes
HSV disease

Herpetic eye

encephalitis

counts seem to correlate with a higher risk of symptomatic HSV in­


Recurrent
infection
Herpetic

herpetic
keratitis

keratitis

fections [148]. The implementation of combination antiretroviral


lesions
Ocular

treatment (tri-therapy) resulted in an increase in CD4+ T cell count and


in low HIV viral loads associated with less opportunistic infections
[149]. However, HSV-2 is shed more frequently in HIV infected in­
Netherlands

Netherlands

Netherlands

dividuals, even when receiving antiretroviral therapy and when CD4+ T


Kingdom

Kingdom

Kingdom
Country

cell counts are high (>400cells/mm3) [150,151]. Acyclovir resistance


United

United

United
The

The

The

was detected at a marked higher frequency in HSV-2 infections than in


HSV-1 infections by Danve-Szatanek et al. (7.3 % versus 0 %), Reyes
et al. (5.3 % versus 0 %), and Ziyaeyan et al. (25 % versus 4.3 %)
Publication

[111,121,127] (Table 6). The reported prevalence of ACVr HSV in­


fections in HIV infected individuals ranged from 0 to 25 %, with most
1990

2008

1989

1990

2016

2013
year

studies reporting a prevalence between 3.4 % and 7.3 %.


First author

5.8. Prevalence of ACV resistance mutations


Duan [130]

Mitterreiter

Van Velzen
Gray [118]
[131]

[107]

[132]
Menage

patients.
Charles
Table 2

[35]

Nine of the included studies performed genotyping of the UL23 and/


or UL30 genes to characterize ACV resistance

8
H.H. Schalkwijk et al.
Table 3
Characteristics of studies reporting on acyclovir resistance in herpes simplex virus among the general immunocompromised population.
First author Publication Country HSV disease Treatment HSV N◦ N◦ N◦ patients N◦ N◦ HSV N◦ resistant Method (cutoff resistance)
year status species patients patients with ACV samples positive HSV isolates
tested resistant HSV isolates
(%)

Carrega [136] 1994 Italy Oral mucositis in ACV treated HSV-1 & 20 10 0 (0 %) 20 10 0 Plaque reduction assay
children with HSV-2 (2)
cancer
Christophers 1998 United Different herpetic ACV treated Untyped 95 95 6 (6.3 %) 146 146 6 Plaque reduction assay (EC50
[116] Kingdom infectionsa ≥ 3 µM)
Collins [109] 1993 Europe Unspecifieda ACV treated Untyped 81 81 18 (22.2 %) 206 206 64 Plaque reduction assay (EC50
≥ 3 µM)
United 208 208 60 (28.8 %) 582 582 105 Dye uptake assay (EC50 ≥ 3
States µg/mL)
Danve- 2004 France Different herpetic Untreated & ACV HSV-1 1168 1168 36 (3.1 %) 1523 1523 -b Dye uptake assay (EC50 ≥ 6.5
Szatanek infectionsa treated µM)
[111] HSV-2 334 334 18 (5.4 %) 411 411 - Dye uptake assay (EC50 ≥
13.5 µM)
Englund [71] 1990 United Herpetic infections Untreated & ACV Untyped 148 148 7 (4.7 %) - 218 23 DNA hybridization (EC50 ≥ 9
States at oral site & treated (1 2 8) µM)
genital tract
Frobert [73] 2014 France Different herpetic Unspecified HSV-1 & 360 360 35 (9.7 %) 360 360 35 Dye uptake assay (EC50 ≥ 6.5
infections in adultsa HSV-2 µM HSV-1, ≥13.5 µM HSV-2),
9

Different herpetic - - - 141 141 7 Sanger sequencing of UL23 &


infections in UL30
childrena
Gray [118] 1989 United Herpetic infections Untreated & ACV HSV-1 49 49 2 (4.1 %) - 101 2 Dye uptake assay
Kingdom of skin or mucous treated
Labrunie 2019 France Different herpetic Unknown, HSV-1 239 200 36 (18 %) 239 200 36 Dye uptake assay (EC50 ≥ 6.5
[218] infections in adults untreated & ACV µM)
& childrena treated (46) 239 35 (14.6 %) 239 UL23 35 Sanger sequencing of UL23 &
148 UL30 UL30
Nugier [112] 1992 France Different herpetic Untreated & ACV HSV-1 156 156 6 (3.8 %) 186 186 13 Dye uptake assay (EC50 ≥ 1.5
infectionsa treated µg/mL)
HSV-2 28 28 4 (14.3 %) 37 37 4 Dye uptake assay (EC50 ≥ 3
µg/mL)
Rabella [219] 2002 Spain Different herpetic Untreated, HSV-1 165 165 8 (4.8 %) 204 131 1 CPE reduction assay (EC50
infectionsa unknown & ACV HSV-2 73 8 ˃3µg/mL)

Biochemical Pharmacology 206 (2022) 115322


treated
Schmidt [123] 2015 Germany Different herpetic Untreated & ACV HSV-1 24 24 6 (25 %) 24 24 6 Sanger sequencing of UL23 &
infectionsa treated UL30 Plaque reduction assay
(≥5x EC50 control strain)
Stránská [113] 2005 The Different herpetic Untreated & ACV HSV-1 & 128 128 9 (7.0 %) 153 153 12 ELVIRA (≥3x EC50 control
Netherlands infectionsa treated HSV-2 strain), Sanger Sequencing of
UL23

ACV: acyclovir, EC50: effective concentration inhibiting 50% of virus growth in cell culture, HSV: herpes simplex virus. a may include infections at immune-privileged sites, b
data not available.
H.H. Schalkwijk et al.
Table 4
Characteristics of studies reporting on acyclovir resistance in herpes simplex virus among solid organ transplant recipients.
First author Publication Country HSV disease Treatment HSV N◦ N◦ N◦ patients N◦ N◦ HSV N◦ resistant Method (cutoff resistance)
year status species patients patients with ACV samples positive isolates
tested resistant HSV isolates
(%)

Boivin [220] 1993 United Different herpetic ACV or GCV HSV-1 & 14 14 0 (0 %) 18 18 0 DNA hybridization assay (EC50
States infectionsa treated HSV-2 ≥ 9 µM)
b c
Christophers 1998 United Different herpetic ACV treated untyped 20 20 2 (10 %) - - 2 Plaque reduction assay (EC50 ≥
[116] Kingdom infectionsa 3 µM)
Danve- 2004 France Different herpetic Untreated & HSV-1 168 168 4b (2.4 %) - - - Dye uptake assay (EC50 ≥ 6.5
Szatanek infectionsa ACV treated µM)
[111] HSV-2 33 33 1b (3.0 %) - - - Dye uptake assay (EC50 ≥ 13.5
µM)
Englund [71] 1990 United Herpetic Untreated (4) & untyped 58 58 2 (3.4 %) - - - DNA hybridization (EC50 ≥ 9
States infections at oral ACV treated µM)
10

site & genital (54)


tract
Frobert [73] 2014 France Different herpetic Unspecified HSV-1 & – – – 94 94 1 Dye uptake assay (EC50 ≥ 6.5
infections in HSV-2 µM HSV-1, ≥13.5 µM HSV-2) &
adultsa Sanger sequencing of UL23 &
Different herpetic – – – 38 38 1 UL30
infections in
childrena
Gray [118] 1989 United Herpetic Unspecified HSV-1 10 10 0 (0 %) 17 17 0 Dye uptake assay
Kingdom infections of skin
or mucous
Stránská [113] 2005 The Different herpetic Untreated & HSV-1 & 17 17 0 (0 %) 17 17 0 ELVIRA (≥3x EC50 control
Netherlands infectionsa ACV treated HSV-2 strain), Sanger sequencing of
UL23

ACV: acyclovir, EC50: effective concentration inhibiting 50% of virus growth in cell culture, GCV: ganciclovir, HSV: herpes simplex virus. a may include infections at immune-privileged sites, b heart or lung transplant

Biochemical Pharmacology 206 (2022) 115322


recipients, c data not available.
H.H. Schalkwijk et al.
Table 5
Characteristics of studies reporting on acyclovir resistance in herpes simplex virus among hematopoietic stem cell transplant recipients.
First author Publication Country HSV disease Treatment HSV N◦ N◦ N◦ patients Prevalence N◦ N◦ HSV N◦ Method (cutoff
year status species patients patients with ACV ACV resistant samples positive resistant resistance)
tested resistant HSVa isolates HSV
HSV (%) isolates

Anton- 2020 United Suspected HSV ACV treated HSV-1 532b 48 8 (16.7 %) 1.5 % -c – – Plaque reduction assay
Vazquez Kingdom infection HSV-2 18 0 (0 %) 0% – – – (EC50 ˃40 µM), Sanger
[144] sequencing of UL23 &
UL30
Ariza Heredia 2018 United Orolabial or ACV treated HSV-1 4028b – 12 0.30 % – – – Plaque reduction assay
[141] States anogenital HSV HSV-2 – 6 0.15 % – – –
infection
Chakrabarti 2000 United Infection of mouth, ACV treated HSV-1 75b 16 4 (25 %) 5.3 % – – – Plaque reduction assay
[221] Kingdom eye, skin, or genitals (EC50 ≥ 1.62 µM)
b
Chen [142] 2000 France Different herpetic ACV treated HSV-1 196 – 14 7.1 % – – – Dye uptake assay (EC50
infectionsd ≥ 6.5 µM)
Danve- 2004 France Different herpetic Untreated & HSV-1 182 182 22 (12.1 %) 12.1 % – – – Dye uptake assay (EC50
Szatanek infectionsd ACV treated ≥ 6.5 µM)
[111] HSV-2 19 19 0 (0 %) 0% – – – Dye uptake assay (EC50
≥ 13.5 µM)
Englund [71] 1990 United Herpetic infections ACV treated Untyped 29 29 4 (13.8 %) 13.8 % – – – DNA hybridization
States at oral site & genital assay (EC50 ≥ 9 µM)
tract
Frobert [73] 2014 France Different herpetic Unspecified HSV-1 & – – – – 90 90 30 Dye uptake assay (EC50
infections in adultsd HSV-2 ≥ 6.5 µM HSV-1, ≥13.5
Different herpetic – – – – 30 30 5 µM HSV-2), Sanger
infections in sequencing of UL23 &
childrend UL30
11

Heidenreich 2020 Germany Different herpetic ACV treated HSV-1 214b -e 9 4.2 % – – – Sanger sequencing,
[143] infectionsd plaque reduction assay
Kakiuchi 2017 Japan (asymptomatic) ACV treated HSV-1 251b 39 11 (28.2 %) 4.4 % 2747 103 35 Plaque reduction assay
[152] oropharyngeal (EC50 > 2 µg/mL),
infection Sanger sequencing of
UL23 & UL30
Langston 2002 United Infection at mucosa, ACV treated HSV 14b 5e 5 (100 %) 35.7 % – – – Plaque reduction assay
[145] States esophagus &
anogenital area
Morfin [139] 2004 France (asymptomatic) Untreated & HSV-1 283b 28 3 (10.7 %) 1.1 % – – – Dye uptake assay (EC50
HSV mucositis ACV treated ≥ 6.5 µM)
Nichols [222] 2003 United Different herpetic ACV treated HSV-1 436b – 14 3.2 % – – – DNA hybridization
States infectionsd HSV-2 – 1 0.23 % – – – assay (EC50 > 4 µg/mL)
Schmidt 2015 Germany Different herpetic Untreated & HSV-1 13 13 6 (46.2 %) 46.2 % 13 13 6 Sanger sequencing of
[123] infectionsd ACV treated UL23 & UL30, Plaque

Biochemical Pharmacology 206 (2022) 115322


reduction assay (≥5x
EC50 control strain)
Stránská 2005 The Different herpetic Untreated & HSV-1 & 28 28 4 (14.3 %) 14.3 % 34 34 5 ELVIRA (≥3x EC50
[113] Netherlands infectionsd ACV treated HSV-2 control strain), Sanger
sequencing of UL23
Van der Beek 2012 The Herpetic oral Untreated & HSV-1 49b 12e 5 (41.7 %) 10.2 % – 191 – Sanger sequencing of
[153] Netherlands ulcerations ACV treated UL23 & UL30, DNA
reduction assay
Wade [140] 1983 United Oropharyngeal & ACV treated HSV-1 & 52 52 3 (5.8 %) 5.8 % – – – Dye uptake assay
States genital herpetic HSV-2
infections

ACV: acyclovir, EC50: effective concentration inhibiting 50% of virus growth in cell culture, HSV: herpes simplex virus. a prevalence was calculated based on the total number of patients included in the study, b
total
number of transplanted patients, not all patients experienced HSV infections c data not available, d may include infections at immune-privileged sites, e only patients refractory to therapy were tested.
H.H. Schalkwijk et al.
Table 6
Characteristics of studies reporting on acyclovir resistance in herpes simplex virus among HIV infected individuals.
First author Publication Country HSV disease Treatment HSV N◦ N◦ N◦ patients N◦ N◦ HSV N◦ Method (cutoff resistance)
year status species patients patients infected with samples positive resistant
tested ACV resistant isolates isolates
HSV (%)

Christophers 1998 United Different herpetic ACV treated Untyped 29b 29 1 (3.4 %) -c – 1 Plaque reduction assay (EC50
[116] Kingdom infectionsa ≥ 3 µM)
Danve- 2004 France Different herpetic Untreated & HSV-1 133 133 0 (0 %) – – – Dye uptake assay (EC50 ≥ 6.5
Szatanek infectionsa ACV treated µM)
[111] HSV-2 177 177 13 (7.3 %) – – – Dye uptake assay (EC50 ≥ 13.5
µM)
Englund [71] 1990 United Herpetic infections Untreated (5) & untyped 14d 14 1 (7.1 %) – – – DNA hybridization (EC50 ≥ 9
States at oral site & genital ACV treated (9) µM)
tract
Frobert [73] 2014 France Herpetic infections Unspecified HSV-1 & – – – 37 37 3 Dye uptake assay (EC50 ≥ 6.5
at oral site, genital HSV-2 µM HSV-1, ≥13.5 µM HSV-2)
tract in adults & Sanger sequencing of UL23
12

& UL30
Muller [223] 2022 South Africa Genital ulcer ACV treated HSV-2 27 27 0 (0 %) 27 27 0 Sanger sequencing of UL23,
disease plaque reduction assay (EC50
≥ 2 µg/mL)
Nag [224] 2021 India Genital or oral Unspecified HSV-1 & 52 8 2 (25 %) 52 8 2 CPE reduction assay
herpetic lesions HSV-2
Reyes [121] 2003 United (Suspected) genital Untreated & HSV-1 – 12 0 (0 %) – 12 0 Plaque reduction assay (EC50
States herpes ACV treated ≥ 2 µg/mL)
HSV-2 – 226 12 (5.3 %) – 226 12 Plaque reduction assay (EC50
≥ 2 µg/mL)
Stránská [113] 2005 The Different herpetic Untreated & HSV-1 & 51 51 2 (3.9 %) 59 59 3 ELVIRA (≥3x EC50 control
Netherlands infectionsa ACV treated HSV-2 strain), Sanger sequencing of
UL23
Ziyaeyan 2007 Iran Orofacial lesionsa Unspecified HSV-1 23e 23 1 (4.3 %) 23 23 1 Plaque reduction assay (EC50
[127] HSV-2 8e 8 2 (25 %) 8 8 2 ≥ 2 µg/mL)

Biochemical Pharmacology 206 (2022) 115322


ACV: acyclovir, AIDS: acquired immunodeficiency syndrome, EC50: effective concentration inhibiting 50% of virus growth in cell culture, HSV: herpes simplex virus. a May include infections at immune-privileged sites, b
17 HIV infected individuals were diagnosed with AIDS, c data not available, d 12 patients with AIDS, 2 with AIDS-related complex, e all patients were diagnosed with AIDS.
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

[73,108,113,123,130,132,144,152,153]. We evaluated the prevalence


The occurrence of mutations conferring acyclovir resistance in articles included in this review. Novel mutations associated with acyclovir resistance are underlined and mutations conferring cross resistance to foscarnet are

of different mutations conferring ACV resistance among the 87 ACVr

[73,108,113,123,130,132,144,152,153]
HSV-1 infected patients and 9 ACVr HSV-2 infected patients included in
these nine studies. An overview of insertions, deletions, and amino acid
changes detected in these patients is available in Table 7.

[73,108,123,144,152,153]
In the samples derived from 87 patients with ACVr HSV-1 infections,
75 mutations conferring ACV resistance were reported in the UL23
Included studies

gene, of which the majority (54.7 %) were insertions or deletions


(Table 7). The G insertion in the homopolymer stretch at nucleotides
433–436 was reported most frequently (15 patients). The G200D mu­

[73,113]
tation was the most frequently reported point mutation (3 patients).

[73]
Mutations introducing a stop codon (E95stop, T172stop, R281stop, and
Q342 stop) were detected in samples from 5 patients. Samples from 68
548–553 (4), Ins C nts 548–553 (3), I194del (1), Ins T nt 613 (1), Del G nts 615–619 (1), Del nts 781–793 (1), Del C nt

E70K (1), G191S (1), L359I (1), L448V (1), E671K (1), G691S (1), A748T (1), G841A (1), E860K (1), A874T (1), A910T
430–436 (15), Del G nts 430–436 (1), Ins A nt 438 (1), Del G nt 438 (1), Del C nt 540 (1), Ins G nt 547 (1), Del C nts

G56D (1), G59W (1), I78S (1), E95stop (1), D162H (1), D162N (1), Y172C (1), T172stop (1), R176Q (1), L178R (2),
S181N (1), A189V (1), G200D (3), G200S (1), R216H (1), R220H (1), R222H (2), L249P (1), E257K (1), D258N (1),

patients were analyzed for UL30 mutations, and mutations conferring


E36del (1), Ins C nt 184 (1), Del C nt 184 (1), Ins C nt 185 (1), Del A nts 184–187 (1), Del C nt 269 (1), Ins G nts

ACV resistance were detected in samples from 10 patients (14.7 %). The
R261C (1), R281stop (2), T287M (1), L298P (1), C336Y (1), Q342stop (1), T354P (1), E371D (1), L327R (1)

majority of these mutations (i.e., A605V, R700G, S724N, I890M, and


G901V) conferred cross-resistance to foscarnet. Substitutions of unclear
significance were detected in both the UL23 and UL30 genes of HSV-1.
Q15K (1), E43A (2), A93V (1), A230V (1), L242P (1), P269S (1), S276N (1), I326V (1), A365T (1)

Six out of nine patients infected with an ACVr HSV-2 strain, harbored
mutations in the UL23 gene conferring ACV resistance, with an insertion
(single G in the G-homopolymer stretch at nucleotides 433–439) and a C
deletion (single C at nucleotides 551–556) reported in two patients each
[73,113]. No mutations conferring resistance were detected in the HSV-
2 UL30 gene, and several substitutions of unknown significance were
detected in both the UL23 and UL30 genes.
A605V (3), R700G (1), A719V (2), S724N (2), I890M (1), G901V (1)

The majority of ACV resistance mutations in the UL23 genes of HSV-


G36E (1), M129I (1), T202A (1), D229H (1), R268W (1), R363C (1)

1 and HSV-2 were frameshift mutations, in agreement with the litera­


ture (section 4.1). However, as only a limited number of HSV samples
were genotyped in the included studies, the prevalence of detected
885 (2), Del C nts 896–900 (1), Del C nts 1061–1064 (1)

mutations might not be representative for the total population of HSV


resistant strains. Comprehensive overviews of known HSV resistance
mutations are available in several reviews [74,82,86,87,95].
Ins C nts 433–439 (2), Del C nts 551–556 (2)

6. Antiviral alternatives for acyclovir resistant HSV infections


K458N (1), G506D (1), K592E (1)

Several antiviral treatment options can be considered in case of an


ACVr HSV infection. In some patients, an increased dose or intravenous
(1), T931A (1), P1114H (1)
Mutation (N◦ of patients)

administration of ACV is still effective to treat ACVr infections [28]. A


limited number of antivirals, all targeting the DNA pol, have been
G56E (1), T288M (1)

approved for the treatment of HSV infections. Although numerous an­


tivirals targeting different steps of HSV replication have been investi­
gated, many failed to show sufficient potency, bioavailability, or
acceptable toxicity. Several alternative antiviral compounds, either
approved or under investigation, will be discussed in the following

sections.
Insertion or

Insertion or

6.1. Nucleoside analogues


mutation

mutation

mutation

mutation

mutation

mutation

mutation

mutation
deletion

deletion
Type of

Point

Point

Point

Point

Point

Point

Point

In addition to ACV and VACV, other nucleoside analogues are


available for the treatment of HSV infections i.e., penciclovir or its


prodrug famciclovir, ganciclovir, brivudine, and trifluridine (tri­
fluorothymidine) (Fig. 4). Penciclovir and ganciclovir are guanosine
significance

significance

significance

significance
Associated
phenotype

analogues similar to ACV but are no obligate chain terminators due to


Resistance

Resistance

Resistance

Resistance
Unclear

Unclear

Unclear

Unclear

the presence of a 3′ hydroxyl on the acyclic sugar moiety. Incorporation


of penciclovir or ganciclovir into the viral DNA allows incorporation of
one additional nucleotide after which DNA elongation is terminated
[95]. Acyclovir and penciclovir differ in their affinity for the viral TK
N◦ resistant

and DNA pol but show a similar spectrum of activity [86]. Ganciclovir is
patients

most frequently used for the treatment of CMV infections but is


approved as topical treatment for herpetic keratitis. Brivudine is a
87

68

deoxyuridine analogue that acts as a competitive dTTP inhibitor and


only shows anti-HSV-1 and anti-VZV activities. Brivudine is dependent
marked in bold.
Gene

UL23

UL30

UL23

UL30

on viral TK activity for conversion to its monophosphate and diphos­


phate form, but the HSV-2 TK is unable to phosphorylate brivudine
Table 7

Virus

HSV-

HSV-

HSV-

HSV-

monophosphate due to its low thymidylate kinase activity [81,154].


1

Penciclovir, famciclovir, ganciclovir, and brivudine are not

13
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

Fig. 4. Chemical structure of several nucleoside analogues active against herpes simplex viruses.

recommended for the treatment of ACVr HSV infections. As these drugs brivudine (HSV-1 only) and ganciclovir. Penciclovir retains activity
have a similar mode of action requiring activation by the viral TK, cross- against several ACVr DNA pol mutants, including HSV-1 strains bearing
resistance between the nucleoside analogues is often observed, espe­ the A605V, A719T, or Y941H mutations, and HSV-2 strains harboring
cially when mutations arise in the viral TK [74,155,156]. Exceptionally, the A724V, S725G, I731F, Y823C or R964H mutations [74,158–161].
ACVr TK mutants, show no cross-resistance to other nucleoside ana­ The first activating phosphorylation step of the thymidine analogue
logues [134,157,158]. Cross-resistance is less frequently observed in trifluridine can be performed by both viral and cellular kinases. Tri­
ACVr DNA pol mutant strains, of which most remain sensitive to fluridine monophosphate irreversibly inhibits the conversion of

Fig. 5. Chemical structures of several antiviral compounds with anti-herpes simplex virus activity. The structures presented are acyclic nucleoside phosphonates
(cidofovir, brincidofovir, adefovir, and tenofovir), pyrophosphate analogue (foscarnet), helicase-primase inhibitors (pritelivir and amenamevir) and n-docosanol.

14
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

deoxyuridine monophosphate (dUMP) to dTMP by the cellular thymi­ some patients might experience treatment failure despite phenotypic
dylate synthase. In its triphosphate form, trifluridine can be incorpo­ susceptibility [142,185]. Thus, cidofovir can be considered to treat ACV
rated by both the cellular and viral DNA polymerase [162,163]. As a and/or foscarnet resistant HSV infections.
result, trifluridine exhibits low selectivity and high cytotoxicity and is Adefovir, an adenosine analogue highly effective against hepatitis B
therefore only available as topical treatment for herpetic keratitis [164]. virus, and tenofovir, an adenosine analogue that potently inhibits the
Mechanisms of trifluridine drug resistance are not well described but HIV and hepatitis B virus reverse transcriptase, show activity against
some studies reported a response to trifluridine treatment in select cases HSV [159,173]. Most isolates resistant to ACV are susceptible to ade­
of clinically ACVr HSV infections [165–167]. Thus, trifluridine therapy fovir and tenofovir although ACVr HSV strains with mutations in the
might be beneficial for some patients with ACVr HSV infections. DNA pol can show cross-resistance to adefovir and tenofovir
[46,159,186]. Furthermore, the activity of adefovir and tenofovir
6.2. Pyrophosphate analogues against HSV is too low to be used therapeutically. Tenofovir 1 % vaginal
microbicide gel has been evaluated for the prevention of HIV and HSV-2
The pyrophosphate analogue foscarnet (Fig. 5, phosphonoformic infections. Although tenofovir gel reduced HSV-2 acquisition in the
acid) selectively and reversibly binds to the pyrophosphate binding site CAPRISA 004 trial, the VOICE trial was unable to demonstrate an effect,
of the viral DNA pol. Binding of foscarnet prevents cleavage of the py­ which was associated with low adherence to antiviral treatment in this
rophosphate from the elongating viral DNA chain, inhibiting further population of patients [187–189].
nucleotide incorporation [168]. Therapy with foscarnet is complicated
due to its associated nephrotoxicity and poor oral bioavailability, only 6.4. Helicase-primase inhibitors
allowing intravenous or topical formulation [169]. Foscarnet is used in
immunocompromised patients for the treatment of mucocutaneous The helicase-primase complex, composed of the helicase, primase,
ACVr HSV infections due to changes in the viral TK but not for ACVr and cofactor subunits, is essential for viral replication and is targeted by
infections having alterations in the DNA pol, since cross-resistance with the helicase-primase inhibitors, a novel class of antivirals. These include
foscarnet occurs frequently [74,95,170]. Furthermore, HSV strains pritelivir (BAY 57–1293) and amenamevir (ASP2151), both showing
resistant to both ACV and foscarnet due to the acquisition of mutations potent anti HSV activity [190,191] (Fig. 5).
in both the TK and DNA pol have been described in the clinic [46,171]. The thiazolylamide pritelivir shows more potent HSV inhibition than
ACV in vitro and acts by inhibiting the single-stranded DNA-dependent
6.3. Acyclic nucleoside phosphonates ATPase activity of the helicase-primase complex [168,192]. Phase I and
II clinical trials in immunocompetent individuals showed effective HSV
Acyclic nucleoside phosphonates are DNA pol inhibitors that have a inhibition and no safety concerns [193–195]. A phase II/III trial
phosphonate group attached to the sugar moiety and therefore require assessing the efficacy and safety of pritelivir for the treatment of ACVr
only two activating phosphorylation steps, both executed by cellular mucocutaneous HSV infections is currently ongoing (NCT03073967).
kinases [50,172,173]. Because acyclic nucleoside phosphonates are in­ Amenamevir is an oxadiazolephenyl derivative that inhibits the
dependent of viral TK activity, they remain active against most ACVr helicase, primase, and single-stranded DNA-dependent ATPase activities
HSV strains. Three acyclic nucleoside phosphonates have been licensed of the helicase-primase complex [196]. In 2017, amenamevir was
for use in the clinic: cidofovir, adefovir, and tenofovir [50] (Fig. 5). approved in Japan for the treatment of herpes zoster caused by VZV
Cidofovir is a cytosine analogue that selectively inhibits herpesvirus reactivation [197]. It proved to be a safe and effective treatment for HSV
DNA pols and terminates viral DNA elongation in case of two successive infections in the immunocompetent population in phase I and II clinical
cidofovir incorporations [172,174,175]. Although cidofovir is only trials [198,199]. A phase III clinical trial (NCT01959295) evaluating the
approved for treatment of CMV retinitis in patients with AIDS, it is used efficacy and safety of amenamevir in patients with herpes simplex in­
off-label for the treatment of ACV and/or foscarnet resistant HSV in­ fections (labial/facial herpes or recurrent genital herpes) has been
fections. Due to its poor bioavailability, it is solely administered intra­ completed but results have not been published.
venously or as topical formulation [50]. Cidofovir treatment is Mutations conferring pritelivir and/or amenamevir resistance map
associated with side effects such as nephrotoxicity and impaired renal to the UL5 and UL52 genes encoding the helicase and primase, respec­
function [169]. Probenecid and intravenous hydration with saline prior tively, and are rapidly selected in vitro [200,201]. However, clinical
to cidofovir administration are indicated to reduce possible nephrotox­ trials did not reveal rapid emergence of resistance to helicase-primase
icity [176]. To improve oral bioavailability and reduce nephrotoxicity, inhibitors [193–195,199]. Amenamevir and pritelivir proved effective
efforts have been made to develop a prodrug. Brincidofovir (Fig. 5.), a against all ACVr HSV strains as these drugs have a different molecular
lipid conjugated prodrug of cidofovir, showed potent in vitro inhibition target than the DNA pol [190,191]. Three immunocompromised pa­
of herpesviruses, including HSV-1 and HSV-2 [177]. Although brinci­ tients with ACVr HSV infections were effectively treated with pritelivir
dofovir is not nephrotoxic, brincidofovir related gastrointestinal disor­ without the emergence of pritelivir resistance [202,203].
ders, misdiagnosed as acute graft-versus-host disease, were reported in
clinical trials evaluating the efficacy of brincidofovir against adenovirus 6.5. n-docosanol
and CMV infections in HSCT recipients and further trials have been
halted [178–180]. The 22-carbon-long saturated alcohol n-docosanol (Fig. 5) is avail­
Cidofovir was shown effective in the treatment of ACVr HSV in­ able over the counter as 10 % topical cream for labial herpes outbreaks.
fections and is active against TK mutants and most DNA pol mutant It appears that n-docosanol prevents HSV fusion and entry by interfering
strains [82,181,182]. Acyclovir resistant HSV strains with deficient or with the cell surface phospholipids [204,205]. N-docosanol reduces
altered TK activity are more susceptible to cidofovir than wild-type symptoms and healing time when therapy is started early during reac­
strains as they induce smaller increases in the dCTP pool, enhancing tivation and is active against ACVr HSV strains in vitro [205,206].
the inhibitory effect of cidofovir [183]. Resistance to cidofovir is ac­
quired by mutations in the DNA pol, and some mutations, including the 6.6. MBX-2168
S724N and L778M mutations in the HSV-1 DNA pol, show cross resis­
tance to ACV and/or foscarnet [98,184]. Most cidofovir resistance A newer class of nucleoside analogues includes the methyl­
associated mutations in the HSV DNA pol selected in vitro show no cross- enecyclopropane nucleoside analogues that possess potent activity
resistance to ACV or foscarnet [184]. Only one case of a cidofovir against various herpesviruses. The third-generation methyl­
resistant HSV infection has been reported in the clinic [185]. However, enecyclopropane nucleoside analogue MBX-2168 shows convincing

15
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

anti-HSV activity in vitro and acts as a competitive inhibitor of viral report in which heterogeneous virus populations of ACVr and wild-type
DNA pols [207]. MBX-2168 is monophosphorylated, converted to syn­ virus were detected, and a neonatal herpes infection with spatial
guanol monophosphate by adenosine deaminase-like protein 1 (ADAL- compartmentalization of wild-type and mutant virus [133,135]. Com­
1), which is subsequently phosphorylated to the active form synguanol bination therapy of ACV and vidarabine was effective in the treatment of
triphosphate [207] (Fig. 6). Synguanol only shows limited activity an HSV-1 infection with mixed populations of ACVr and wild-type
against HSV, likely due to inefficient phosphorylation of this compound strains [134]. Combination therapy with ganciclovir and cidofovir was
by HSV thymidine kinases. MBX-2168 phosphorylation is, at least in effective in treating a persistent HSV-1 infection that showed both viral
part, performed by the serine/threonine-protein kinase TAO3 (TAOK3) compartmentalization between lesions and mixed virus populations of
and MBX-2168 is therefore active against ACVr HSV with TK mutations (multi)-drug resistant strains [217]. These case reports show the po­
[207,208]. MBX-2168 showed low cytotoxicity in vitro, making it a tential of combination therapy and highlight that this strategy should be
promising antiviral for which further development is warranted [209]. considered for treatment of recalcitrant infections and/or for therapy of
infections caused by multiple strains presenting different drug suscep­
tibility patterns. Combinations with other antivirals, in particular of
6.7. Combination therapy compounds with different modes of action, might prove effective as well
in the management of ACVr HSV infections, although this requires
HSV infections can show heterogeneity of virus populations and further study.
spatial compartmentalization, with different viral strains infecting
distinct body sites [46,135]. When infected with multiple strains 7. Future perspectives and final remarks
showing dissimilar drug susceptibilities, combination therapy might be
necessary to effectively treat all variants. Combination therapy has the In this review we provided an overview of prevalence data on ACV
potential to prevent emergence of drug resistance and to treat resistant resistance in HSV infections. The studies reporting prevalence data had a
HSV infections. Although combination therapy is a common strategy for high variability in study design, methodology, treatment strategies,
the treatment of HIV and hepatitis C virus infections, it is only sporad­ treatment histories, immunosuppressive regimens, study population,
ically used for treatment of herpesvirus infections [210]. Previous and patient selection. This makes it difficult to define the global prev­
studies have shown additive or synergistic activity of several antiviral alence of ACV resistance in HSV infections. In the immunocompetent
combinations against HSV, including combinations of helicase primase population, the prevalence of ACV resistance in HSV infections did not
inhibitors and nucleoside analogues [162,211–213]. seem to be associated with ACV treatment history. However, in in­
Multiple case reports, have shown the effective use of foscarnet and fections at immune-privileged sites and in immunocompromised pa­
ACV combination therapy to treat HSV infections that were ACVr or tients, an impaired immune system allows frequent recurrent infections
refractory to ACV therapy [135,142,214–216]. These include a case

Fig. 6. Proposed mechanism of activation of MBX-2168. Initial phosphorylation is at least in part performed by the serine/threonine-protein kinase TAO3 (TAOK3).
MBX-2168 monophosphate (MBX-2168-MP) is then converted to synguanol monophosphate (synguanol-MP) by adenosine deaminase-like protein 1 (ADAL-1), which
is subsequently phosphorylated to its active form synguanol triphosphate (synguanol-TP) which competitively inhibits the viral DNA polymerase.

16
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

and ongoing viral replication, that require long-term ACV exposure, [9] K.J. Looker, A.S. Magaret, M.T. May, K.M. Turner, P. Vickerman, S.L. Gottlieb, L.
M. Newman, Global and Regional Estimates of Prevalent and Incident Herpes
predisposing for the development of ACVr HSV infections. The immu­
Simplex Virus Type 1 Infections in 2012, PLoS ONE 10 (10) (2015) e0140765.
nocompromised population is a highly diverse population, with [10] K.J. Looker, A.S. Magaret, K.M. Turner, P. Vickerman, S.L. Gottlieb, L.
different pathologies underlying immunosuppression. Distinguishing M. Newman, Global estimates of prevalent and incident herpes simplex virus type
between dissimilar subpopulations of immunocompromised patients, 2 infections in 2012, PLoS ONE 10 (1) (2015) e114989.
[11] C. Kaushic, K.L. Roth, V. Anipindi, F. Xiu, Increased prevalence of sexually
provides a better indication of the true prevalence of ACVr HSV in­ transmitted viral infections in women: the role of female sex hormones in
fections among different groups of immunosuppressed hosts. regulating susceptibility and immune responses, J. Reprod. Immunol. 88 (2)
Although ACV prophylaxis is effective in reducing HSV reactivation, (2011) 204–209.
[12] R.J. Whitley, B. Roizman, Herpes simplex virus infections, Lancet 357 (9267)
resistant infections remain a serious and frequent complication in (2001) 1513–1518.
herpetic keratitis and in immunocompromised individuals. Acyclovir [13] J.S. Pepose, T.L. Keadle, L.A. Morrison, Ocular herpes simplex: changing
should therefore be rationally used to prevent emergence of resistance. epidemiology, emerging disease patterns, and the potential of vaccine prevention
and therapy, Am. J. Ophthalmol. 141 (3) (2006) 547–557.
Furthermore, it is beneficial to test patients with unresponsive HSV in­ [14] C.M. Roberts, J.R. Pfister, S.J. Spear, Increasing proportion of herpes simplex
fections for antiviral resistance, as this can guide clinicians in the virus type 1 as a cause of genital herpes infection in college students, Sex. Transm.
adjustment of antiviral therapy. Ideally both phenotypical and geno­ Dis. 30 (10) (2003) 797–800.
[15] D. Durukan, C.K. Fairley, C.S. Bradshaw, T.R.H. Read, J. Druce, M. Catton,
typical characterization of HSV clinical samples should be performed to L. Caly, E.P.F. Chow, Increasing proportion of herpes simplex virus type 1 among
avoid false-positive phenotypical results and misinterpretation of novel women and men diagnosed with first-episode anogenital herpes: a retrospective
amino acid changes with unknown significance. observational study over 14 years in Melbourne, Australia, Sex Transm Infect 95
(4) (2019) 307–313.
The lack of antivirals with a different mechanism of action poses a
[16] F. Xu, M.R. Sternberg, B.J. Kottiri, G.M. McQuillan, F.K. Lee, A.J. Nahmias, S.
problem for the treatment of ACVr HSV infections, in particular when M. Berman, L.E. Markowitz, Trends in herpes simplex virus type 1 and type 2
resistance is caused by mutations in the DNA pol. Although several seroprevalence in the United States, JAMA 296 (8) (2006) 964–973.
promising antivirals are under development, there is still a need for [17] C.H. Mercer, C. Tanton, P. Prah, B. Erens, P. Sonnenberg, S. Clifton,
W. Macdowall, R. Lewis, N. Field, J. Datta, A.J. Copas, A. Phelps, K. Wellings, A.
novel antiviral treatments. Combination therapy has the potential to M. Johnson, Changes in sexual attitudes and lifestyles in Britain through the life
treat ACVr infections and should be further investigated. course and over time: findings from the National Surveys of Sexual Attitudes and
Lifestyles (Natsal), Lancet 382 (9907) (2013) 1781–1794.
[18] R. Gupta, T. Warren, A. Wald, Genital herpes, Lancet 370 (9605) (2007)
CRediT authorship contribution statement 2127–2137.
[19] J.K. Benedetti, J. Zeh, L. Corey, Clinical reactivation of genital herpes simplex
virus infection decreases in frequency over time, Ann. Intern. Med. 131 (1) (1999)
Hanna Helena Schalkwijk: Conceptualization, Methodology, Vali­ 14–20.
dation, Visualization, Writing – original draft, Writing – review & [20] W.E. Lafferty, R.W. Coombs, J. Benedetti, C. Critchlow, L. Corey, Recurrences
editing. Robert Snoeck: Conceptualization, Methodology, Supervision, after oral and genital herpes simplex virus infection, Influence of site of infection
and viral type, N Engl J Med 316 (23) (1987) 1444–1449.
Writing – review & editing, Funding acquisition. Graciela Andrei:
[21] R. Engelberg, D. Carrell, E. Krantz, L. Corey, A. Wald, Natural history of genital
Conceptualization, Methodology, Supervision, Validation, Writing – herpes simplex virus type 1 infection, Sex. Transm. Dis. 30 (2) (2003) 174–177.
review & editing. [22] J.W. Gnann Jr., R.J. Whitley, CLINICAL PRACTICE, Genital Herpes, N Engl J Med
375 (7) (2016) 666–674.
[23] S.H. James, J.S. Sheffield, D.W. Kimberlin, Mother-to-Child Transmission of
Herpes Simplex Virus, J Pediatric Infect Dis Soc 1 (Suppl 1) (3 Suppl 2014,)
Declaration of Competing Interest S19–S23.
[24] K.J. Looker, A.S. Magaret, M.T. May, K.M.E. Turner, P. Vickerman, L.M. Newman,
S.L. Gottlieb, First estimates of the global and regional incidence of neonatal
The authors declare that they have no known competing financial herpes infection, Lancet Glob Health 5 (3) (2017) e300–e309.
interests or personal relationships that could have appeared to influence [25] D.W. Kimberlin, Neonatal herpes simplex infection, Clin. Microbiol. Rev. 17 (1)
the work reported in this paper. (2004) 1–13.
[26] S.G. Pinninti, D.W. Kimberlin, Neonatal herpes simplex virus infections, Semin.
Perinatol. 42 (3) (2018) 168–175.
Acknowledgements [27] R.J. Whitley, A.J. Nahmias, S.J. Soong, G.G. Galasso, C.L. Fleming, C.A. Alford,
Vidarabine therapy of neonatal herpes simplex virus infection, Pediatrics 66 (4)
(1980) 495–501.
Financial support was provided by Research Foundation Flanders [28] R. Whitley, J. Baines, Clinical management of herpes simplex virus infections:
(FWO, grant number G08818N). The authors wish to thank Kristel past, present, and future F1000Res 7 (2018).
Paque, Thomas Vandendriessche, and Krizia Tuand, the biomedical [29] D.W. Kimberlin, Advances in the treatment of neonatal herpes simplex infections,
Rev. Med. Virol. 11 (3) (2001) 157–163.
reference librarians of the KU Leuven libraries, 2Bergen learning Centre [30] D.W. Kimberlin, C.Y. Lin, R.F. Jacobs, D.A. Powell, L. Corey, W.C. Gruber,
Désiré Collen (Leuven, Belgium), for their help in conducting the sys­ M. Rathore, J.S. Bradley, P.S. Diaz, M. Kumar, A.M. Arvin, K. Gutierrez,
tematic literature search. M. Shelton, L.B. Weiner, J.W. Sleasman, T.M. de Sierra, S. Weller, S.J. Soong,
J. Kiell, F.D. Lakeman, R.J. Whitley, Safety and efficacy of high-dose intravenous
acyclovir in the management of neonatal herpes simplex virus infections,
References Pediatrics 108 (2) (2001) 230–238.
[31] L. Remeijer, A. Osterhaus, G. Verjans, Human herpes simplex virus keratitis: the
pathogenesis revisited, Ocul Immunol Inflamm 12 (4) (2004) 255–285.
[1] J.P. Weir, Genomic organization and evolution of the human herpesviruses, Virus
[32] S. Burrel, D. Boutolleau, G. Azar, S. Doan, C. Deback, I. Cochereau, H. Agut, E.
Genes 16 (1) (1998) 85–93.
E. Gabison, Phenotypic and genotypic characterization of acyclovir-resistant
[2] A.J. Davison, Evolution of the herpesviruses, Vet. Microbiol. 86 (1–2) (2002)
corneal HSV-1 isolates from immunocompetent patients with recurrent herpetic
69–88.
keratitis, J. Clin. Virol. 58 (1) (2013) 321–324.
[3] A.J. Davison, R. Eberle, B. Ehlers, G.S. Hayward, D.J. McGeoch, A.C. Minson, P.
[33] I. McCormick, C. James, N.J. Welton, P. Mayaud, K.M.E. Turner, S.L. Gottlieb,
E. Pellett, B. Roizman, M.J. Studdert, E. Thiry, The order Herpesvirales, Arch.
A. Foster, K.J. Looker, Incidence of herpes simplex virus keratitis and other ocular
Virol. 154 (1) (2009) 171–177.
disease: global review and estimates, Ophthalmic Epidemiol. (2021) 1–10.
[4] R.S. Sellar, K.S. Peggs, Management of multidrug-resistant viruses in the
[34] A.V. Farooq, D. Shukla, Herpes simplex epithelial and stromal keratitis: an
immunocompromised host, Br. J. Haematol. 156 (5) (2012) 559–572.
epidemiologic update, Surv. Ophthalmol. 57 (5) (2012) 448–462.
[5] E. Cesarman, Gammaherpesvirus and lymphoproliferative disorders in
[35] M. van Velzen, D.A. van de Vijver, F.B. van Loenen, A.D. Osterhaus, L. Remeijer,
immunocompromised patients, Cancer Lett. 305 (2) (2011) 163–174.
G.M. Verjans, Acyclovir prophylaxis predisposes to antiviral-resistant recurrent
[6] J.I. Cohen, Herpesvirus latency, J Clin Invest 130 (7) (2020) 3361–3369.
herpetic keratitis, J. Infect. Dis. 208 (9) (2013) 1359–1365.
[7] S.H. James, D.W. Kimberlin, R.J. Whitley, Antiviral therapy for herpesvirus
[36] J.W. Gnann Jr., R.J. Whitley, Herpes Simplex Encephalitis: an Update, Curr Infect
central nervous system infections: neonatal herpes simplex virus infection, herpes
Dis Rep 19 (3) (2017) 13.
simplex encephalitis, and congenital cytomegalovirus infection, Antiviral Res. 83
[37] A. Hjalmarsson, P. Blomqvist, B. Sköldenberg, Herpes simplex encephalitis in
(3) (2009) 207–213.
Sweden, 1990–2001: incidence, morbidity, and mortality, Clin. Infect. Dis. 45 (7)
[8] C. James, M. Harfouche, N.J. Welton, K.M. Turner, L.J. Abu-Raddad, S.
(2007) 875–880.
L. Gottlieb, K.J. Looker, Herpes simplex virus: global infection prevalence and
incidence estimates, 2016, Bull. World Health Organ. 98 (5) (2020) 315–329.

17
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

[38] E. Puchhammer-Stöckl, E. Presterl, C. Croÿ, S. Aberle, T. Popow-Kraupp, [65] R. Gupta, A. Wald, E. Krantz, S. Selke, T. Warren, M. Vargas-Cortes, G. Miller,
M. Kundi, H. Hofmann, U. Wenninger, I. Gödl, Screening for possible failure of L. Corey, Valacyclovir and acyclovir for suppression of shedding of herpes
herpes simplex virus PCR in cerebrospinal fluid for the diagnosis of herpes simplex virus in the genital tract, J. Infect. Dis. 190 (8) (2004) 1374–1381.
simplex encephalitis, J. Med. Virol. 64 (4) (2001) 531–536. [66] K.H. Fife, J. Almekinder, S. Ofner, A comparison of one year of episodic or
[39] J. Granerod, H.E. Ambrose, N.W. Davies, J.P. Clewley, A.L. Walsh, D. Morgan, suppressive treatment of recurrent genital herpes with valacyclovir, Sex. Transm.
R. Cunningham, M. Zuckerman, K.J. Mutton, T. Solomon, K.N. Ward, M.P. Lunn, Dis. 34 (5) (2007) 297–301.
S.R. Irani, A. Vincent, D.W. Brown, N.S. Crowcroft, Causes of encephalitis and [67] R.A. Taplitz, E.B. Kennedy, E.J. Bow, J. Crews, C. Gleason, D.K. Hawley, A.
differences in their clinical presentations in England: a multicentre, population- A. Langston, L.J. Nastoupil, M. Rajotte, K.V. Rolston, L. Strasfeld, C.R. Flowers,
based prospective study, Lancet Infect. Dis. 10 (12) (2010) 835–844. Antimicrobial Prophylaxis for Adult Patients With Cancer-Related
[40] U. Sili, A. Kaya, A. Mert, Herpes simplex virus encephalitis: clinical Immunosuppression: ASCO and IDSA Clinical Practice Guideline Update, J. Clin.
manifestations, diagnosis and outcome in 106 adult patients, J. Clin. Virol. 60 (2) Oncol. 36 (30) (2018) 3043–3054.
(2014) 112–118. [68] L. Henze, C. Buhl, M. Sandherr, O.A. Cornely, W.J. Heinz, Y. Khodamoradi, T.
[41] J.P. Stahl, A. Mailles, T. De Broucker, Herpes simplex encephalitis and R. Kiderlen, P. Koehler, A. Seidler, R. Sprute, M. Schmidt-Hieber, M. von
management of acyclovir in encephalitis patients in France, Epidemiol. Infect. Lilienfeld-Toal, Management of herpesvirus reactivations in patients with solid
140 (2) (2012) 372–381. tumours and hematologic malignancies: update of the Guidelines of the Infectious
[42] T. Chew, K.E. Taylor, K.L. Mossman, Innate and adaptive immune responses to Diseases Working Party (AGIHO) of the German Society for Hematology and
herpes simplex virus, Viruses 1 (3) (2009) 979–1002. Medical Oncology (DGHO) on herpes simplex virus type 1, herpes simplex virus
[43] C.I. Bustamante, J.C. Wade, Herpes simplex virus infection in the type 2, and varicella zoster virus, Ann. Hematol. 101 (3) (2022) 491–511.
immunocompromised cancer patient, J. Clin. Oncol. 9 (10) (1991) 1903–1915. [69] M. Christopeit, M. Schmidt-Hieber, R. Sprute, D. Buchheidt, M. Hentrich, M.
[44] J.D. Meyers, N. Flournoy, E.D. Thomas, Infection with herpes simplex virus and Karthaus, O. Penack, M. Ruhnke, F. Weissinger, O.A. Cornely, G. Maschmeyer,
cell-mediated immunity after marrow transplant, J. Infect. Dis. 142 (3) (1980) Prophylaxis, diagnosis and therapy of infections in patients undergoing high-dose
338–346. chemotherapy and autologous haematopoietic stem cell transplantation. 2020
[45] R.F. Pass, R.J. Whitley, J.D. Whelchel, A.G. Diethelm, D.W. Reynolds, C.A. Alford, update of the recommendations of the Infectious Diseases Working Party
Identification of patients with increased risk of infection with herpes simplex (AGIHO) of the German Society of Hematology and Medical Oncology (DGHO),
virus after renal transplantation, J. Infect. Dis. 140 (4) (1979) 487–492. Ann Hematol 100(2) (2021) 321-336.
[46] G. Andrei, A. Georgala, D. Topalis, P. Fiten, M. Aoun, G. Opdenakker, R. Snoeck, [70] M. Ifversen, R. Meisel, P. Sedlacek, K. Kalwak, L. Sisinni, D. Hutt, T. Lehrnbecher,
Heterogeneity and evolution of thymidine kinase and DNA polymerase mutants of A. Balduzzi, T. Diesch, A. Jarisch, T. Güngör, J. Stein, I. Yaniv, H. Bonig, M.
herpes simplex virus type 1: implications for antiviral therapy, J. Infect. Dis. 207 Kuhlen, M. Ansari, T. Nava, J.H. Dalle, C. Diaz-de-Heredia, E. Trigoso, U.
(8) (2013) 1295–1305. Falkenberg, M. Hartmann, M. Deiana, M. Canesi, C. Broggi, A. Bertaina, B.
[47] P. Ljungman, M.N. Ellis, R.C. Hackman, D.H. Shepp, J.D. Meyers, Acyclovir- Gibson, G. Krivan, K. Vettenranta, T. Matic, J. Buechner, A. Lawitschka, C. Peters,
resistant herpes simplex virus causing pneumonia after marrow transplantation, A. Yesilipek, K. Yalçin, G. Lucchini, S. Bakhtiar, D. Turkiewicz, R. Niinimäki, J.
J. Infect. Dis. 162 (1) (1990) 244–248. Wachowiak, S. Cesaro, A. Dalissier, S. Corbacioglu, A.M. Willasch, P. Bader,
[48] G.B. Elion, P.A. Furman, J.A. Fyfe, P. de Miranda, L. Beauchamp, H.J. Schaeffer, Supportive Care During Pediatric Hematopoietic Stem Cell Transplantation:
Selectivity of action of an antiherpetic agent, 9-(2-hydroxyethoxymethyl) Prevention of Infections. A Report From Workshops on Supportive Care of the
guanine, Proc Natl Acad Sci U S A 74 (12) (1977) 5716–5720. Paediatric Diseases Working Party (PDWP) of the European Society for Blood and
[49] J.A. Fyfe, P.M. Keller, P.A. Furman, R.L. Miller, G.B. Elion, Thymidine kinase Marrow Transplantation (EBMT), Front Pediatr 9 (2021) 705179.
from herpes simplex virus phosphorylates the new antiviral compound, 9-(2- [71] J.A. Englund, M.E. Zimmerman, E.M. Swierkosz, J.L. Goodman, D.R. Scholl, H.
hydroxyethoxymethyl)guanine, J. Biol. Chem. 253 (24) (1978) 8721–8727. H. Balfour Jr., Herpes simplex virus resistant to acyclovir, A study in a tertiary
[50] E. De Clercq, J. Neyts, Antiviral agents acting as DNA or RNA chain terminators, care center, Ann Intern Med 112 (6) (1990) 416–422.
Handb. Exp. Pharmacol. 189 (2009) 53–84. [72] K.R. Beutner, Valacyclovir: a review of its antiviral activity, pharmacokinetic
[51] W.H. Miller, R.L. Miller, Phosphorylation of acyclovir (acycloguanosine) properties, and clinical efficacy, Antiviral Res. 28 (4) (1995) 281–290.
monophosphate by GMP kinase, J. Biol. Chem. 255 (15) (1980) 7204–7207. [73] E. Frobert, S. Burrel, S. Ducastelle-Lepretre, G. Billaud, F. Ader, J.S. Casalegno,
[52] W.H. Miller, R.L. Miller, Phosphorylation of acyclovir diphosphate by cellular V. Nave, D. Boutolleau, M. Michallet, B. Lina, F. Morfin, Resistance of herpes
enzymes, Biochem. Pharmacol. 31 (23) (1982) 3879–3884. simplex viruses to acyclovir: an update from a ten-year survey in France, Antiviral
[53] J.E. Reardon, T. Spector, Herpes simplex virus type 1 DNA polymerase, Res. 111 (2014) 36–41.
Mechanism of inhibition by acyclovir triphosphate, J Biol Chem 264 (13) (1989) [74] A. Sauerbrei, K. Bohn-Wippert, M. Kaspar, A. Krumbholz, M. Karrasch, R. Zell,
7405–7411. Database on natural polymorphisms and resistance-related non-synonymous
[54] D. Derse, Y.C. Cheng, P.A. Furman, M.H. St Clair, G.B. Elion, Inhibition of purified mutations in thymidine kinase and DNA polymerase genes of herpes simplex virus
human and herpes simplex virus-induced DNA polymerases by 9-(2- types 1 and 2, J. Antimicrob. Chemother. 71 (1) (2016) 6–16.
hydroxyethoxymethyl)guanine triphosphate. Effects on primer-template function, [75] N.K. Balasubramaniam, V. Veerisetty, G.A. Gentry, Herpesviral deoxythymidine
J. Biol. Chem. 256 (22) (1981) 11447–11451. kinases contain a site analogous to the phosphoryl-binding arginine-rich region of
[55] A.K. Vashishtha, R.D. Kuchta, Effects of Acyclovir, Foscarnet, and porcine adenylate kinase; comparison of secondary structure predictions and
Ribonucleotides on Herpes Simplex Virus-1 DNA Polymerase: Mechanistic conservation, J. Gen. Virol. 71 (Pt 12) (1990) 2979–2987.
Insights and a Novel Mechanism for Preventing Stable Incorporation of [76] J.S. Evans, K.P. Lock, B.A. Levine, J.N. Champness, M.R. Sanderson, W.
Ribonucleotides into DNA, Biochemistry 55 (7) (2016) 1168–1177. C. Summers, P.J. McLeish, A. Buchan, Herpesviral thymidine kinases: laxity and
[56] D.H. King, History, pharmacokinetics, and pharmacology of acyclovir, J. Am. resistance by design, J. Gen. Virol. 79 (Pt 9) (1998) 2083–2092.
Acad. Dermatol. 18 (1 Pt 2) (1988) 176–179. [77] A.T. Jamieson, G.A. Gentry, J.H. Subak-Sharpe, Induction of both thymidine and
[57] K.L. Seley-Radtke, M.K. Yates, The evolution of nucleoside analogue antivirals: A deoxycytidine kinase activity by herpes viruses, J. Gen. Virol. 24 (3) (1974)
review for chemists and non-chemists. Part 1: Early structural modifications to 465–480.
the nucleoside scaffold, Antiviral Res. 154 (2018) 66–86. [78] D.M. Coen, M. Kosz-Vnenchak, J.G. Jacobson, D.A. Leib, C.L. Bogard, P.
[58] P.A. Furman, M.H. St Clair, J.A. Fyfe, J.L. Rideout, P.M. Keller, G.B. Elion, A. Schaffer, K.L. Tyler, D.M. Knipe, Thymidine kinase-negative herpes simplex
Inhibition of herpes simplex virus-induced DNA polymerase activity and viral virus mutants establish latency in mouse trigeminal ganglia but do not reactivate,
DNA replication by 9-(2-hydroxyethoxymethyl)guanine and its triphosphate, Proc Natl Acad Sci U S A 86 (12) (1989) 4736–4740.
J. Virol. 32 (1) (1979) 72–77. [79] R.B. Tenser, K.A. Hay, W.A. Edris, Latency-associated transcript but not
[59] L.M. Beauchamp, G.F. Orr, P. De Miranda, T. Burnette, T.A. Krenitsky, Amino acid reactivatable virus is present in sensory ganglion neurons after inoculation of
ester prodrugs of acyclovir, Antiviral Chem. Chemother. 3 (3) (1992) 157–164. thymidine kinase-negative mutants of herpes simplex virus type 1, J. Virol. 63 (6)
[60] S. Weller, M.R. Blum, M. Doucette, T. Burnette, D.M. Cederberg, P. de Miranda, (1989) 2861–2865.
M.L. Smiley, Pharmacokinetics of the acyclovir pro-drug valaciclovir after [80] D. Deville-Bonne, C. El Amri, P. Meyer, Y. Chen, L.A. Agrofoglio, J. Janin, Human
escalating single- and multiple-dose administration to normal volunteers, Clin. and viral nucleoside/nucleotide kinases involved in antiviral drug activation:
Pharmacol. Ther. 54 (6) (1993) 595–605. structural and catalytic properties, Antiviral Res. 86 (1) (2010) 101–120.
[61] S.L. Spruance, R. Nett, T. Marbury, R. Wolff, J. Johnson, T. Spaulding, Acyclovir [81] J.A. Fyfe, Differential phosphorylation of (E)-5-(2-bromovinyl)-2’-deoxyuridine
cream for treatment of herpes simplex labialis: results of two randomized, double- monophosphate by thymidylate kinases from herpes simplex viruses types 1 and 2
blind, vehicle-controlled, multicenter clinical trials, Antimicrob. Agents and varicella zoster virus, Mol. Pharmacol. 21 (2) (1982) 432–437.
Chemother. 46 (7) (2002) 2238–2243. [82] C. Gilbert, J. Bestman-Smith, G. Boivin, Resistance of herpesviruses to antiviral
[62] D.E. Balderson, G. Cai, M.A. Fries, D.M. Kleinman, M.M. McLaughlin, T. drugs: clinical impacts and molecular mechanisms, Drug Resist Updat 5 (2)
M. Trivedi, J.I. Wurzelmann, S.B. Young, A systematic review and meta-analysis (2002) 88–114.
to compare the efficacy of acyclovir 3% ophthalmic ointment to idoxuridine in [83] A. Gaudreau, E. Hill, H.H. Balfour Jr., A. Erice, G. Boivin, Phenotypic and
curing herpetic keratitis by Day 7 of treatment, BMC Ophthalmol 15 (2015) 42. genotypic characterization of acyclovir-resistant herpes simplex viruses from
[63] J. Piret, G. Boivin, Resistance of herpes simplex viruses to nucleoside analogues: immunocompromised patients, J. Infect. Dis. 178 (2) (1998) 297–303.
mechanisms, prevalence, and management, Antimicrob. Agents Chemother. 55 [84] N. Malartre, R. Boulieu, N. Falah, J.C. Cortay, B. Lina, F. Morfin, E. Frobert,
(2) (2011) 459–472. Effects of mutations on herpes simplex virus 1 thymidine kinase functionality: an
[64] WHO Guidelines for the Treatment of Genital Herpes Simplex Virus, WHO in vitro assay based on detection of monophosphate forms of acyclovir and
Guidelines for the Treatment of Genital Herpes Simplex Virus, World Health thymidine using HPLC/DAD, Antiviral Res. 95 (3) (2012) 224–228.
Organization Copyright © World Health Organization 2016., Geneva, 2016. [85] E.L. Hill, G.A. Hunter, M.N. Ellis, In vitro and in vivo characterization of herpes
simplex virus clinical isolates recovered from patients infected with human

18
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

immunodeficiency virus, Antimicrob. Agents Chemother. 35 (11) (1991) [112] F. Nugier, J.N. Colin, M. Aymard, M. Langlois, Occurrence and characterization of
2322–2328. acyclovir-resistant herpes simplex virus isolates: Report on a two—year
[86] G. Andrei, R. Snoeck, Herpes simplex virus drug-resistance: new mutations and sensitivity screening survey, J. Med. Virol. 36 (1) (1992) 1–12.
insights, Curr Opin Infect Dis 26 (6) (2013) 551–560. [113] R. Stránská, R. Schuurman, E. Nienhuis, I.W. Goedegebuure, M. Polman, J.
[87] F. Morfin, D. Thouvenot, Herpes simplex virus resistance to antiviral drugs, F. Weel, P.M. Wertheim-Van Dillen, R.J. Berkhout, A.M. van Loon, Survey of
J. Clin. Virol. 26 (1) (2003) 29–37. acyclovir-resistant herpes simplex virus in the Netherlands: prevalence and
[88] F. Morfin, G. Souillet, K. Bilger, T. Ooka, M. Aymard, D. Thouvenot, Genetic characterization, J. Clin. Virol. 32 (1) (2005) 7–18.
characterization of thymidine kinase from acyclovir-resistant and -susceptible [114] T.H. Bacon, R.J. Boon, M. Schultz, C. Hodges-Savola, Surveillance for antiviral-
herpes simplex virus type 1 isolated from bone marrow transplant recipients, agent-resistant herpes simplex virus in the general population with recurrent
J. Infect. Dis. 182 (1) (2000) 290–293. herpes labialis, Antimicrob. Agents Chemother. 46 (9) (2002) 3042–3044.
[89] S. Burrel, C. Aime, L. Hermet, Z. Ait-Arkoub, H. Agut, D. Boutolleau, Surveillance [115] R.J. Boon, T.H. Bacon, H.L. Robey, T.J. Coleman, A. Connolly, P. Crosson, S.
of herpes simplex virus resistance to antivirals: a 4-year survey, Antiviral Res. 100 L. Sacks, Antiviral susceptibilities of herpes simplex virus from immunocompetent
(2) (2013) 365–372. subjects with recurrent herpes labialis: a UK-based survey, J. Antimicrob.
[90] P.E. Boehmer, I.R. Lehman, Herpes simplex virus DNA replication, Annu. Rev. Chemother. 46 (6) (2000) 1051.
Biochem. 66 (1997) 347–384. [116] J. Christophers, J. Clayton, J. Craske, R. Ward, P. Collins, M. Trowbridge,
[91] S. Liu, J.D. Knafels, J.S. Chang, G.A. Waszak, E.T. Baldwin, M.R. Deibel Jr., D. G. Darby, Survey of resistance of herpes simplex virus to acyclovir in northwest
R. Thomsen, F.L. Homa, P.A. Wells, M.C. Tory, R.A. Poorman, H. Gao, X. Qiu, A. England, Antimicrob. Agents Chemother. 42 (4) (1998) 868–872.
P. Seddon, Crystal structure of the herpes simplex virus 1 DNA polymerase, [117] K.H. Fife, C.S. Crumpacker, G.J. Mertz, E.L. Hill, G.S. Boone, Recurrence and
J. Biol. Chem. 281 (26) (2006) 18193–18200. resistance patterns of herpes simplex virus following cessation of ≥6 years of
[92] S.L. Terrell, D.M. Coen, The pre-NH(2)-terminal domain of the herpes simplex chronic suppression with acyclovir, J. Infect. Dis. 169 (6) (1994) 1338–1341.
virus 1 DNA polymerase catalytic subunit is required for efficient viral [118] J.J. Gray, T.G. Wreghitt, T.P. Baglin, Susceptibility to acyclovir of herpes simplex
replication, J. Virol. 86 (20) (2012) 11057–11065. virus: emergence of resistance in patients with lymphoid and myeloid neoplasia,
[93] S.L. Terrell, J.M. Pesola, D.M. Coen, Roles of conserved residues within the pre- J. Infect. 19 (1) (1989) 31–40.
NH2-terminal domain of herpes simplex virus 1 DNA polymerase in replication [119] T. Hasegawa, T. Kawana, T. Okuda, M. Horii, T. Tsukada, K. Shiraki,
and latency in mice, J. Gen. Virol. 95 (Pt 4) (2014) 940–947. Susceptibility to acyclovir of herpes simplex virus isolates obtained between 1977
[94] Y. Narita, A. Sugimoto, D. Kawashima, T. Watanabe, T. Kanda, H. Kimura, and 1996 in Japan, J. Med. Virol. 63 (1) (2001) 57–63.
T. Tsurumi, T. Murata, A Herpesvirus Specific Motif of Epstein-Barr Virus DNA [120] M. Hashido, T. Kawana, In vitro acyclovir sensitivity of herpes simplex viruses
Polymerase Is Required for the Efficient Lytic Genome Synthesis, Sci. Rep. 5 isolated from female genital herpes in Japan, Kansenshogaku zasshi, The Journal
(2015) 11767. of the Japanese Association for Infectious Diseases 62 (2) (1988) 141–146.
[95] D. Topalis, S. Gillemot, R. Snoeck, G. Andrei, Distribution and effects of amino [121] M. Reyes, N.S. Shaik, J.M. Graber, R. Nisenbaum, N.T. Wetherall, K. Fukuda, W.
acid changes in drug-resistant α and β herpesviruses DNA polymerase, Nucleic C. Reeves, V., Task Force Herpes Simplex, Acyclovir-resistant genital herpes
Acids Res. 44 (20) (2016) 9530–9554. among persons attending sexually transmitted disease and human
[96] K. Zarrouk, J. Piret, G. Boivin, Herpesvirus DNA polymerases: Structures, immunodeficiency virus clinics, Arch. Intern. Med. 163 (1) (2003) 76–80.
functions and inhibitors, Virus Res. 234 (2017) 177–192. [122] A. Sangdara, P. Bhattarakosol, Acyclovir susceptibility of herpes simplex virus
[97] C.V. Kuny, M.L. Szpara, Alphaherpesvirus Genomics: Past, Present and Future, isolates at King Chulalongkorn Memorial Hospital, Bangkok, Journal of the
Curr Issues Mol Biol 42 (2021) 41–80. Medical Association of Thailand 91 (6) (2008) 908–912.
[98] J. Bestman-Smith, G. Boivin, Drug resistance patterns of recombinant herpes [123] S. Schmidt, K. Bohn-Wippert, P. Schlattmann, R. Zell, A. Sauerbrei, Sequence
simplex virus DNA polymerase mutants generated with a set of overlapping Analysis of Herpes Simplex Virus 1 Thymidine Kinase and DNA Polymerase Genes
cosmids and plasmids, J. Virol. 77 (14) (2003) 7820–7829. from over 300 Clinical Isolates from 1973 to 2014 Finds Novel Mutations That
[99] M. Mercier-Darty, D. Boutolleau, C. Rodriguez, S. Burrel, Added value of ultra- May Be Relevant for Development of Antiviral Resistance, Antimicrob. Agents
deep sequencing (UDS) approach for detection of genotypic antiviral resistance of Chemother. 59 (8) (2015) 4938–4945.
herpes simplex virus (HSV), Antiviral Res. 168 (2019) 128–133. [124] K. Tateishi, Y. Toh, H. Minagawa, H. Tashiro, Detection of herpes simplex virus
[100] Y.K. Shin, G.Y. Cai, A. Weinberg, J.J. Leary, M.J. Levin, Frequency of acyclovir- (HSV) in the saliva from 1,000 oral surgery outpatients by the polymerase chain
resistant herpes simplex virus in clinical specimens and laboratory isolates, reaction (PCR) and virus isolation, J. Oral Pathol. Med. 23 (2) (1994) 80–84.
J. Clin. Microbiol. 39 (3) (2001) 913–917. [125] R.B. Varella, M.A. Guimarães, A.C. Guimarães, A. Atalla, M. Nucci, M. Ramos-e-
[101] R. Stránská, R. Schuurman, D.R. Scholl, J.A. Jollick, C.J. Shaw, C. Loef, Silva, Acyclovir susceptibility of herpes simplex virus isolates from transplanted
M. Polman, A.M. van Loon, ELVIRA HSV, a yield reduction assay for rapid herpes and nontransplanted patients in Rio de Janeiro, Brazil, Skinmed 10 (4) (2012)
simplex virus susceptibility testing, Antimicrob. Agents Chemother. 48 (6) (2004) 208–211.
2331–2333. [126] C. Yoosook, W. Chantratita, P. Rimdusit, Recovery frequencies of herpes simplex
[102] C. McLaren, M.N. Ellis, G.A. Hunter, A colorimetric assay for the measurement of virus types 1 and 2 from symptomatic and asymptomatic genital herpes cases and
the sensitivity of herpes simplex viruses to antiviral agents, Antiviral Res. 3 (4) antiviral sensitivities of isolates, Journal of the Medical Association of Thailand
(1983) 223–234. =, Chotmaihet thangphaet 72 (10) (1989) 572–576.
[103] B. Svennerholm, A. Vahlne, G.B. Löwhagen, A. Widell, E. Lycke, Sensitivity of [127] M. Ziyaeyan, A. Alborzi, A. Japoni, M. Kadivar, M.A. Davarpanah, B. Pourabbas,
HSV strains isolated before and after treatment with acyclovir, Scand. J. Infect. A. Abassian, Frequency of acyclovir-resistant herpes simplex viruses isolated from
Dis. Suppl. 47 (1985) 149–154. the general immunocompetent population and patients with acquired
[104] E.M. Swierkosz, D.R. Scholl, J.L. Brown, J.D. Jollick, C.A. Gleaves, Improved DNA immunodeficiency syndrome, Int. J. Dermatol. 46 (12) (2007) 1263–1266.
hybridization method for detection of acyclovir-resistant herpes simplex virus, [128] S.M. Swetter, E.L. Hill, E.R. Kern, D.M. Koelle, C.M. Posavad, W. Lawrence,
Antimicrob. Agents Chemother. 31 (10) (1987) 1465–1469. S. Safrin, Chronic vulvar ulceration in an immunocompetent woman due to
[105] T.H. Bacon, M.J. Levin, J.J. Leary, R.T. Sarisky, D. Sutton, Herpes simplex virus acyclovir-resistant, thymidine kinase-deficient herpes simplex virus, J. Infect. Dis.
resistance to acyclovir and penciclovir after two decades of antiviral therapy, 177 (3) (1998) 543–550.
Clin. Microbiol. Rev. 16 (1) (2003) 114–128. [129] T. Czartoski, C. Liu, D.M. Koelle, S. Schmechel, A. Kalus, A. Wald, Fulminant,
[106] T.N. Thi, C. Deback, I. Malet, P. Bonnafous, Z. Ait-Arkoub, H. Agut, Rapid acyclovir-resistant, herpes simplex virus type 2 hepatitis in an immunocompetent
determination of antiviral drug susceptibility of herpes simplex virus types 1 and woman, J. Clin. Microbiol. 44 (4) (2006) 1584–1586.
2 by real-time PCR, Antiviral Res. 69 (3) (2006) 152–157. [130] R. Duan, R.D. de Vries, A.D. Osterhaus, L. Remeijer, G.M. Verjans, Acyclovir-
[107] M.J. Menage, E. de Clercq, A. van Lierde, V.S. Easty, J.M. Darville, S.D. Cook, D. resistant corneal HSV-1 isolates from patients with herpetic keratitis, J. Infect.
L. Easty, Antiviral drug sensitivity in ocular herpes simplex virus infection, Br. J. Dis. 198 (5) (2008) 659–663.
Ophthalmol. 74 (9) (1990) 532–535. [131] S.J. Charles, J.J. Gray, Ocular herpes simplex virus infections: reduced sensitivity
[108] Y. Wang, Q. Wang, Q. Zhu, R. Zhou, J. Liu, T. Peng, Identification and to acyclovir in primary disease, Br. J. Ophthalmol. 74 (5) (1990) 286–288.
characterization of acyclovir-resistant clinical HSV-1 isolates from children, [132] J.G. Mitterreiter, M.J. Titulaer, G.P. van Nierop, J.J. van Kampen, G.I. Aron, A.
J. Clin. Virol. 52 (2) (2011) 107–112. D. Osterhaus, G.M. Verjans, W.J. Ouwendijk, Prevalence of Intrathecal Acyclovir
[109] P. Collins, M.N. Ellis, Sensitivity monitoring of clinical isolates of herpes simplex Resistant Virus in Herpes Simplex Encephalitis Patients, PLoS ONE 11 (5) (2016)
virus to acyclovir, J Med Virol Suppl 1 (1993) 58–66. e0155531.
[110] S. Safrin, T. Elbeik, L. Phan, D. Robinson, J. Rush, A. Elbaggari, J. Mills, [133] K. Schepers, A. Hernandez, G. Andrei, S. Gillemot, P. Fiten, G. Opdenakker, J.
Correlation between response to acyclovir and foscarnet therapy and in vitro C. Bier, P. David, M.L. Delforge, F. Jacobs, R. Snoeck, Acyclovir-resistant herpes
susceptibility result for isolates of herpes simplex virus from human simplex encephalitis in a patient treated with anti-tumor necrosis factor-α
immunodeficiency virus-infected patients, Antimicrob. Agents Chemother. 38 (6) monoclonal antibodies, J. Clin. Virol. 59 (1) (2014) 67–70.
(1994) 1246–1250. [134] S. Kakiuchi, S. Nonoyama, H. Wakamatsu, K. Kogawa, L. Wang, H. Kinoshita-
[111] C. Danve-Szatanek, M. Aymard, D. Thouvenot, F. Morfin, G. Agius, I. Bertin, Yamaguchi, M. Takayama-Ito, C.K. Lim, N. Inoue, M. Mizuguchi, T. Igarashi,
S. Billaudel, B. Chanzy, M. Coste-Burel, L. Finkielsztejn, H. Fleury, T. Hadou, M. Saijo, Neonatal herpes encephalitis caused by a virologically confirmed
C. Henquell, H. Lafeuille, M.E. Lafon, A. Le Faou, M.C. Legrand, L. Maille, acyclovir-resistant herpes simplex virus 1 strain, J. Clin. Microbiol. 51 (1) (2013)
C. Mengelle, P. Morand, F. Morinet, E. Nicand, S. Omar, B. Picard, B. Pozzetto, 356–359.
J. Puel, D. Raoult, C. Scieux, M. Segondy, J.M. Seigneurin, R. Teyssou, [135] M. Bache, G. Andrei, L. Bindl, L. Bofferding, J. Bottu, C. Géron, C. Neuhäuser,
C. Zandotti, Surveillance network for herpes simplex virus resistance to antiviral S. Gillemot, P. Fiten, G. Opdenakker, R. Snoeck, Antiviral Drug-Resistance Typing
drugs: 3-year follow-up, J. Clin. Microbiol. 42 (1) (2004) 242–249. Reveals Compartmentalization and Dynamics of Acyclovir-Resistant Herpes
Simplex Virus Type-2 (HSV-2) in a Case of Neonatal Herpes, J Pediatric Infect Dis
Soc 3 (2) (2014) e24–e27.

19
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

[136] G. Carrega, E. Castagnola, A. Canessa, P. Argenta, R. Haupt, G. Dini, A. Garaventa, [159] G. Andrei, S. Gillemot, D. Topalis, R. Snoeck, The Anti-Human Immunodeficiency
Herpes simplex virus and oral mucositis in children with cancer, Support. Care Virus Drug Tenofovir, a Reverse Transcriptase Inhibitor, Also Targets the Herpes
Cancer 2 (4) (1994) 266–269. Simplex Virus DNA Polymerase, J Infect Dis 217 (5) (2018) 790–801.
[137] R.R. Razonable, Management of viral infections in solid organ transplant [160] M.R. Boyd, S. Safrin, E.R. Kern, Penciclovir: A review of its spectrum of activity,
recipients, Expert Rev Anti Infect Ther 9 (6) (2011) 685–700. selectivity, and cross-resistance pattern, Antiviral Chemistry and Chemotherapy,
[138] D.H. Lee, R.A. Zuckerman, Herpes simplex virus infections in solid organ Supplement 4 (1) (1993) 3–11.
transplantation: Guidelines from the American Society of Transplantation [161] C.B. Hwang, K.L. Ruffner, D.M. Coen, A point mutation within a distinct
Infectious Diseases Community of Practice, Clin. Transplant. 33 (9) (2019) conserved region of the herpes simplex virus DNA polymerase gene confers drug
e13526. resistance, J. Virol. 66 (3) (1992) 1774–1776.
[139] F. Morfin, K. Bilger, A. Boucher, A. Thiebaut, F. Najioullah, N. Bleyzac, N. Raus, [162] J.A. Hobden, M. Kumar, H.E. Kaufman, C. Clement, E.D. Varnell, P.
S. Bosshard, M. Aymard, M. Michallet, D. Thouvenot, HSV excretion after bone S. Bhattacharjee, J.M. Hill, In vitro synergism of trifluorothymidine and
marrow transplantation: a 4-year survey, J. Clin. Virol. 30 (4) (2004) 341–345. ganciclovir against HSV-1, Invest. Ophthalmol. Vis. Sci. 52 (2) (2011) 830–833.
[140] J.C. Wade, C. McLaren, J.D. Meyers, Frequency and significance of acyclovir- [163] T. Emura, N. Suzuki, M. Yamaguchi, H. Ohshimo, M. Fukushima, A novel
resistant herpes simplex virus isolated from marrow transplant patients receiving combination antimetabolite, TAS-102, exhibits antitumor activity in FU-resistant
multiple courses of treatment with acyclovir, J. Infect. Dis. 148 (6) (1983) human cancer cells through a mechanism involving FTD incorporation in DNA,
1077–1082. Int. J. Oncol. 25 (3) (2004) 571–578.
[141] E.J. Ariza-Heredia, R.F. Chemaly, L.R. Shahani, Y. Jang, R.E. Champlin, V. [164] E. De Clercq, Selective anti-herpesvirus agents, Antivir Chem Chemother 23 (3)
E. Mulanovich, Delay of alternative antiviral therapy and poor outcomes of (2013) 93–101.
acyclovir-resistant herpes simplex virus infections in recipients of allogeneic stem [165] H.A. Kessler, S. Hurwitz, C. Farthing, C.A. Benson, J. Feinberg, D.R. Kuritzkes, T.
cell transplant - a retrospective study, Transpl. Int. 31 (6) (2018) 639–648. C. Bailey, S. Safrin, R.T. Steigbigel, S.H. Cheeseman, G.F. McKinley, B. Wettlaufer,
[142] Y. Chen, C. Scieux, V. Garrait, G. Socié, V. Rocha, J.M. Molina, D. Thouvenot, S. Owens, T. Nevin, J.A. Korvick, Pilot study of topical trifluridine for the
F. Morfin, L. Hocqueloux, L. Garderet, H. Espérou, F. Sélimi, A. Devergie, treatment of acyclovir-resistant mucocutaneous herpes simplex disease in patients
G. Leleu, M. Aymard, F. Morinet, E. Gluckman, P. Ribaud, Resistant herpes with AIDS (ACTG 172), AIDS Clinical Trials Group, J Acquir Immune Defic Syndr
simplex virus type 1 infection: an emerging concern after allogeneic stem cell Hum Retrovirol 12 (2) (1996) 147–152.
transplantation, Clin. Infect. Dis. 31 (4) (2000) 927–935. [166] M. Murphy, A. Morley, R.P. Eglin, E. Monteiro, Topical trifluridine for
[143] D. Heidenreich, S. Kreil, N. Mueller, M. Jawhar, F. Nolte, W.K. Hofmann, S. mucocutaneous acyclovir-resistant herpes simplex II in AIDS patient, Lancet 340
A. Klein, Topical treatment of acyclovir-resistant herpes simplex virus stomatitis (8826) (1992) 1040.
after allogeneic hematopoietic cell transplantation, Oncology research and [167] W. Zhang, T. Suzuki, A. Shiraishi, I. Shimamura, Y. Inoue, Y. Ohashi, Dendritic
treatment 43 (12) (2020) 672–678. keratitis caused by an acyclovir-resistant herpes simplex virus with frameshift
[144] V. Anton-Vazquez, V. Mehra, J.L. Mbisa, D. Bradshaw, T.N. Basu, M.L. Daly, G. mutation, Cornea 26 (1) (2007) 105–106.
J. Mufti, A. Pagliuca, V. Potter, M. Zuckerman, Challenges of aciclovir-resistant [168] J. Piret, G. Boivin, Antiviral Drugs Against Herpesviruses, Adv. Exp. Med. Biol.
HSV infection in allogeneic bone marrow transplant recipients, J. Clin. Virol. 128 1322 (2021) 1–30.
(2020). [169] W. Leowattana, Antiviral Drugs and Acute Kidney Injury (AKI), Infect. Disord.
[145] A.A. Langston, I. Redei, A.M. Caliendo, J. Somani, D. Hutcherson, S. Lonial, Drug Targets 19 (4) (2019) 375–382.
S. Bucur, J. Cherry, A. Allen, E.K. Waller, Development of drug-resistant herpes [170] K.S. Erlich, M.A. Jacobson, J.E. Koehler, S.E. Follansbee, D.P. Drennan, L. Gooze,
simplex virus infection after haploidentical hematopoietic progenitor cell S. Safrin, J. Mills, Foscarnet therapy for severe acyclovir-resistant herpes simplex
transplantation, Blood 99 (3) (2002) 1085–1088. virus type-2 infections in patients with the acquired immunodeficiency syndrome
[146] E.E. Freeman, H.A. Weiss, J.R. Glynn, P.L. Cross, J.A. Whitworth, R.J. Hayes, (AIDS), An uncontrolled trial, Ann Intern Med 110 (9) (1989) 710–713.
Herpes simplex virus 2 infection increases HIV acquisition in men and women: [171] I. Schmit, G. Boivin, Characterization of the DNA polymerase and thymidine
systematic review and meta-analysis of longitudinal studies, Aids 20 (1) (2006) kinase genes of herpes simplex virus isolates from AIDS patients in whom
73–83. acyclovir and foscarnet therapy sequentially failed, J. Infect. Dis. 180 (2) (1999)
[147] S.L. Rowland-Jones, Timeline: AIDS pathogenesis: what have two decades of HIV 487–490.
research taught us? Nat. Rev. Immunol. 3 (4) (2003) 343–348. [172] H.T. Ho, K.L. Woods, J.J. Bronson, H. De Boeck, J.C. Martin, M.J. Hitchcock,
[148] E.K. Bagdades, D. Pillay, S.B. Squire, C. O’Neil, M.A. Johnson, P.D. Griffiths, Intracellular metabolism of the antiherpes agent (S)-1-[3-hydroxy-2-
Relationship between herpes simplex virus ulceration and CD4+ cell counts in (phosphonylmethoxy)propyl]cytosine, Mol. Pharmacol. 41 (1) (1992) 197–202.
patients with HIV infection, Aids 6 (11) (1992) 1317–1320. [173] E. De Clercq, T. Sakuma, M. Baba, R. Pauwels, J. Balzarini, I. Rosenberg, A. Holý,
[149] B. Ledergerber, M. Egger, V. Erard, R. Weber, B. Hirschel, H. Furrer, M. Battegay, Antiviral activity of phosphonylmethoxyalkyl derivatives of purine and
P. Vernazza, E. Bernasconi, M. Opravil, D. Kaufmann, P. Sudre, P. Francioli, pyrimidines, Antiviral Res. 8 (5–6) (1987) 261–272.
A. Telenti, AIDS-related opportunistic illnesses occurring after initiation of potent [174] J.M. Cherrington, S.J. Allen, B.H. McKee, M.S. Chen, Kinetic analysis of the
antiretroviral therapy: the Swiss HIV Cohort Study, JAMA 282 (23) (1999) interaction between the diphosphate of (S)-1-(3-hydroxy-2-
2220–2226. phosphonylmethoxypropyl)cytosine, ddCTP, AZTTP, and FIAUTP with human
[150] M. Augenbraun, J. Feldman, K. Chirgwin, J. Zenilman, L. Clarke, J. DeHovitz, DNA polymerases β and γ, Biochemical Pharmacology 48 (10) (1994) 1986–1988.
S. Landesman, H. Minkoff, Increased genital shedding of herpes simplex virus [175] X. Xiong, J.L. Smith, M.S. Chen, Effect of incorporation of cidofovir into DNA by
type 2 in HIV-seropositive women, Ann. Intern. Med. 123 (11) (1995) 845–847. human cytomegalovirus DNA polymerase on DNA elongation, Antimicrob. Agents
[151] L. Corey, A. Wald, C.L. Celum, T.C. Quinn, The effects of herpes simplex virus-2 Chemother. 41 (3) (1997) 594–599.
on HIV-1 acquisition and transmission: a review of two overlapping epidemics, [176] J.P. Lalezari, W.L. Drew, E. Glutzer, C. James, D. Miner, J. Flaherty, P.E. Fisher,
J. Acquir. Immune Defic. Syndr. 35 (5) (2004) 435–445. K. Cundy, J. Hannigan, J.C. Martin, et al., (S)-1-[3-hydroxy-2-
[152] S. Kakiuchi, M. Tsuji, H. Nishimura, T. Yoshikawa, L. Wang, M. Takayama-Ito, (phosphonylmethoxy)propyl]cytosine (cidofovir): results of a phase I/II study of a
H. Kinoshita, C.K. Lim, H. Fujii, S. Yamada, S. Harada, A. Oka, M. Mizuguchi, novel antiviral nucleotide analogue, J. Infect. Dis. 171 (4) (1995) 788–796.
S. Taniguchi, M. Saijo, Association of the Emergence of Acyclovir-Resistant [177] S.L. Williams-Aziz, C.B. Hartline, E.A. Harden, S.L. Daily, M.N. Prichard, N.
Herpes Simplex Virus Type 1 With Prognosis in Hematopoietic Stem Cell L. Kushner, J.R. Beadle, W.B. Wan, K.Y. Hostetler, E.R. Kern, Comparative
Transplantation Patients, J. Infect. Dis. 215 (6) (2017) 865–873. activities of lipid esters of cidofovir and cyclic cidofovir against replication of
[153] M.T. van der Beek, A. Laheij, J.E. Raber-Durlacher, P.A. von dem Borne, herpesviruses in vitro, Antimicrob. Agents Chemother. 49 (9) (2005) 3724–3733.
R. Wolterbeek, C.S. van der Blij-de Brouwer, C. van Loveren, E.C.J. Claas, A.C. [178] C.J. Detweiler, S.B. Mueller, A.D. Sung, J.L. Saullo, V.K. Prasad, D.M. Cardona,
M. Kroes, J.J. de Soet, A. Vossen, Viral loads and antiviral resistance of Brincidofovir (CMX001) Toxicity Associated With Epithelial Apoptosis and Crypt
herpesviruses and oral ulcerations in hematopoietic stem cell transplant Drop Out in a Hematopoietic Cell Transplant Patient: Challenges in Distinguishing
recipients, Bone Marrow Transplant. 47 (9) (2012) 1222–1228. Drug Toxicity From GVHD, J. Pediatr. Hematol. Oncol. 40 (6) (2018) e364–e368.
[154] E. De Clercq, E)-5-(2-bromovinyl)-2’-deoxyuridine (BVDU, Med. Res. Rev. 25 (1) [179] F.M. Marty, D.J. Winston, S.D. Rowley, E. Vance, G.A. Papanicolaou, K.
(2005) 1–20. M. Mullane, T.M. Brundage, A.T. Robertson, S. Godkin, H. Momméja-Marin,
[155] A. Sauerbrei, K. Bohn, A. Heim, J. Hofmann, B. Weissbrich, P. Schnitzler, M. Boeckh, CMX001 to prevent cytomegalovirus disease in hematopoietic-cell
D. Hoffmann, R. Zell, G. Jahn, P. Wutzler, K. Hamprecht, Novel resistance- transplantation, N. Engl. J. Med. 369 (13) (2013) 1227–1236.
associated mutations of thymidine kinase and DNA polymerase genes of herpes [180] F.M. Marty, D.J. Winston, R.F. Chemaly, K.M. Mullane, T.B. Shore, G.
simplex virus type 1 and type 2, Antivir Ther 16 (8) (2011) 1297–1308. A. Papanicolaou, G. Chittick, T.M. Brundage, C. Wilson, M.E. Morrison, S.
[156] R.T. Sarisky, M.R. Quail, P.E. Clark, T.T. Nguyen, W.S. Halsey, R.J. Wittrock, A. Foster, W.G. Nichols, M.J. Boeckh, A. Randomized, Double-Blind, Placebo-
J. O’Leary Bartus, M.M. Van Horn, G.M. Sathe, S. Van Horn, M.D. Kelly, T. Controlled Phase 3 Trial of Oral Brincidofovir for Cytomegalovirus Prophylaxis in
H. Bacon, J.J. Leary, Characterization of herpes simplex viruses selected in Allogeneic Hematopoietic Cell Transplantation, Biol Blood Marrow Transplant 25
culture for resistance to penciclovir or acyclovir, J. Virol. 75 (4) (2001) (2) (2019) 369–381.
1761–1769. [181] J. Lalezari, T. Schacker, J. Feinberg, J. Gathe, S. Lee, T. Cheung, F. Kramer,
[157] T. Karamitros, I. Harrison, R. Piorkowska, A. Katzourakis, G. Magiorkinis, J. H. Kessler, L. Corey, W.L. Drew, J. Boggs, B. McGuire, H.S. Jaffe, S. Safrin,
L. Mbisa, De Novo Assembly of Human Herpes Virus Type 1 (HHV-1) Genome, A randomized, double-blind, placebo-controlled trial of cidofovir gel for the
Mining of Non-Canonical Structures and Detection of Novel Drug-Resistance treatment of acyclovir-unresponsive mucocutaneous herpes simplex virus
Mutations Using Short- and Long-Read Next Generation Sequencing infection in patients with AIDS, J. Infect. Dis. 176 (4) (1997) 892–898.
Technologies, PLoS ONE 11 (6) (2016) e0157600. [182] T. Kopp, A. Geusau, A. Rieger, G. Stingl, Successful treatment of an aciclovir-
[158] E. Pelosi, G.B. Mulamba, D.M. Coen, Penciclovir and pathogenesis phenotypes of resistant herpes simplex type 2 infection with cidofovir in an AIDS patient, Br. J.
drug-resistant Herpes simplex virus mutants, Antiviral Res. 37 (1) (1998) 17–28. Dermatol. 147 (1) (2002) 134–138.

20
H.H. Schalkwijk et al. Biochemical Pharmacology 206 (2022) 115322

[183] D.B. Mendel, D.B. Barkhimer, M.S. Chen, Biochemical basis for increased [202] A. Serris, A. Pouvaret, C. Loiseau, H. Abid, S. Burrel, J. Fourgeaud, C. Rouzaud,
susceptibility to Cidofovir of herpes simplex viruses with altered or deficient F. Lanternier, D. Boutolleau, P. Frange, Pritelivir for recurrent aciclovir-resistant
thymidine kinase activity, Antimicrob. Agents Chemother. 39 (9) (1995) herpes simplex virus 2 infections in immunocompromised patients, J. Antimicrob.
2120–2122. Chemother. (2022).
[184] G. Andrei, P. Fiten, M. Froeyen, E. De Clercq, G. Opdenakker, R. Snoeck, DNA [203] L. Cannon, E. Tholouli, C. Ward, H. Farooq, M. Kingston, Use of pritelivir in
polymerase mutations in drug-resistant herpes simplex virus mutants determine refractory aciclovir-resistant herpes simplex virus type 2, Int. J. STD AIDS 32 (10)
in vivo neurovirulence and drug-enzyme interactions, Antivir Ther 12 (5) (2007) (2021) 978–980.
719–732. [204] L.E. Pope, J.F. Marcelletti, L.R. Katz, J.Y. Lin, D.H. Katz, M.L. Parish, P.G. Spear,
[185] D.L. Wyles, A. Patel, N. Madinger, M. Bessesen, P.R. Krause, A. Weinberg, The anti-herpes simplex virus activity of n-docosanol includes inhibition of the
Development of herpes simplex virus disease in patients who are receiving viral entry process, Antiviral Res. 40 (1–2) (1998) 85–94.
cidofovir, Clin. Infect. Dis. 41 (5) (2005) 676–680. [205] D.H. Katz, J.F. Marcelletti, M.H. Khalil, L.E. Pope, L.R. Katz, Antiviral activity of
[186] L. Naesens, R. Snoeck, G. Andrei, J. Balzarini, J. Neyts, E. De Clercq, HPMPC 1-docosanol, an inhibitor of lipid-enveloped viruses including herpes simplex,
(cidofovir), PMEA (adefovir) and related acyclic nucleoside phosphonate Proc Natl Acad Sci U S A 88 (23) (1991) 10825–10829.
analogues, A review of their pharmacology and clinical potential in the treatment [206] S.L. Sacks, R.A. Thisted, T.M. Jones, R.A. Barbarash, D.J. Mikolich, G.E. Ruoff, J.
of viral infections, Antiviral Chemistry and Chemotherapy 8 (1) (1997) 1–23. L. Jorizzo, L.B. Gunnill, D.H. Katz, M.H. Khalil, P.R. Morrow, G.J. Yakatan, L.
[187] S.S. Abdool Karim, Q. Abdool Karim, A.B. Kharsany, C. Baxter, A.C. Grobler, E. Pope, J.E. Berg, Clinical efficacy of topical docosanol 10% cream for herpes
L. Werner, A. Kashuba, L.E. Mansoor, N. Samsunder, A. Mindel, T.N. Gengiah, simplex labialis: A multicenter, randomized, placebo-controlled trial, J. Am.
Tenofovir Gel for the Prevention of Herpes Simplex Virus Type 2 Infection, Acad. Dermatol. 45 (2) (2001) 222–230.
N. Engl. J. Med. 373 (6) (2015) 530–539. [207] K.J. Vollmer, A.C. Burns, H.E. Sauer, J.D. Williams, G. Komazin-Meredith,
[188] J.M. Marrazzo, L. Rabe, C. Kelly, B. Richardson, C. Deal, J.L. Schwartz, Z. S. Cardinale, M. Butler, Z. Aron, I. Hussein, M.G. Busch, T.L. Bowlin, B.G. Gentry,
M. Chirenje, J. Piper, R.A. Morrow, C.W. Hendrix, M.A. Marzinke, S.L. Hillier, Activation of 6-Alkoxy-Substituted Methylenecyclopropane Nucleoside Analogs
Tenofovir Gel for Prevention of Herpes Simplex Virus Type 2 Acquisition: Requires Enzymatic Modification by Adenosine Deaminase-Like Protein 1,
Findings From the VOICE Trial, J. Infect. Dis. 219 (12) (2019) 1940–1947. Antimicrob. Agents Chemother. 63 (10) (2019).
[189] J.M. Marrazzo, G. Ramjee, B.A. Richardson, K. Gomez, N. Mgodi, G. Nair, [208] G. Komazin-Meredith, S.C. Cardinale, K. Comeau, K.J. Magalhaes, C.B. Hartline,
T. Palanee, C. Nakabiito, A. van der Straten, L. Noguchi, C.W. Hendrix, J.Y. Dai, J.D. Williams, T.J. Opperman, M.N. Prichard, T.L. Bowlin, TAOK3 phosphorylates
S. Ganesh, B. Mkhize, M. Taljaard, U.M. Parikh, J. Piper, B. Mâsse, C. Grossman, the methylenecyclopropane nucleoside MBX 2168 to its monophosphate,
J. Rooney, J.L. Schwartz, H. Watts, M.A. Marzinke, S.L. Hillier, I.M. McGowan, Z. Antiviral Res. 119 (2015) 23–27.
M. Chirenje, Tenofovir-based preexposure prophylaxis for HIV infection among [209] M.N. Prichard, J.D. Williams, G. Komazin-Meredith, A.R. Khan, N.B. Price, G.
African women, N. Engl. J. Med. 372 (6) (2015) 509–518. M. Jefferson, E.A. Harden, C.B. Hartline, N.P. Peet, T.L. Bowlin, Synthesis and
[190] K. Katsumata, A. Weinberg, K. Chono, S. Takakura, T. Kontani, H. Suzuki, antiviral activities of methylenecyclopropane analogs with 6-alkoxy and 6-
Susceptibility of herpes simplex virus isolated from genital herpes lesions to alkylthio substitutions that exhibit broad-spectrum antiviral activity against
ASP2151, a novel helicase-primase inhibitor, Antimicrob. Agents Chemother. 56 human herpesviruses, Antimicrob. Agents Chemother. 57 (8) (2013) 3518–3527.
(7) (2012) 3587–3591. [210] V. Pirrone, N. Thakkar, J.M. Jacobson, B. Wigdahl, F.C. Krebs, Combinatorial
[191] H.J. Field, M.L. Huang, E.M. Lay, I. Mickleburgh, H. Zimmermann, A. Birkmann, approaches to the prevention and treatment of HIV-1 infection, Antimicrob.
Baseline sensitivity of HSV-1 and HSV-2 clinical isolates and defined acyclovir- Agents Chemother. 55 (5) (2011) 1831–1842.
resistant strains to the helicase-primase inhibitor pritelivir, Antiviral Res. 100 (2) [211] K. Chono, K. Katsumata, H. Suzuki, K. Shiraki, Synergistic activity of amenamevir
(2013) 297–299. (ASP2151) with nucleoside analogs against herpes simplex virus types 1 and 2
[192] G. Kleymann, R. Fischer, U.A. Betz, M. Hendrix, W. Bender, U. Schneider, and varicella-zoster virus, Antiviral Res. 97 (2) (2013) 154–160.
G. Handke, P. Eckenberg, G. Hewlett, V. Pevzner, J. Baumeister, O. Weber, [212] Z.W. Greeley, N.J. Giannasca, M.J. Porter, B.J. Margulies, Acyclovir, cidofovir,
K. Henninger, J. Keldenich, A. Jensen, J. Kolb, U. Bach, A. Popp, J. Mäben, and amenamevir have additive antiviral effects on herpes simplex virus TYPE 1,
I. Frappa, D. Haebich, O. Lockhoff, H. Rübsamen-Waigmann, New helicase- Antiviral Res. 176 (2020), 104754.
primase inhibitors as drug candidates for the treatment of herpes simplex disease, [213] G. Kleymann, New antiviral drugs that target herpesvirus helicase primase
Nat. Med. 8 (4) (2002) 392–398. enzymes, Herpes 10 (2) (2003) 46–52.
[193] P.T. Edlefsen, A. Birkmann, M.L. Huang, C.A. Magaret, J.J. Kee, K. Diem, [214] E.C. Schulte, A. Sauerbrei, D. Hoffmann, C. Zimmer, B. Hemmer, M. Mühlau,
T. Goldner, B. Timmler, S. Stoelben, H. Ruebsamen-Schaeff, H. Zimmermann, Acyclovir resistance in herpes simplex encephalitis, Ann. Neurol. 67 (6) (2010)
T. Warren, A. Wald, L. Corey, No Evidence of Pritelivir Resistance Among Herpes 830–833.
Simplex Virus Type 2 Isolates After 4 Weeks of Daily Therapy, J. Infect. Dis. 214 [215] Z.G. Gayretli Aydin, G. Tanir, C. Genc Sel, Y. Tasci Yıldız, T. Aydin Teke,
(2) (2016) 258–264. A. Kaman, Acyclovir Unresponsive Herpes Simplex Encephalitis in a child
[194] A. Wald, L. Corey, B. Timmler, A. Magaret, T. Warren, S. Tyring, C. Johnston, successfully treated with the addition of Foscarnet 117 (2019) e47–e51. Case
J. Kriesel, K. Fife, L. Galitz, S. Stoelben, M.L. Huang, S. Selke, H.P. Stobernack, report,.
H. Ruebsamen-Schaeff, A. Birkmann, Helicase-primase inhibitor pritelivir for [216] N. Katyal, A.M. Taqui, D. Tepper, J.M. Beary, C.R. Newey, Fulminant Herpes
HSV-2 infection, N. Engl. J. Med. 370 (3) (2014) 201–210. Simplex Virus Type I Encephalitis Despite Maximal Medical Therapy, Cureus 10
[195] A. Wald, B. Timmler, A. Magaret, T. Warren, S. Tyring, C. Johnston, K. Fife, (4) (2018) e2467.
S. Selke, M.L. Huang, H.P. Stobernack, H. Zimmermann, L. Corey, A. Birkmann, [217] H.H. Schalkwijk, S. Gillemot, M. Reynders, D. Selleslag, G. Andrei, R. Snoeck,
H. Ruebsamen-Schaeff, Effect of Pritelivir Compared With Valacyclovir on Genital Heterogeneity and viral replication fitness of HSV-1 clinical isolates with
HSV-2 Shedding in Patients With Frequent Recurrences: A Randomized Clinical mutations in the thymidine kinase and DNA polymerase, J. Antimicrob.
Trial, JAMA 316 (23) (2016) 2495–2503. Chemother. (2022).
[196] K. Chono, K. Katsumata, T. Kontani, M. Kobayashi, K. Sudo, T. Yokota, K. Konno, [218] T. Labrunie, S. Ducastelle, C. Domenech, F. Ader, F. Morfin, E. Frobert, UL23,
Y. Shimizu, H. Suzuki, ASP2151, a novel helicase-primase inhibitor, possesses UL30, and UL5 characterization of HSV1 clinical strains isolated from hematology
antiviral activity against varicella-zoster virus and herpes simplex virus types 1 department patients, Antiviral Res. 168 (2019) 114–120.
and 2, J. Antimicrob. Chemother. 65 (8) (2010) 1733–1741. [219] N. Rabella, M. Otegui, R. Labeaga, P. Rodriguez, N. Margall, M. Gurgui, G. Prats,
[197] N. Shoji, K. Tanese, A. Sasaki, T. Horiuchi, Y. Utsuno, K. Fukuda, Y. Hoshino, Antiviral susceptibility of herpes simplex viruses and its clinical correlates: A
S. Noda, H. Minami, W. Asakura, Pharmaceuticals and Medical Device Agency single center’s experience, Clin. Infect. Dis. 34 (8) (2002) 1055–1060.
approval summary: Amenamevir for the treatment of herpes zoster, J. Dermatol. [220] G. Boivin, A. Erice, D.D. Crane, D.L. Dunn, H.H. Balfour Jr, Acyclovir
47 (7) (2020) 683–688. susceptibilities of herpes simplex virus strains isolated from solid organ transplant
[198] T. Kusawake, J.J. Keirns, D. Kowalski, M. den Adel, D. Groenendaal-van de recipients after acyclovir or ganciclovir prophylaxis, Antimicrob. Agents
Meent, A. Takada, Y. Ohtsu, M. Katashima, Pharmacokinetics and Safety of Chemother. 37 (2) (1993) 357–359.
Amenamevir in Healthy Subjects: Analysis of Four Randomized Phase 1 Studies, [221] S. Chakrabarti, D. Pillay, D. Ratcliffe, P.A. Cane, K.E. Collingham, D.W. Milligan,
Adv Ther 34 (12) (2017) 2625–2637. Resistance to antiviral drugs in herpes simplex virus infections among allogeneic
[199] S. Tyring, A. Wald, N. Zadeikis, S. Dhadda, K. Takenouchi, R. Rorig, ASP2151 for stem cell transplant recipients: risk factors and prognostic significance, J. Infect.
the treatment of genital herpes: a randomized, double-blind, placebo- and Dis. 181 (6) (2000) 2055–2058.
valacyclovir-controlled, dose-finding study, J. Infect. Dis. 205 (7) (2012) [222] W.G. Nichols, M. Boeckh, R.A. Carter, A. Wald, L. Corey, Transferred herpes
1100–1110. simplex virus immunity after stem-cell transplantation: Clinical implications,
[200] K. Chono, K. Katsumata, T. Kontani, K. Shiraki, H. Suzuki, Characterization of J. Infect. Dis. 187 (5) (2003) 801–808.
virus strains resistant to the herpes virus helicase-primase inhibitor ASP2151 [223] E.E. Muller, D.V. Maseko, R.S. Kularatne, Phenotypic and genotypic acyclovir
(Amenamevir), Biochem. Pharmacol. 84 (4) (2012) 459–467. resistance surveillance of genital herpes simplex virus 2 in South Africa, Antiviral
[201] S. Biswas, L. Jennens, H.J. Field, Single amino acid substitutions in the HSV-1 Res. 200 (2022), 105277.
helicase protein that confer resistance to the helicase-primase inhibitor BAY [224] S. Nag, M. Sengupta, S. Sarkar, Y. Ray, D. Chattopadhyay, M. Sengupta, Isolation,
57–1293 are associated with increased or decreased virus growth characteristics characterization and acyclovir susceptibility of herpes simplex virus isolates
in tissue culture, Arch. Virol. 152 (8) (2007) 1489–1500. among immunocompromised patients, Indian Journal of Sexually Transmitted
Diseases and Aids 42 (1) (2021) 19–23.

21

You might also like