ArticlePDF Available

Cytomegalovirus Antiviral Drug Resistance: Future Prospects for Prevention, Detection and Management

Taylor & Francis
Future Microbiology
Authors:

Abstract

Human cytomegalovirus (CMV) is one of the most important viral pathogens affecting hematopoietic stem cell transplant (HSCT) and solid organ transplant (SOT) recipients, because infection is frequent and it exerts short- and long-term effects on host immunity. The direct effects of active CMV infection include fever, bone marrow suppression and invasive end organ disease. Indirectly, CMV increases risk for other infections and is associated with increased rejection, graft loss, mortality and transplant costs [1]. Given the impact of CMV on transplant outcomes, prevention of active infection and aggressive treatment of invasive disease are essential. Prevention strategies include universal antiviral prophylaxis or pre-emptive therapy that is based on sequential viral load monitoring in blood. Both approaches prevent CMV disease in at-risk patients, but they are limited by the toxicities of available anti-CMV drugs (i.e., cytopenias from valganiclovir, ganciclovir and cidofovir, and nephrotoxicity from foscarnet and cidofovir) and the potential for the virus to develop antiviral drug resistance (AVR). Furthermore, optimal viral load thresholds for the initiation of pre-emptive therapy have not been defined and it is likely that these cut-offs will vary based on host factors as well as the quantitative PCR assay used for testing. Due to these complexities, an ideal preventative approach has yet to be identified. Only prophylaxis would be expected to prevent CMV indirect effects, but early treatment of viremia (i.e., pre-emptive therapy) may allow some reconstitution of protective immunity. Current approaches HSCT programs generally apply preemptive therapy to avoid the marrow suppressive effects of valganciclovir (VGCV) or ganciclovir (GCV). Some centers also utilize prophylaxis for unrelated, HLAmismatched or cord blood transplants. SOT programs also typically use prophylaxis for the highest risk patients (CMV seropositive donor [D+]/CMV seronegative recipient [R-]), and may consider pre-emptive approaches for moderate-risk R+ liver or kidney recipients. The routine use of prolonged CMV prophylaxis (i.e., ≥6 months of VGCV) has raised concern about AVR,
1545
ISSN 1746- 0913
Future Microbiol. (2015) 10(10) , 1545 –1548
part of
10.2217/fmb.15.82 © 2015 Future Medicine Ltd
EDITORIAL
Cytomegalovirus antiviral drug
resistance: future prospects
for prevention, detection and
management
Kimberly E Hanson1,2 & Sankar Swaminathan*,1
Keywords
adoptive immunotherapy
antiviral drug resistance
cytomegalovirus human CMV
Human cytomegalovirus (CMV) is one
of the most important viral pathogens
affecting hematopoietic stem cell trans-
plant (HSCT) and solid organ transplant
(SOT) recipients, because infection is fre-
quent and it exerts short- and long-term
effects on host immunity. The direct
effects of active CMV infection include
fever, bone marrow suppression and inva-
sive end organ disease. Indirectly, CMV
increases risk for other infections and is
associated with increased rejection, graft
loss, mortality and transplant costs [1] .
Given the impact of CMV on transplant
outcomes, prevention of active infection
and aggressive treatment of invasive disease
are essential.
Prevention strategies include univer-
sal antiviral prophylaxis or pre-emptive
therapy that is based on sequential viral
load monitoring in blood. Both approaches
prevent CMV disease in at-risk patients,
but they are limited by the toxicities of
available anti-CMV drugs (i.e., cytopenias
from valganiclovir, ganciclovir and cidofo-
vir, and nephrotoxicity from foscarnet and
cidofovir) and the potential for the virus to
develop antiviral drug resistance (AVR).
Furthermore, optimal viral load thresholds
for the initiation of pre-emptive therapy
have not been defined and it is likely that
these cut-offs will vary based on host fac-
tors as well as the quantitative PCR assay
used for testing. Due to these complexi-
ties, an ideal preventative approach has
yet to be identified. Only prophylaxis
would be expected to prevent CMV indi-
rect effects, but early treatment of viremia
(i.e., pre-emptive therapy) may allow some
reconstitution of protective immunity.
Current approaches
HSCT programs generally apply pre-
emptive therapy to avoid the marrow sup-
pressive effects of valganciclovir (VGCV)
or ganciclovir (GCV). Some centers also
utilize prophylaxis for unrelated, HLA-
mismatched or cord blood transplants. SOT
programs also typically use prophylaxis for
the highest risk patients (CMV seroposi-
tive donor [D+]/CMV seronegative recipi-
ent [R-]), and may consider pre-emptive
approaches for moderate-risk R+ liver or
kidney recipients. The routine use of pro-
longed CMV prophylaxis (i.e., 6 months
of VGCV) has raised concern about AVR,
1Department o f Medicine, Division of Infec tious Diseases, Universit y of Utah, UT, USA
2Departm ent of Pathology, ARUP Laboratories, Universit y of Utah, UT, USA
*Author for correspondence: sankar.swaminathan@hsc.utah.edu
Antiviral drug resistance is a
dreaded complication
because it makes treatment
more difficult, is associated
with poor clinical outcomes
and may lead to allograft loss
or death.
For reprint orders, please contact: reprints@futuremedicine.com
Future Microbiol. (2015) 10(10)
154 6
editorial Hanson & Swaminathan
future science group
with the potential to develop cross-resistance to
currently available drugs. A recent report also
described a high incidence of AVR among D+/R-
kidney recipients receiving pre-emptive ther-
apy [2] . AVR is a dreaded complication because
it makes treatment more difficult, is associated
with poor clinical outcomes and may lead to
allograft loss or death [3] .
GCV and its oral prodrug VGCV are the cur-
rent cornerstones of CMV therapy. These agents
are nucleoside analogs that require phosphoryla-
tion by the viral protein kinase (UL97 gene) in
order to inhibit the CMV DNA polymerase
(UL54 gene) that is required for viral replica-
tion. Certain mutations within the UL97 and/
or UL54 genes have been linked to GCV and
VGCV resistance (reviewed in [4]). Cidofovir
(CDV) and foscarnet (FOS) act at the level of
the DNA polymerase. Mutations in the UL54
gene can confer resistance to CDV, FOS or cross-
resistance to both CDV and GCV. CDV resist-
ance has been reported following GCV therapy,
while FOS resistance typically requires previous
exposure to that drug.
CMV AVR is suspected when viral load
increases or plateaus despite at least 2 weeks of
appropriate antiviral therapy. Drug resistance
is then confirmed by detecting specific gene
mutations or with phenotypic drug susceptibility
testing. Most laboratories use standard Sanger
DNA sequencing of known resistance loci to
detect drug resistance mutations. Confirmation
of genotypic resistance is preferred because host
factors or inadequate drug levels may also affect
treatment response, and empiric changes in anti-
viral therapy often have adverse side effects. A
limitation of current laboratory practice is that
standard DNA sequencing may not detect drug-
resistant heterogeneity (<20–30%) in a mixed
viral population. Minority CMV populations
can be clinically relevant and newer technolo-
gies such as next-generation sequencing [5] , that
can detect subpopulations on the order of 1–3%,
are being explored for the enhanced detection of
genotypic resistance.
Most AVR has been reported in the context
of long-term GCV or VGCV use, usually after
at least 6 weeks of drug exposure [6] . Precise esti-
mates of AVR prevalence and risk in an era of
VGCV prophylaxis and pre-emptive therapy are
limited by the paucity of large prospective stud-
ies. In general, CMV AVR appears to be rela-
tively uncommon after HSCT [7] but may affect
as many 5–27% of viremic SOT recipients, with
the highest incidence following lung transplan-
tation [8]. Risk factors for AVR include D+/R-
serostatus in SOT, lung transplantation, high
viral load, persistent viremia on therapy and sub-
optimal GCV concentrations [9] , as well as hap-
loidentical T-cell-depleted HSCT [10 ] . The exact
role, if any, that therapeutic drug monitoring
should have in detecting subtherapeutic GCV
drug concentrations needs to be better defined.
Future possibilities
Given the safety profile of traditional drugs in
conjunction with the problem of AVR, better-
tolerated anti-CMV agents with different
mechanisms of action are needed. The primary
risk for active infection after transplantation is a
reduced number and function of CMV-reactive
T cells. Therefore, reduction in immunosuppres-
sion when feasible is an essential component of
CMV management.
Adoptive transfer of donor-derived or third
party T cells is being explored as a way to pro-
mote immune reconstitution and reduce viral
burden. Perhaps the most exciting approach is
the generation and expansion of broad-spectrum,
cytotoxic donor T lymphocytes in vitro which
recognize virus-specific epitopes, persist and pro-
liferate in vivo [1 1] . These strategies have been
used in prophylactic and pre-emptive regimens
as well as for the treatment of clinically resist-
ant infection. Despite significant heterogeneity,
most studies have observed that CMV-specific
T-cell immunity can be restored without induc-
ing graft-versus-host disease or other significant
toxicity [12–14]. It is possible that adoptive T-cell
therapy could be used in combination with
shorter course antiviral prophylaxis to enhance
cellular immune protection while minimizing
antiviral drug side effects, cost and AVR.
Three experimental CMV antivirals have
entered Phase II and III clinical trials. Maribavir
(MBV) and letermovir (LTV) have unique
mechanisms of action. MBV is an oral UL97
protein kinase inhibitor that initially failed
Phase III prophylaxis trials possibly due to
inappropriate dose selection [15] . More recently,
two as of yet unpublished Phase II dose-rang-
ing studies have been completed that examined
higher doses of MBV as pre-emptive therapy
(EuraCT: 2010-024247-32) and for refractory
or resistant CMV (ClinicalTrials.gov identi-
fier: NCT01611974). Since UL97 is needed to
phosphorylate GCV, therapy with MBV and
GCV would be expected to be antagonistic.
Three experimental
human cytomegalovirus
antivirals have entered
Phase II and III clinical
trials.
1547
Cytomegalovirus antiviral drug resistance editorial
future science group www.futuremedicine.com
However, the UL97 mutations elicited by MBV
and GCV are different and MBV retains in vitro
activity against most GCV- and CDV-resistant
viruses [16 ] . Of note, there is a single p-loop
mutation (F342S) that has been shown to confer
dual cross-resistance between MBV and GCV,
but does not affect kinase activity or growth in
cell culture [17 ] . The F342S mutation has not yet
been observed in vivo.
LTV (formerly AIC-246) is a CMV-specific
viral terminase inhibitor with potent in vitro
activity against wild-type and drug-resistant
virus. Oral and intravenous formulations have
been developed, which is of potential ben-
efit for patients unable to tolerate or absorb
oral therapy. LTV recently entered Phase III
prophylaxis studies (ClinicalTrials.gov iden-
tifier: NCT02137772) after successful com-
pletion of an HSCT Phase II dose-escalation
trial [18]. No AVR information was provided in
the Phase II report. However, high-grade resist-
ance mutations in the UL56 terminase gene are
readily selected in vitro and await clinical cor-
relation [19] . The potentially low barrier to LTV
resistance may limit the utility of this agent as
monotherapy for severe disease or high-grade
viremia.
Brincidofovir (formerly CMX-001) is a broad-
spectrum, orally bioavailable lipid conjugate of
CDV. Unlike CDV, brincidofovir (BCV) is not
a substrate for human organic anion transport-
ers (hOATs) and therefore has reduced potential
to cause renal impairment. A Phase II prophy-
laxis study in HSCT was completed wherein no
development of AVR was noted [2 0] . Phase III
trials are currently underway (ClinicalTrials.gov
identifier: NCT0176170). Despite overlapping
resistance mechanisms, BCV may have a role in
treating genotypically resistant infection because
the drug has potent in vitro activity and achieves
high intracellular concentrations.
Combination antiviral therapy is the standard
of care for other viral infections, such as HIV
and HCV. Dual DNA polymerase inhibitor
treatment for CMV, however, has been limited
by additive toxicity, cross-resistance and a lack
of evidence to support effectiveness. We have
now entered an exciting time of late-phase drug
development for several anti-CMV agents with
novel drug targets, potent activity and reduced
toxicity. Additionally, CMV-specific immuno-
therapies are under active investigation. These
agents used alone or in combination, create new
opportunities to refine our management strate-
gies and potentially provide therapeutic alterna-
tives for drug-resistant CMV. Studies are now
needed to determine whether combinations of
the new agents with different mechanisms of
action can improve treatment efficacy and/or
reduce the risk for AVR. Additionally, stud-
ies comparing prophylactic versus pre-emptive
approaches, that utilize standardized CMV
assays and are powered to assess both the direct
as well as indirect effects of CMV, will better
inform clinical practice in the future.
Financial & competing interests disclosure
The authors have no relevant affiliations or financial
involvement with any organization or entity with a finan-
cial interest in or financial conflict with the subject matter
or materials discussed in the manuscript. This includes
employment, consultancies, honoraria, stock ownership or
options, expert testimony, grants or patents received or
pending, or royalties.
No writing assistance was utilized in the production of
this manuscript.
We have now entered
an exciting time of late-
phase drug
development for several
anti-cytomegalovirus
agents with novel drug
targets, potent activity and
reduced toxicity.
References
1 Roman A, Manito N, Ca mpistol JM et al.
The impact of the prevention strategies on the
indirect effects of CMV infection in solid
organ transplant recipients. Transplant. Rev.
28(2), 84–91 (2014).
2 Couzi L , Helou S, Bachelet T et al. High
incidence of anticytomegalovirus drug
resistance among D+R- kidney transplant
recipients receiving preemptive therapy. Am.
J. Transplant. 12(1), 202–209 (2012).
3 Eid AJ, Arthurs SK, Deziel PJ, Wilhelm MP,
Razonable RR . Emergence of drug-resistant
cytomegalovirus in the era of valganciclovir
prophylax is: therapeutic implications and
outcomes. Clin. Transplant. 22(2), 162–170
(2008).
4 Lurain NS, Chou S. Antivira l drug resistance
of human cytomegalovirus. Clin . Microbiol.
Rev. 23(4), 689–712 (2010).
5 Sahoo MK, Lefterova MI, Yama moto F
et al. Detection of cytomegalovirus drug
resistance mutations by next-generation
sequencing. J. Clin. Microbiol. 51(11),
3700 –3710 (2013).
6 Kotton CN, Kumar D, Caliendo AM et al.
Updated international consensus guidelines
on the management of cytomega lovirus in
solid-organ transplantation. Transplantation
96(4), 333–360 (2 013).
7 van der Beek MT, Marijt EW, Vossen AC
et al. Failure of pre-emptive treatment of
cytomegalovirus infections and antiviral
resistance in stem cell transplant recipients.
Antiviral Ther. 17(1), 45–51 (2012).
8 Limaye A P. Ganciclovir-resistant
cytomegalovirus in organ transplant
recipients. Clin. Infect. Dis. 35(7), 866 –872
(2002).
9 Le Page AK, Jager MM, Iwasenko JM, Scott
GM, Alain S, Rawlinson WD. Clinical
aspects of cytomegalovirus antiviral
resistance in solid organ transplant
recipients. Clin. Infect. Dis. 5 6(7) , 1018 –1029
(2013) .
Future Microbiol. (2015) 10(10)
154 8
editorial Hanson & Swaminathan
future science group
10 Shmueli E, Or R, Shapira MY et al . High rate
of cytomegalovirus drug resistance a mong
patients receiving preemptive antiviral treat ment
after haploidentical stem cell transplantation.
J. Infect. Dis. 209(4), 557–561 (2014).
11 Papadopoulou A, Gerdemann U, Katari UL
et al. Activity of broad-spectrum T cells as
treatment for AdV, EBV, CMV, BKV, and
HHV6 infections after HSCT. Sci. Translat.
Med. 6(242), 242ra283 (2014).
12 Schmitt A, Tonn T, Busch DH et al. Adoptive
transfer and selective reconstitution of
streptamer-selected cytomegalovirus-specific
CD8+ T cells leads to virus clearance in
patients after allogeneic peripheral blood stem
cell transplantation. Transfusion 51(3),
591–599 (2011).
13 Uhlin M, Gertow J, Uzunel M et al. Rapid
salvage treatment with virus-specific T cells
for therapy-resistant disease. Clin. Infect. Dis.
55( 8) , 1064 –1073 (2 012).
14 Leen A M, Bollard CM, Mendizabal AM et al.
Multicenter study of banked third-party
virus-specific T cells to treat severe viral
infections after hematopoietic stem cell
transplantation. Blood 121(26), 5113–5123
(2013) .
15 Marty FM, Boeckh M. Maribavir and human
cytomegalovirus-what happened in the
clinical trials and why might the drug have
failed? Curr. Opin. Virol. 1(6), 555–562
(2 011).
16 Drew WL, Miner RC, Marousek GI, Chou S.
Maribavir sensitivity of cytomega lovirus
isolates resistant to ganciclovir, cidofovir or
foscarnet. J. Clin. Virol. 37(2), 124–127
(2006).
17 Chou S, Ercolani RJ, Marousek G, Bowlin
TL. Cytomegalovirus UL97 kinase catalytic
domain mutations that confer multidrug
resistance. Antimicrob. Agents Chemother.
57(7), 3375–3379 (2013).
18 Chemaly RF, Ullmann AJ, Stoelben S et al.
Letermovir for cy tomegalovirus prophylax is
in hematopoietic-cell transplantation.
N. Engl. J. Med. 370(19), 1781–1789 ( 2014) .
19 Goldner T, Hempel C, Ruebsamen-Schaeff H,
Zimmermann H, Lischka P. Geno- and
phenotypic characterization of human
cytomegalovirus mutants selected in vitro after
letermovir (AIC246) exposure. Antimicrob.
Agents Chemother. 58(1) , 610 –613 (2014 ).
20 Marty FM, Winston DJ, Rowley SD et al.
CMX001 to prevent cytomegalovirus disease
in hematopoietic-cell transplantation.
N. Engl. J. Med. 369 (13), 1227–1236 (2013).
... Antiviral drug medication has significantly reduced the spread of epidemics, but their continuous usage has led in the emergence of drug resistant mutants over a period of time [31][32][33]. Several antiviral drugs have low oral bioavailability, show adverse side effects and are too expensive [34][35][36]. Hence, there is important necessity to develop safe and effective nonnucleoside antiviral agents. ...
... This compound is found to be more potent than the drug, zileuton (Fig. 10) [6]. Chloro acetylation of 2-(methylthio)-1H-benzo[d]imidazole (34) in presence of NaH and DMF, gives pure product imidazole derivative (35), which undergoes cyclization with thiourea in presence of ethanol resulting in aminothiazole derivatives (36). Compound 36 on alkylation with the equivalent amount of chloroacetone in presence of acetone in anhydrous K2CO3 gives benzo[d]thiazole derivative (37). ...
Article
Full-text available
Thiazoles are notable five-membered heterocyclic rings and their moieties can be found in several biologically active compounds of natural origin, as well as synthetic molecules that possess a wide range of pharmacological activities. Inflammation is the common cause that is associated with different disorders and diseases such as psoriasis, arthritis, infections, asthma, cancer, etc. In this article, the synthesis pattern of these novel molecules are discussed and their anti-inflammatory activities against cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2) and lipoxygenase (LOX) were reviewed and documented. The potent 26 thiazole analogs were validated with molecular docking against main protease (6LU7) and spike binding domain ACE2 receptor (6M0J) to defeat from the COVID-19 infections. Among this, THI- 9a showed excellent binding energy and affinity against deadly SAR CoV-2. The reviewed and theoretical study information strongly suggested that thiazole derivatives can be used for the development of futuristic target drugs against death-causing diseases like SAR-CoV-2.
... Antiviral drug medication has significantly reduced the spread of epidemics, but their continuous usage has led in the emergence of drug resistant mutants over a period of time [31][32][33]. Several antiviral drugs have low oral bioavailability, show adverse side effects and are too expensive [34][35][36]. Hence, there is important necessity to develop safe and effective nonnucleoside antiviral agents. ...
... This compound is found to be more potent than the drug, zileuton (Fig. 10) [6]. Chloro acetylation of 2-(methylthio)-1H-benzo[d]imidazole (34) in presence of NaH and DMF, gives pure product imidazole derivative (35), which undergoes cyclization with thiourea in presence of ethanol resulting in aminothiazole derivatives (36). Compound 36 on alkylation with the equivalent amount of chloroacetone in presence of acetone in anhydrous K2CO3 gives benzo[d]thiazole derivative (37). ...
Article
Thiazoles are notable five-membered heterocyclic rings and their moieties can be found in several biologically active compounds of natural origin, as well as synthetic molecules that possess a wide range of pharmacological activities. Inflammation is the common cause that is associated with different disorders and diseases such as psoriasis, arthritis, infections, asthma, cancer, etc. In this article, the synthesis pattern of these novel molecules are discussed and their anti-inflammatory activities against cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2) and lipoxygenase (LOX) were reviewed and documented. The potent 26 thiazole analogs were validated with molecular docking against main protease (6LU7) and spike binding domain ACE2 receptor (6M0J) to defeat from the COVID-19 infections. Among this, THI- 9a showed excellent binding energy and affinity against deadly SAR CoV-2. The reviewed and theoretical study information strongly suggested that thiazole derivatives can be used for the development of futuristic target drugs against death-causing diseases like SAR-CoV-2.
... Antiviral drug medication has significantly reduced the spread of epidemics, but their continuous usage has led in the emergence of drug resistant mutants over a period of time [31][32][33]. Several antiviral drugs have low oral bioavailability, show adverse side effects and are too expensive [34][35][36]. Hence, there is important necessity to develop safe and effective nonnucleoside antiviral agents. ...
... This compound is found to be more potent than the drug, zileuton (Fig. 10) [6]. Chloro acetylation of 2-(methylthio)-1H-benzo[d]imidazole (34) in presence of NaH and DMF, gives pure product imidazole derivative (35), which undergoes cyclization with thiourea in presence of ethanol resulting in aminothiazole derivatives (36). Compound 36 on alkylation with the equivalent amount of chloroacetone in presence of acetone in anhydrous K2CO3 gives benzo[d]thiazole derivative (37). ...
Article
Thiazoles are notable five-membered heterocyclic rings and their moieties can be found in several biologically active compounds of natural origin, as well as synthetic molecules that possess a wide range of pharmacological activities. Inflammation is the common cause that is associated with different disorders and diseases such as psoriasis, arthritis, infections, asthma, cancer, etc. In this article, the synthesis pattern of these novel molecules are discussed and their anti-inflammatory activities against cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2) and lipoxygenase (LOX) were reviewed and documented. The potent 26 thiazole analogs were validated with molecular docking against main protease (6LU7) and spike binding domain ACE2 receptor (6M0J) to defeat from the COVID-19 infections. Among this, THI- 9a showed excellent binding energy and affinity against deadly SAR CoV-2. The reviewed and theoretical study information strongly suggested that thiazole derivatives can be used for the development of futuristic target drugs against death-causing diseases like SAR-CoV-2.
... Antiviral drug medication has significantly reduced the spread of epidemics, but their continuous usage has led in the emergence of drug resistant mutants over a period of time [31][32][33]. Several antiviral drugs have low oral bioavailability, show adverse side effects and are too expensive [34][35][36]. Hence, there is important necessity to develop safe and effective nonnucleoside antiviral agents. ...
... This compound is found to be more potent than the drug, zileuton (Fig. 10) [6]. Chloro acetylation of 2-(methylthio)-1H-benzo[d]imidazole (34) in presence of NaH and DMF, gives pure product imidazole derivative (35), which undergoes cyclization with thiourea in presence of ethanol resulting in aminothiazole derivatives (36). Compound 36 on alkylation with the equivalent amount of chloroacetone in presence of acetone in anhydrous K2CO3 gives benzo[d]thiazole derivative (37). ...
Article
Thiazoles are notable five-membered heterocyclic rings and their moieties can be found in several biologically active compounds of natural origin, as well as synthetic molecules that possess a wide range of pharmacological activities. Inflammation is the common cause that is associated with different disorders and diseases such as psoriasis, arthritis, infections, asthma, cancer, etc. In this article, the synthesis pattern of these novel molecules are discussed and their anti-inflammatory activities against cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2) and lipoxygenase (LOX) were reviewed and documented. The potent 26 thiazole analogs were validated with molecular docking against main protease (6LU7) and spike binding domain ACE2 receptor (6M0J) to defeat from the COVID-19 infections. Among this, THI-9a showed excellent binding energy and affinity against deadly SAR CoV-2. The reviewed and theoretical study information strongly suggested that thiazole derivatives can be used for the development of futuristic target drugs against death-causing diseases like SAR-CoV-2.
... Many antiviral medicines have significant adverse effects and are economically unfeasible. As a result, the development of new, safe, and efficient antiviral drugs for viral infection treatment is critical [118][119][120][121][122][123]. Furthermore, the dramatic situation created by the unanticipated SARS-CoV-2 outbreak has undoubtedly aroused worries about the possibility of a new influenza pandemic in the future, as well as the need to expand treatment efficacy with newer antiviral drugs [124,125]. ...
Article
Viruses are still the most prevalent infectious pathogens on a worldwide scale, with many of them causing life-threatening illnesses in humans. Influenza viruses, because of their significant morbidity and mortality, continue to pose a major threat to human health. According to WHO statistics, seasonal influenza virus epidemics are predicted to cause over 2 million severe illness cases with high death rates yearly. The whole world has been suffering from the Covid-19 epidemic for two years and is still suffering so far, and the deaths from this virus have exceeded three million cases. Because the great majority of viral infections do not have a specific medication or vaccination, discovering novel medicines remains a vital task. This review covers reports in the patent literature from 1980 to the end of 2021 on the antiviral activities of pyrimidine moieties. The patent database, SciFinder, was used to locate patent applications. A large variety of pyrimidine molecules have been produced and tested for antiviral activity over the last decade. These molecules were reported to inhibit a wide range of viruses, including influenza virus, respiratory syncytial virus, rhinovirus, dengue virus, herpes virus, hepatitis B and C, and human immunodeficiency virus. The cytotoxicity of the developed pyrimidine derivatives was tested in almost all reported studies and the selectivity index was calculated to show the selectivity and safety of such molecules. From the remarkable activity of pyrimidine compounds as antivirals for several dangerous viruses, we expect that these derivatives will be used as potent drugs in the very near future.
... Many antiviral medicines have significant adverse effects and are economically unfeasible. As a result, the development of new, safe, and efficient antiviral drugs for viral infections treatment is critical [7,[105][106][107][108][109]. Furthermore, the dramatic situation created by the unanticipated SARS-CoV-2 outbreak has undoubtedly aroused worries about the possibility of a new influenza pandemic in the future, as well as the need to expand treatment efficacy with newer antiviral drugs [110,111]. ...
Article
Introduction: Several viral infections cause life-threatening consequences in humans, making them the most serious public health concerns. Despite the fact that several antiviral medicines are available on the market, there is no full treatment for many important viral infections. To date, antiviral medicines have significantly reduced the spread of epidemics, but their continued use has resulted in the creation of drug-resistant variants throughout time. As a result, the development of new, safe, and efficient antiviral drugs is critical. Areas covered: This review covered reports in the patent literature in the period 2014 to the first quarter of 2021 on the antiviral activities of thiazole derivatives. These molecules were reported to inhibit a wide range of viruses including influenza viruses, coronaviruses, herpes viruses, hepatitis B and C, bovine viral diarrhea virus, chikungunya virus and human immunodeficiency viruses. Expert opinion: The most bioactive molecules can be used as lead structures for the development of new thiazole compounds with potent and selective antiviral activity. In addition, more efforts are needed to better understand the host-virus interactions for the discovery and development of new therapeutic agents and creative treatment strategies that are supposed to improve rates of clinical cure of the serious viruses.
... To date, antiviral drugs have shown a noteworthy reduction in the spread of epidemics but at the same time, their unremitting usage has resulted in the development of drug-resistant mutants over the time [9][10][11][12][13][14][15][16][17][18]. Many antiviral drugs display pronounced side-effects and are too expensive to be affordable [19][20][21][22][23]. Consequently, the discovery of novel, safe and effective antiviral drug is urgently needed for the treatment of viral infections. ...
Article
Full-text available
Viruses exemplify the most common reason of infectious diseases worldwide and those with prompt propagation and high infection rates cause animal and human pandemics. These rapid-spreading diseases are usually treated with antiviral drugs but, often, drug resistance occurs because of the capability of the pathogens to mutate quickly and become less vulnerable to the treatments. Several compounds have been developed and some of them have exposed good antiviral activity. This review provides an overview on some already known compounds, pointing the attention on the running progresses in identifying and designing novel compounds with outstanding antiviral activity.
Article
Aims: Development of some potent bis-thiazole and bis-thiazine derivatives that could be used as antiviral prototypes. Materials & methods: Xylenyl-spaced bis-carbazone scaffold 3 was used as a versatile building block for bis-thiazole derivatives 6a–e and 9a–d and bis-thiazine derivatives 12a–f. These bis-heterocycles were screened as herpes simplex virus type 1 (HSV-1) inhibitors. Results: The new bis-heterocyclic compounds showed remarkable antiviral activity (e.g., compound 6d cytotoxicity concentration CC50 >500 μg/ml). The antiviral capacity of the synthesized bis-compounds was supported by a molecular docking study against the glycoprotein D receptor of HSV-1. Compounds 6b, 9b, and 12c displayed the best binding coefficients. Conclusion: A new series of xylenyl-spaced bis-carbazone scaffolds were used as a building scaffold to construct a host of bis-thiazole/thiazine derivatives that could be used as antiviral prototypes.
Article
Despite extensive research in the field of drug discovery and development, still there is a need to develop novel molecular entities. Literature reveals a substantial heterocyclic nucleus named, piperazine, which shows an immense therapeutic voyage. For several decades, molecules having the piperazine nucleus have entered the market as a drug exhibiting biological potential. It was known to possess antipsychotic, antihistamine, antianginal, antidepressant, anticancer, antiviral, cardioprotective, and anti-inflammatory activity with a specific basis for structural activity relationship. Thus, it is regarded as a key structural feature in most of the already available therapeutic drugs in the market. Reports also suggest that the extensive utilization of these currently available drugs having a piperazine nucleus shows increasing tolerance significantly day by day. In addition to this, various other factors like solubility, low bioavailability, cost-effectiveness, and imbalance between pharmacokinetics and pharmacodynamics profile limit their utilization. Focusing on that issues, various structural modification studies were performed on the piperazine moiety to develop new derivatives/analogs to overcome the problems associated with available marketed drugs. Thus, this review article aims to gain insight into the number of structural modifications at the N-1 and N-4 positions of the piperazine scaffold. This SAR approach may prove to be the best way to overcome the above-discussed drawbacks and lead to the design of drug molecules with better efficacy and affinity. Hence, there is an urgent need to focus on the structural features of this scaffold which paves further work for deeper exploration and may help medicinal chemists as well as pharmaceutical industries.
Article
Herpesviruses can either cause primary infection or may get reactivated after both hematopoietic cell and solid organ transplantations. In general, viral infections increase post-transplant morbidity and mortality. Prophylactic, preemptive, or therapeutically administered antiviral drugs may be associated with serious side effects and may induce viral resistance. Virus-specific T cells represent a valuable addition to antiviral treatment, with high rates of response and minimal side effects. Even low numbers of virus-specific T cells manufactured by direct selection methods can reconstitute virus-specific immunity after transplantation and control viral replication. Virus-specific T cells belong to the advanced therapy medicinal products, and their production is regulated by appropriate legislation; also, strict safety regulations are required to minimize their side effects.
Article
Full-text available
It remains difficult to treat the multiplicity of distinct viral infections that afflict immunocompromised patients. Adoptive transfer of virus-specific T cells (VSTs) can be safe and effective, but such cells have been complex to prepare and limited in antiviral range. We now demonstrate the feasibility and clinical utility of rapidly generated single-culture VSTs that recognize 12 immunogenic antigens from five viruses (Epstein-Barr virus, adenovirus, cytomegalovirus, BK virus, and human herpesvirus 6) that frequently cause disease in immunocompromised patients. When administered to 11 recipients of allogeneic transplants, 8 of whom had up to four active infections with the targeted viruses, these VSTs proved safe in all subjects and produced an overall 94% virological and clinical response rate that was sustained long-term.
Article
Full-text available
Cytomegalovirus (CMV) infection is a leading cause of illness and death in patients who have undergone allogeneic hematopoietic-cell transplantation. Available treatments are restricted by clinically significant toxic effects and drug resistance. In this phase 2 study, we evaluated the effect of letermovir (also known as AIC246), a new anti-CMV drug with a novel mechanism of action, on the incidence and time to onset of prophylaxis failure in CMV-seropositive recipients of allogeneic hematopoietic-cell transplants from matched related or unrelated donors. From March 2010 through October 2011, we randomly assigned 131 transplant recipients in a 3:1 ratio to three sequential study cohorts according to a double-blind design. Patients received oral letermovir (at a dose of 60, 120, or 240 mg per day, or matching placebo) for 12 weeks after engraftment. The primary end point was all-cause prophylaxis failure, defined as discontinuation of the study drug because of CMV antigen or DNA detection, end-organ disease, or any other cause. Patients underwent weekly surveillance for CMV infection. The reduction in the incidence of all-cause prophylaxis failure was dose-dependent. The incidence of prophylaxis failure with letermovir, as compared with placebo, was 48% versus 64% at a daily letermovir dose of 60 mg (P=0.32), 32% at a dose of 120 mg (P=0.01), and 29% at a dose of 240 mg (P=0.007). Kaplan-Meier time-to-onset profiles for prophylaxis failure showed a significant difference in the comparison of letermovir at a dose of 240 mg per day with placebo (P=0.002). The safety profile of letermovir was similar to placebo, with no indication of hematologic toxicity or nephrotoxicity. Letermovir, as compared with placebo, was effective in reducing the incidence of CMV infection in recipients of allogeneic hematopoietic-cell transplants. The highest dose (240 mg per day) had the greatest anti-CMV activity, with an acceptable safety profile. (Funded by AiCuris; ClinicalTrials.gov number, NCT01063829.).
Article
Full-text available
Letermovir is a novel antiviral compound currently in clinical development for the prevention of human cytomegalovirus (HCMV) infections. In contrast to all currently approved anti-HCMV drugs that target the viral DNA polymerase, letermovir acts via a distinct mode of action involving the viral terminase subunit pUL56. To extend our understanding of potential letermovir resistance mechanisms, we used marker transfer to characterize mutations identified in letermovir-resistant HCMV variants that were selected in cell culture.
Article
Full-text available
The use of available antiviral agents for the prevention of cytomegalovirus (CMV) disease is limited by frequent toxic effects and the emergence of resistance. CMX001 has potent in vitro activity against CMV and other double-stranded DNA viruses. We evaluated the safety and anti-CMV activity of CMX001 in patients who had undergone allogeneic hematopoietic-cell transplantation. From December 2009 through June 2011, a total of 230 patients with data that could be evaluated were enrolled in the study. We randomly assigned these adult CMV-seropositive transplant recipients from 27 centers to oral administration of CMX001 or placebo. Patients were assigned in a 3:1 ratio to five sequential study cohorts according to a dose-escalating, double-blind design. Randomization was stratified according to the presence or absence of acute graft-versus-host disease and CMV DNA in plasma. Patients received the study drug after engraftment for 9 to 11 weeks, until week 13 after transplantation. Polymerase-chain-reaction analysis of CMV DNA in plasma was performed weekly. Patients in whom CMV DNA was detected at a level that required treatment discontinued the study drug and received preemptive treatment against CMV infection. The primary end point was a CMV event, defined as CMV disease or a plasma CMV DNA level greater than 200 copies per milliliter when the study drug was discontinued. The analysis was conducted in the intention-to-treat population. The incidence of CMV events was significantly lower among patients who received CMX001 at a dose of 100 mg twice weekly than among patients who received placebo (10% vs. 37%; risk difference, -27 percentage points; 95% confidence interval, -42 to -12; P=0.002). Diarrhea was the most common adverse event in patients receiving CMX001 at doses of 200 mg weekly or higher and was dose-limiting at 200 mg twice weekly. Myelosuppression and nephrotoxicity were not observed. Treatment with oral CMX001 at a dose of 100 mg twice weekly significantly reduced the incidence of CMV events in recipients of hematopoietic-cell transplants. Diarrhea was dose-limiting in this population at a dose of 200 mg twice weekly. (Funded by Chimerix; CMX001-201 ClinicalTrials.gov number, NCT00942305.).
Article
Full-text available
Antiviral therapy for cytomegalovirus (CMV) plays an important role in the management of solid organ and hematopoietic stem cell transplant recipients. However, CMV antiviral therapy can be complicated by drug resistance associated with mutations in the phosphotransferase UL97 and the DNA polymerase UL54. We have developed an amplicon-based high-throughput sequencing strategy for detecting CMV drug resistance mutations in clinical plasma specimens using a microfluidics PCR platform for multiplexed library preparation and a bench-top next-generation sequencing instrument. Plasmid clones of the UL97 and UL54 genes were used to demonstrate a low overall empiric error rate of the assay (0.189%), and to develop a statistical algorithm for identifying authentic low-abundance variants. The ability of the assay to detect resistance mutations was tested with mixes of wild-type and mutant plasmids, as well as clinical CMV isolates and plasma samples known to contain mutations that confer resistance. Finally, 48 clinical plasma specimens with a range of viral loads (394 - 2,191,011 copies/mL plasma) were sequenced, using multiplexing of up to 24 specimens per run. This led to the identification of seven resistance mutations, three of which were present at < 20% of the sequenced population. Thus this assay offers more sensitive minor variant detection and increased multiplexing relative to current methods for genotypic detection of CMV drug resistance mutations.
Article
Full-text available
Human cytomegalovirus UL97 kinase mutations that commonly confer ganciclovir resistance cluster in different parts of the gene than those conferring resistance to maribavir, an experimental UL97 kinase inhibitor. The drug resistance, growth and autophosphorylation phenotypes of several unusual UL97 mutations in the kinase catalytic domain were characterized. Mutations V466G and P521L, described in clinical specimens from ganciclovir-treated subjects, conferred a UL97 kinase knockout phenotype with no autophosphorylation, a severe growth defect and high-level ganciclovir, cyclopropavir and maribavir resistance, similar to mutations at the catalytic lysine residue K355. Mutations F342S and V356G, observed after propagation under cyclopropavir in vitro, showed much less growth attenuation and moderate to high level resistance to all three drugs while maintaining UL97 autophosphorylation competence and normal cytopathic effect in cell culture, a novel phenotype. F342S is located in the ATP-binding P-loop and is homologous to a c-Abl kinase mutation conferring resistance to imatinib. UL97 mutants with relatively preserved growth fitness and multi-drug resistance are of greater concern in antiviral therapy than the severely growth-impaired UL97 knockout mutants. Current diagnostic genotyping assays are unlikely to detect F342S and V356G and the frequency of their appearance in clinical specimens remains undefined.
Article
Full-text available
Virus-specific T cell (VST) lines could provide useful antiviral prophylaxis and treatment of immune-deficient patients if it were possible to avoid the necessity of generating a separate line for each patient, often on an emergency basis. We prepared a bank of 32 virus-specific lines from individuals with common HLA polymorphisms who were immune to Epstein-Barr virus (EBV), cytomegalovirus, or adenovirus. A total of 18 lines were administered to 50 patients with severe, refractory illness because of infection with one of these viruses after hematopoietic stem cell transplant. The cumulative rates of complete or partial responses at 6 weeks postinfusion were 74.0% (95% CI, 58.5%-89.5%) for the entire group (n = 50), 73.9% (95% CI, 51.2% -96.6%) for cytomegalovirus (n = 23), 77.8% for adenovirus (n = 18), and 66.7% (95% CI, 36.9%-96.5%) for EBV (n = 9). Only 4 responders had a recurrence or progression. There were no immediate infusion-related adverse events, and de novo graft-versus-host disease developed in only 2 patients. Despite the disparity between the lines and their recipients, the mean frequency of VSTs increased significantly postinfusion, coincident with striking decreases in viral DNA and resolution of clinical symptoms. The use of banked third-party VSTs is a feasible and safe approach to rapidly treat severe or intractable viral infections after stem cell transplantation. This study is registered at www.clinicaltrials.gov as NCT00711035.
Article
Full-text available
Despite advances in the prophylaxis and acute treatment of cytomegalovirus (CMV), it remains an important pathogen affecting the short- and long-term clinical outcome of solid organ transplant. The emergence of CMV resistance in a patient reduces the clinical efficacy of antiviral therapy, complicates therapeutic and clinical management decisions, and in some cases results in loss of the allograft and/or death of the patient. There is increasing use of antiviral prophylaxis after transplant with little expansion in the range of antiviral agents effective in treatment of CMV. Further understanding is needed of the risk factors for development of CMV antiviral resistance and of therapeutic strategies for treating patients infected with resistant viruses. We review the current status of CMV resistance in solid organ transplant recipients, and provide diagnostic and therapeutic suggestions for the clinician in managing antiviral resistance.
Article
We examined the rate, clinical impact, and risk factors of cytomegalovirus (CMV) drug resistance in 561 patients who underwent 616 hematopoietic stem cell transplantations (HSCTs) over 5 years. Drug resistance was exclusively identified in haploidentical (haplo)-HSCT recipients receiving preemptive therapy, among whom the rate was 14.5%. Resistance appeared after prolonged treatment (median, 70 days), was associated with higher preceding viral load (P < .001), and was the strongest predictor for disease by multivariate analysis. The high rate of drug resistance as interlinked with severe disease in haplo-HSCT recipients suggests the potential advantage of prophylactic over preemptive treatment in high-risk patients and highlights the need for better-tolerable anti-CMV drugs.