ArticlePDF AvailableLiterature Review

Management of Ganciclovir Resistant Cytomegalovirus Retinitis in a Solid Organ Transplant Recipient: A Review of Current Evidence and Treatment Approaches

Taylor & Francis
Ocular Immunology and Inflammation
Authors:
  • Manchester Univesrsity NHS Foundation Trust, Manchester, United Kingdon

Abstract and Figures

Purpose: Cytomegalovirus retinitis (CMVR) is a serious and potentially sight-threatening infection in immunocompromised individuals. Strategies for the management of drug-resistant CMVR are described. Methods: A case of severe bilateral CMVR in a single lung transplant patient, with UL97 mutation conferring ganciclovir-resistance, is presented. Treatment with standard antiviral agent and adjuvant leflunomide, immunosuppression modifications (calcineurin inhibitors and corticosteroid), intravitreal antiviral therapy and novel use of CMV-immunoglobulin is described. A literature review to support drug-resistant CMVR management is presented. Results: Severe and progressive CMV retinitis was refractory to intravitreal foscarnet and systemic leflunomide. Drug-toxicity restricted systemic antiviral therapy options. The use of combined leflunomide and CMV-immunoglobulins, in the absence of viremia, has not been previously reported. Loss of ganciclovir-resistance was eventually observed permitting successful treatment with systemic and intravitreal ganciclovir. Conclusions: Drug-resistant CMVR is a complex clinical challenge. Multiple systemic and local treatment strategies may be necessary but toxicity, resistance, and co-morbidities may severely restrict available options.
Content may be subject to copyright.
Full Terms & Conditions of access and use can be found at
https://www.tandfonline.com/action/journalInformation?journalCode=ioii20
Ocular Immunology and Inflammation
ISSN: 0927-3948 (Print) 1744-5078 (Online) Journal homepage: https://www.tandfonline.com/loi/ioii20
Management of Ganciclovir Resistant
Cytomegalovirus Retinitis in a Solid Organ
Transplant Recipient: A Review of Current Evidence
and Treatment Approaches
L. Fu, K. Santhanakrishnan, M. Al-Aloul, N. P. Jones & L. R. Steeples
To cite this article: L. Fu, K. Santhanakrishnan, M. Al-Aloul, N. P. Jones & L. R. Steeples (2019):
Management of Ganciclovir Resistant Cytomegalovirus Retinitis in a Solid Organ Transplant
Recipient: A Review of Current Evidence and Treatment Approaches, Ocular Immunology and
Inflammation, DOI: 10.1080/09273948.2019.1645188
To link to this article: https://doi.org/10.1080/09273948.2019.1645188
Published online: 17 Oct 2019.
Submit your article to this journal
View related articles
View Crossmark data
LETTER TO THE EDITOR
Management of Ganciclovir Resistant
Cytomegalovirus Retinitis in a Solid Organ
Transplant Recipient: A Review of Current Evidence
and Treatment Approaches
L. Fu, FRCOphth,
1,2
K. Santhanakrishnan, MRCP,
3
M. Al-Aloul, FRCP,
3
N. P. Jones, FRCOphth,
1,2
and
L. R. Steeples, FRCOphth
1,2
1
Manchester Royal Eye Hospital, Manchester University NHS Foundation Trust, Manchester Academic
Health Sciences Centre, Manchester, UK,
2
Centre for Ophthalmology and Vision Sciences, Faculty of Medical
and Human Sciences, University of Manchester, Manchester, UK, and
3
Department of Cardiothoracic
Transplant, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
ABSTRACT
Purpose: Cytomegalovirus retinitis (CMVR) is a serious and potentially sight-threatening infection in immuno-
compromised individuals. Strategies for the management of drug-resistant CMVR are described.
Methods: A case of severe bilateral CMVR in a single lung transplant patient, with UL97 mutation conferring
ganciclovir-resistance, is presented. Treatment with standard antiviral agent and adjuvant leunomide, immu-
nosuppression modications (calcineurin inhibitors and corticosteroid), intravitreal antiviral therapy and novel
use of CMV-immunoglobulin is described. A literature review to support drug-resistant CMVR management is
presented.
Results: Severe and progressive CMV retinitis was refractory to intravitreal foscarnet and systemic leunomide.
Drug-toxicity restricted systemic antiviral therapy options. The use of combined leunomide and CMV-
immunoglobulins, in the absence of viremia, has not been previously reported. Loss of ganciclovir-resistance
was eventually observed permitting successful treatment with systemic and intravitreal ganciclovir.
Conclusions: Drug-resistant CMVR is a complex clinical challenge. Multiple systemic and local treatment
strategies may be necessary but toxicity, resistance, and co-morbidities may severely restrict available options.
Keywords: Cytomegalovirus (CMV) retinitis, drug-resistance, ganciclovir-resistance, immunoglobulins, UL97
Cytomegalovirus (CMV) is an ubiquitous human
herpes virus and causes signicant morbidity and mor-
tality in immunocompromised hosts.
1
Cytomegalovirus
retinitis (CMVR) is severe and potentially blinding and
was commonly seen in patients with acquired immuno-
deciency syndrome (AIDS) prior to modern highly
active antiretroviral therapy (HAART). Following the
introduction of HAART a dramatic reduction in the
incidence of CMVR in the HIV population was
observed.
2
In the non-HIV immunosuppressed popula-
tion, particularly solid organ transplant (SOT) or hemo-
poietic cell transplant (HCT) recipients, CMV infection
is common and tissue-invasive disease, including
CMVR, is important.
3
Treating viremia and tissue-
invasive disease may be challenging in the context of
systemic immunosuppression. Toxicity and emergence
of drug resistance are major concerns with currently
available antiviral therapies.
4
Current evidence for the management of drug-
resistant CMVR, particularly the use of novel therapies,
is limited and low-grade. Approaches are directed by
evidence for management of non-ocular drug-resistant
disease in transplant patients. We present a clinically
challenging case of ganciclovir (GCV)-resistant CMVR
Received 30 April 2019; revised 9 July 2019; accepted 15 July 2019
Correspondence: L. Fu, Manchester Royal Eye Hospital, Oxford Road, Manchester, M13 9Wl, UK. E-mail: Lana.Fu@mft.nhs.uk
Color versions of one or more of the gures in the article can be found online at www.tandfonline.com/ioii.
Ocular Immunology & Inammation, 2019; 0(0): 17
© 2019 Taylor & Francis Group, LLC
ISSN: 0927-3948 print / 1744-5078 online
DOI: 10.1080/09273948.2019.1645188
1
in a SOT patient. Strategies for management are dis-
cussed including standard and adjuvant systemic anti-
viral agents, local antiviral therapy, modication of
immunosuppression and novel use of CMV immunoglo-
bulins (CMV IgG) for retinitis.
CASE REPORT
A 55-year-old male presented with bilateral CMVR 20
months after right single lung transplant for idio-
pathic pulmonary brosis. He had received standard
immunosuppression with rabbit antithymocyte glo-
bulin (RATG) induction (1.5 mg/Kg for 3 days) then
maintenance therapy with azathioprine, ciclosporin,
and prednisolone (15 mg/day). The CMV status was
both donor and recipient positive (D+/R+) and pro-
phylactic valganciclovir (VGCV) was administered
(450 mg daily) for 3 months.
Surveillance bronchoscopy and biopsy 3 weeks post-
transplant showed mild acute rejection. Intravenous (IV)
methylprednisolone was administered and ciclosporin
was switched to tacrolimus (2 mg twice daily). Three
months later, a further mild rejection episode was suc-
cessfully treated with IV methylprednisolone. Nine
months post-transplant, CMV viremia was detected for
the rst time (log 4.89). Induction therapy with IV GCV
followed by oral VGCV achieved reduction in viral load
to undetectable levels. However, 3 weeks later rising
CMV viral load was observed (log 3.46) and complicated
by CMV pneumonitis. Resistance testing identied the
UL97 C670Y mutation, conferring resistance to GCV;
UL54 testing was negative. Intravenous foscarnet (FOS)
therapy replaced GCV but severe nephrotoxicity devel-
oped and it was discontinued. Systemic leunomide and
weekly CMV immunoglobulin (CMV-IG, Cytotec®)
therapy achieved disease control with recovery of pul-
monary function and eventual resolution of viremia (18
months post transplant). Leunomide was continued as
long-term antiviral therapy alongside oral tacrolimus
(2 mg twice daily) and prednisolone (5 mg daily).
Two months after resolution of viremia (20 months
post transplant) the patient presented with oaters
and reduced vision and was referred for tertiary uvei-
tis opinion. His vision was right (R) 1.0 and left (L) 0.6
logMAR. There was bilateral low-grade granuloma-
tous anterior uveitis and mild vitritis. Bilateral per-
ipheral necrotizing retinitis was seen with 4 and 2
clock-hours of retinitis in the R and L eyes, respec-
tively. The patient was pseudophakic. Aqueous sam-
pling was positive for CMV and negative for herpes
simplex, varicella zoster, and syphilis. HIV and syphi-
lis serology were negative. Blood PCR for CMV was
negative. Intravitreal FOS (2.4 mg/0.1 ml) was
initiated in the right then the left eye and repeated
at 12-week intervals.
Despite systemic leunomide, and repeated intra-
vitreal FOS over 15 months (3 injections to the right,
and 16 to the left eye) bilateral persistent and progres-
sive retinitis was observed. Chorioretinal scarring
developed in previously necrotizing areas and cir-
cumferential spread of CMVR was seen (Figure 1).
Systemic immunosuppression was switched from
tacrolimus to sirolimus (1.5 mg daily) for secondary
antiviral properties. During the switch, increased pre-
dnisolone (8 mg daily) was given for 2 months.
Bilateral extension of retinitis to zone 2 was of signi-
cant clinical concern. Serial aqueous sampling demon-
strated persistent high viral load and GCV-resistant
virus (UL97 C607Y mutation positive); UL54 mutation
testing was negative with no evidence of resistance to
FOS. Blood PCR remained negative for CMV.
Systemic FOS was contraindicated due to persistent
renal impairment.
Two-weekly CMVIG (Cytotec®) was adminis-
tered alongside 12 weekly bilateral intravitreal FOS
over 2 months. A partial response was observed: in
the right eye retinitis remained active but progression
was halted; in the left eye reduction in serial aqueous
FIGURE 1. Wide-eld retinal image (Optos ®) right (a) and left (b) eyes demonstrating active necrotizing retinitis, with circumfer-
ential progressive spread and extensive chorioretinal scarring in previously necrotizing areas.
2L. Fu et al.
Ocular Immunology & Inammation
viral load and reduction in the area of retinitis was
observed after 3 weeks of treatment. The right eye
developed rhegmatogenous, macula-off, retinal
detachment with deterioration in vision to 1.0
logMAR. Pars plana vitrectomy, endolaser, and oil
tamponade was successfully performed. Under oil
tamponade retinitis resolved with no disease
recurrence.
Subsequently, left eye retinitis progressed cir-
cumferentially, threatening the macula with sequen-
tial extensive peripheral scarring. (Figure 2)Despite
regular intravitreal FOS (12 weekly), rising left eye
aqueous viral load (peak log 6.4) and persistent
activity was observed. There was no CMV viremia
even after temporary withdrawal of leunomide.
Further UL97 and UL54 mutation analysis testing
was negative, indicating loss of resistance.
Induction therapy, adjusted for renal function, was
recommenced with intravenous GCV (2.5 mg/kg,
daily for 4 weeks) plus weekly CMV-IG (2 mg/kg
for 4 weeks). Weekly left eye intravitreal GCV
(2 mg/0.1 ml) for 4 weeks was given and a rapid
drop in ocular viral load and full resolution of CMV
retinitis was observed. (Figure 3)
Sirolimus and VGCV (450 mg daily) therapy were
continued with no recurrence of retinitis during 12
months follow-up and with stable renal function.
Right eye silicone oil removal and epiretinal mem-
brane peel was performed 6 months post-
detachment repair and the retina has remained
attached. The vision remains 0.4 LogMAR in both
eyes.
DISCUSSION
This case demonstrates the complexities of drug-
resistant CMV retinitis in immunocompromised
patients. In transplant recipients, mandatory immu-
nosuppression for transplant survival restricts oppor-
tunity for immune reconstitution and permits the
possibility of viral reactivation and tissue-invasive
disease. Incomplete suppression of viral replication
risks the eventual development of drug-resistant dis-
ease adding to the challenge of treatment.
5,6
In this
population, co-morbidities are frequent and further
restrict treatment options.
Antiviral resistance can manifest from true viral
resistance secondary to one or more genetic mutations
or as a result of clinical resistancein the absence of
any detectable mutations. The latter is attributable to
host or viral factors. In general, CMV drug-resistance
FIGURE 2. Sequential wide-eld retinal images (Optos ®) of left eye demonstrates persistent and progressive CMV retinitis. CMV
retinitis was refractory to treatment and circumferential disease progression was observed from baseline (a) with retinitis advancing
superiorly and temporally (b, c) and threatening the macula (d). Resultant extensive chorioretinal scarring is observed involving
zones 13.
Management of Ganciclovir CMV Retinitis in SOT 3
© 2019 Taylor & Francis Group, LLC
should be suspected in patients who have received
several weeks of full dose antiviral therapies and have
rising or persistent CMV viral load or tissue
disease.
5,7
Resistance is unusual in the rst 6 weeks
of therapy.
8
Other causes for sub-optimal response
should always be considered and conrmation of
drug resistance is desirable because empirical treat-
ment changes may cause drug toxicity.
5
Drug resistance is caused by mutations either in the
phosphotransferase-gene (UL97) or in the viral poly-
merase gene (UL54). Mutations emerge rst in UL97,
conferring high- or low-grade GCV resistance. Later,
UL54 mutations may emerge with cross-resistance to
GCV-cidofovir (CDV), GCV-FOS, GCV-CDV-FOS, or
FOS alone even without prior exposure to CDV or
FOS.
9
There are many known mutations, with new
mutations detected continuously. Different levels of
resistance are conferred with individual mutations.
Prolonged drug exposure (>3 months) and incom-
plete suppression of viral replication with sub-
therapeutic drug activity (resulting from inadequate
dosing, absorption or prodrug conversion and varia-
tion in drug clearance) are major risk factors for drug
resistance.
5,7,10
Other risks are high peak viral load,
recurrent infection, T-cell depletion, delayed immune
reconstitution and prior drug exposure, although pre-
emptive therapy in HCT has been shown to be
protective.
11
In renal impairment, absorption during
adjusted dosing is uncertain. In our patient, signi-
cant immunosuppression with high-dose corticoster-
oid during transplant rejection episodes and T-cell
inhibition immunomodulation therapy (IMT) were
signicant risks for emergence of drug resistance.
There may be no detectable CMV in blood or other
tissue at the time of presentation with CMVR. Active
ocular disease, including persistent or progressive reti-
nitis, with persistent or rising intraocular viral load,
may be the only indicator of drug resistance. This high-
lights the utility of ocular sampling for PCR, further to
diagnostic purposes, for serial quantication of viral
load and/or resistance testing. However, the small
volume typically obtained (~0.1 ml from the anterior
chamber) may restrict the number of tests and clinical
priority must be considered. Antivirals in high concen-
tration interferes with PCR analysis, and sampling
should always be performed prior to intravitreal
injection.
Several challenging factors and unusual virus beha-
viors were evident in this case. Firstly, the patient
rapidly developed true GCV resistance following short
exposure to a high GCV dose. This was deemed highly
atypical by transplant physicians. Concurrent immuno-
suppression undoubtedly accelerated the development
of resistance. Secondly, eye disease was diagnosed and
persisted when there was no systemic viremia and other
end-organ disease was controlled, indicating seques-
tered intraocular virus. Thirdly, the detected UL97
mutation conferred resistance to ganciclovir but not
FOS. UL54 mutation testing was repeatedly negative,
with no evidence of foscarnet resistance. However,
CMVR remained refractory to intensive intravitreal
FOS suggesting additional clinical resistance. Systemic
leunomide therapy was ongoing during refractory
ocular disease and prevented viremia; however,
whether the dose was achieving sufcient intraocular
concentration to limit viral replication is uncertain.
Finally and exceptionally, after months of treatment,
loss of detectable resistance mutations was observed
and permitted re-introduction of systemic and local
GCV therapy with complete clinical resolution. This
may be explained by heterogeneous intraocular virus
populations with mixed ganciclovir sensitivities.
The outcome of CMVR is inuenced by several
factors including immune function, co-morbidities,
toxicity, virulence, previous therapy and the develop-
ment of drug resistance. The site, extent and respon-
siveness of CMVR alongside complications determine
long-term visual outcomes, with high risk of signi-
cant vision loss. Complications can be severe and
include retinal detachment (up to one third of eyes),
FIGURE 3. Wide-eld retinal images (Optos ®) left eye before (a) and (b) 4 weeks post induction therapy with systemic ganciclovir
and intravitreal ganciclovir plus CMV-IG after identication of loss of resistance. Complete resolution of CMV retinitis was observed.
4L. Fu et al.
Ocular Immunology & Inammation
macular edema and ischemia, optic atrophy, glau-
coma, and cataract.
12
Treatment
Standard Antiviral Therapy
Systemic Therapy. There are four established anti-
viral drugs for CMV infection: ganciclovir, valganci-
clovir, foscarnet, and cidofovir (CDV); all target DNA
polymerase. Oral or intravenous GCV is standard
rst-line therapy. In patients with renal impairment
or impaired absorption, IV therapy is favored.
9
All
available antivirals are associated with signicant
toxicity and emergence of drug resistance. Foscarnet
is second-line for resistant or refractory disease. If
UL97 resistance is diagnosed during GCV or VGCV
therapy the principal approach is to switch to IV FOS
and reduce systemic immunosuppression if possible.
Alternatively, for mutations conferring low-level GCV
resistance, dose escalation or combined GCV/FOS,
with monitoring for hematological toxicity, is an alter-
native approach.
12
Systemic adjuvant therapy may be
used alongside FOS, particularly leunomide. Use of
cidofovir is limited by toxicity, particularly severe
nephrotoxicity and especially with concurrent calci-
neurin inhibitor therapy.
9
If there is inadequate clin-
ical response or toxicity with FOS, one or more
investigational medicines must be considered.
5
UL54
mutation resistance is treated according to cross-
resistance, with FOS, CDV and GCV used according
to sensitivity and tolerance. Drug level monitoring is
encouraged, particularly in renal-adjusted dosing.
13
Intravitreal Therapy. Currently available treatments
are foscarnet (2.4 mg/0.1 ml) and ganciclovir (12mg/
0.1 ml). Foscarnet is used rst-line in our service with
reported lower risk of retinal toxicity.
14
Intensive induc-
tion therapy once- to twice-weekly is used in addition to
systemic therapy. In refractory disease, switching
between the two agents should be considered, with test-
ing for resistance. As seen in our case, local treatment
may need to be intense and prolonged to halt disease
progression where standard systemic treatment options
are severely restricted by toxicity and resistance.
Adjuvant Therapies
Leunomide. Leunomide, an immunosuppres-
sive drug approved for use in rheumatoid arthritis,
has in-vivo coincident anti-CMV activity, either
alone or in combination in CMV treatment and sec-
ondary prophylaxis.
9,15
It has variable efcacy in
multi-drug resistant and refractory disease.
9,1416
It
may also interfere with CMV virion assembly.
15,16
There are few reports of its use in CMVR. Rifkin
et al. reported successful control of CMVR with
adjuvant leunomide alongside other antiviral
therapy in two SOT recipients with UL-97
mutation.
17
In a further case report of leunomide
in multi-drug-resistant CMVR the authors reported
detection of the drug in vitreous samples.
18
However, the ocular penetration and necessary
intra-ocular therapeutic concentration is unknown.
Toxicity can occur including abnormal liver
enzymes and impaired bone marrow function.
7
Immune Function
Reconstitution of immune function and host defenses is
an important aspect of the management of CMV infec-
tion. Return of CMV-specic T-cell immunity results in
control of and/or cessation of infection recurrences.
Consideration should be given to reducing immuno-
suppression where feasible. Adjusting immunosup-
pression to a mammalian Target Of Rapamycin
(mTOR) inhibitor may have a particular role.
mTOR Inhibitors. Cytomegalovirus uses the mTOR
pathway for viral replication. The mTOR inhibitors sir-
olimus and everolimus, used primarily as immunomo-
dulatory (IMT) agents, have been associated with
reduced incidence of CMV infections in transplant
recipients.
19
Switching is established in guidelines for
management of non-ocular drug-resistant CMV where
possible and requires expert physician management.
20
Temporary increases in systemic corticosteroid dose
may be necessary during switches in IMT agents, with
potential for increased viral replication/activity.
Adoptive Immunotherapy. With demonstration of the
importance of T-cell immune function, use of T-cell
adoptive treatment is of interest and described as
benecial.
21
There is however no current evidence
for use in CMV retinitis.
CMV Intravenous Immune Immunoglobulin (CMV-IVIG).
IVIG is FDA-approved for prophylaxis of CMV disease
and is used as rescue treatment, alongside antivirals, in
refractory CMV disease.
22
A role in combination with
standard antivirals is uncertain with no evidence for
superiority of combined use with GCV/VGCV versus
appropriate antiviral therapy alone.
23
In addition, renal-
adjusted dosing is necessary. Specically for CMVR dos-
ing, ocular penetration and clinical efcacy are uncertain.
Our case is the rst documented use of Cytotec® IVIG in
the absence of systemic viremia to treat ocular tissue
disease. Overall there is a lack of evidence for use in
CMVR.
Novel Therapies
Letermovir. Letermovir is a novel CMV-terminase
inhibitor, blocking cleaving and packaging of viral
DNA. It is FDA approved for prophylaxis in HCT reci-
pients. It maintains high potency against GCV, FOS,
and CDV-resistant CMV and may be promising in
Management of Ganciclovir CMV Retinitis in SOT 5
© 2019 Taylor & Francis Group, LLC
multi-drug-resistant CMV disease.
7,24
However, there is
in-vitro evidence of rapid emergence of resistance and
recent experience of in-vivo resistance. This included
off-label use in a lung-transplant recipient conferring
letermovir resistance primarily mapping to the UL56
component of the terminase complex.
24,25
In clinical
trials, a favorable side-effect prole was reported with
no nephrotoxicity or myelosuppression.
11,26
Arolein
treatment of CMVR is not established.
Maribavir and Brincidofovir. Brincidofovir, an oral
lipid conjugate of CDV, and maribavir, an oral benzi-
midazole L-riboside that inhibits the UL97 viral pro-
tein kinase of CMV, are new treatments with potential
in the prophylaxis of CMV and treatment of refrac-
tory/resistant CMV.
9
Brincidofovir is more potent
and less nephrotoxic than CDV but associated with
increased gastrointestinal toxicity.
27,28
There is no evi-
dence of use of either agent in CMV retinitis.
CONCLUSION
Treatment of drug-resistant CMV retinitis can be extre-
mely challenging and must consider host factors,
including immune function and organ toxicity and
virus factors particularly clinical and genetic resistance.
Ganciclovir resistance and previous FOS toxicity pre-
vented use of standard systemic antivirals, and the risk
of renal failure with cidofovir or foscarnet was deemed
unacceptable. Immunomodulation was switched to an
mTOR inhibition regime with lowest possible mainte-
nance corticosteroid dose. With limited therapeutic
options, a strategy of virustatic therapy (systemic leu-
nomide and intravitreal FOS (>50 total injections in
eyes) was used in parallel with Cytotec® IVIG to ame-
liorate tissue invasive disease. The response was limited
with partial efcacy. Identication of loss of resistance
provided a fortuitous opportunity for a major therapeu-
tic switch with an excellent outcome. Further research
on novel therapies with different antiviral mechanisms
in drug-resistant CMV retinitis is necessary.
DECLARATION OF INTEREST
The authors report no conicts of interest. The authors
alone are responsible for the content and writing of the
paper.
ORCID
L. Fu http://orcid.org/0000-0003-1959-9749
REFERENCES
1. Port AD, Orlin A, Kiss S, et al. Cytomegalovirus retinitis: a
review. JOculPharmacolTher.2017;33:224234. doi:10.1089/
jop.2016.0140.
2. Kedhar SR, Jabs DA. Cytomegalovirus retinitis in the era of
highly active antiretroviral therapy. Herpes-Cambridge.
2007;14:66.
3. Limaye A. Ganciclovir-resistant cytomegalovirus in organ
transplant recipients. Clin Infect Dis.2002;35(7):866872.
doi:10.1086/342385.
4. Chemaly R, Chou S, Einsele H, et al. Denitions of resis-
tant and refractory cytomegalovirus infection and disease
in transplant recipients for use in clinical trials. Clin Infect
Dis.2018;68(8):14201426. doi:10.1093/cid/ciy696.
5. Chou S. Approach to drug-resistant cytomegalovirus in
transplant recipients. Curr Opin Infect Dis.2015;28:293.
doi:10.1097/QCO.0000000000000170.
6. Schneider EW, Elner SG, van Kuijk FJ, et al. Chronic retinal
necrosis: cytomegalovirus necrotizing retinitis associated
with panretinal vasculopathy in non-HIV patients. Retina.
2013;33:17911799. doi:10.1097/IAE.0b013e318285f486.
7. El Chaer F, Shah DP, Chemaly RF. How I treat resistant
cytomegalovirus infection in hematopoietic cell transplan-
tation recipients. Blood.2016;128:26242636. doi:10.1182/
blood-2016-06-688432.
8. Lurain NS, Chou S. Antiviral drug resistance of human
cytomegalovirus. Clin Microbiol Rev.2010;23(4):689712.
doi:10.1128/CMR.00009-10.
9. Koval CE. Prevention and treatment of cytomegalovirus
infections in solid organ transplant recipients. Infect Dis
Clinics.2018;32:581597. doi:10.1016/j.idc.2018.04.008.
10. Nichols WG, Corey L, Gooley T, et al. Rising pp65 anti-
genemia during preemptive anticytomegalovirus therapy
after allogeneic hematopoietic stem cell transplantation:
risk factors, correlation with DNA load, and outcomes.
Blood.2001;97:867874. doi:10.1182/blood.v97.4.867.
11. Marty FM, Ljungman P, Chemaly RF, et al. Letermovir
prophylaxis for cytomegalovirus in hematopoietic-cell
transplantation. N Engl J Med.2017;377:24332444.
doi:10.1056/NEJMoa1706640.
12. Gracia-Ahunger I, Gutiérrez-Aroca J, Cordero E, et al. Use of
high-dose ganciclovir for the treatment of cytomegalovirus
replication in solid organ transplant patients with ganciclovir
resistance-inducing mutations. Transplantation.2013;
95:10151020. doi:10.1097/TP.0b013e31828555ac.
13. Bedino G, Esposito P, Bosio F, et al. The role of therapeutic
drug monitoring in the treatment of cytomegalovirus dis-
ease in kidney transplantation. Int Urol Nephrol.
2013;45:18091813. doi:10.1007/s11255-012-0293-y.
14. Wong R, Pavesio CE, Laidlaw DA, et al. Acute retinal
necrosis: the effects of intravitreal foscarnet and virus
type on outcome. Ophthalmology.2010;117:556560.
doi:10.1016/j.ophtha.2009.08.003.
15. Waldman WJ, Knight DA, Lurain NS, et al. Novel mechan-
ism of inhibition of cytomegalovirus by the experimental
immunosuppressive agent leunomide1, 2. Transplantation.
1999;68:814825. doi:10.1097/00007890-199909270-00014.
16. Waldman WJ, Knight DA, Blinder L, et al. Inhibition of
cytomegalovirus in vitro and in vivo by the experimental
immunosuppressive agent leunomide. Intervirology.
1999;42:412418. doi:10.1159/000053979.
17. Rifkin LM, Minkus CL, Pursell K, et al. Utility of leuno-
mide in the treatment of drug resistant cytomegalovirus
retinitis. Ocul Immunol Inamm.2017;25:9396. doi:10.3109/
09273948.2015.1071406.
6L. Fu et al.
Ocular Immunology & Inammation
18. Dunn JH, Weinberg A, Chan LK, et al. Long-term suppres-
sion of multidrug-resistant cytomegalovirus retinitis with
systemically administered leunomide. JAMA Ophthalmol.
2013;131:958960. doi:10.1001/jamaophthalmol.2013.1589.
19. Nashan B, Gaston R, Emery V, et al. Review of cytomega-
lovirus infection ndings with mammalian target of rapa-
mycin inhibitor-based immunosuppressive therapy in de
novo renal transplant recipients. Transplantation.
2012;93:10751085. doi:10.1097/TP.0b013e31824810e6.
20. Torre-Cisneros J, Aguado JM, Caston JJ, et al. Management of
cytomegalovirus infection in solid organ transplant recipients:
SET/GESITRA-SEIMC/REIPI recommendations. Transplant
Rev.2016;30:119143. doi:10.1016/j.trre.2016.04.001.
21. 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 Transl
Med.2014;25:242ra83. doi:10.1126/scitranslmed.3008825.
22 . Schulz U, Solidoro P, Müller V, et al. CMV immunoglobulins for the
treatment of CMV infections in thoracic transplant recipients.
Transplantation.2016;100:S5S10. doi:10.1097/TP.0000000000001097.
23. Razonable R, Humar A. Cytomegalovirus in solid organ
transplant recipientsguidelines of the American Society
of Transplantation Infectious Diseases Community of
Practice. Clin Transplant.2019;e13512. doi:10.1111/
ctr.13512.
24. Chou S. A third component of the human cytomegalovirus
terminase complex is involved in letermovir resistance.
Antiviral Res.2017;148:14. doi:10.1016/j.antiviral.2017.10.019.
25. Cherrier L, Nasar A, Goodlet KJ, et al. Emergence of leter-
movir resistance in a lung transplant recipient with ganci-
clovir-resistant cytomegalovirus infection. Am J Transplant.
2018;18:30603064. doi:10.1111/ajt.15135.
26. Chemaly RF, Ullmann AJ, Stoelben S, et al. Letermovir for
cytomegalovirus prophylaxis in hematopoietic-cell
transplantation. N Engl J Med.2014;370:17811789.
doi:10.1056/NEJMoa1309533.
27. Hostetler KY. Alkoxyalkyl prodrugs of acyclic nucleoside
phosphonates enhance oral antiviral activity and reduce
toxicity: current state of the art. Antiviral Res.2009;82:
A8498.
28. Painter W, Robertson A, Trost LC, et al. First pharmaco-
kinetic and safety study in humans of the novel lipid
antiviral conjugate CMX001, a broad-spectrum oral drug
active against double-stranded DNA viruses. Antimicrob
Agents Chemother.2012;56:27262734. doi:10.1128/
AAC.05983-11.
Management of Ganciclovir CMV Retinitis in SOT 7
© 2019 Taylor & Francis Group, LLC
... The most severe manifestation of CMV infection in the eye, is CMV retinitis; uncontrolled virus replication in the retina may indeed lead to cell death, retinal detachment, blurred vision and ultimately blindness (2). Prior to highly active antiretroviral therapy, CMV retinitis was commonly observed in patients with acquired immunodeficiency syndrome, and to a lesser extent, in other conditions associated with deficient T-cell response such as solid organ transplantation (3,14,15), hematopoietic stem cell and bone marrow transplantation (16-18). ...
Article
Full-text available
In immunocompetent individuals, cytomegalovirus (CMV) infection is usually mild but may cause severe complications such as retinitis, pneumonitis, and encephalitis in immunocompromised individuals. So far, cases of CMV retinitis in patients with medulloblastoma undergoing chemotherapy and radiotherapy, have not been reported. We herein report the case of a pediatric patient with high-risk medulloblastoma who experienced an unexpected CMV retinopathy and leukoencephalopathy following high dose thiotepa and proton irradiation. The patient underwent a four-course induction therapy (1st cycle: methotrexate and vinorelbine; 2nd cycle: etoposide and hematopoietic stem cells apheresis; 3rd cycle: cyclophosphamide and vinorelbine; 4th cycle: carboplatin and vinorelbine) and then a consolidation phase consisting in high dose thiotepa followed by autologous HSC transplant and proton cranio-spinal irradiation plus boost to the primary tumor site and pituitary site with concomitant vinorelbine. After two months of maintenance treatment with lomustine and vinorelbine, the patient showed complete blindness and leukoencephalopathy. A diagnosis of CMV retinopathy was made and oral valganciclovir was administered. CMV retinopathy was judged to be possibly related to the use of high dose thiotepa worsened by radiotherapy. This case report suggests that in pediatric patients undergoing immunosuppressive chemo-radiotherapy, CMV reactivation should be carefully monitored to prevent serious complications such as retinopathy and visual loss.
... While the role of IVIG in severe suppurative keratitis in humans is unclear, a study on a mouse ocular infection model demonstrated that treating epithelial herpes simplex keratitis with topical immunoglobulins attenuates the spread and incidence rate of HSV-1 [42]. In a recent study, IVIG was used as rescue treatment in a ganciclovir-resistant cytomegalovirus retinitis [43]. In our study, the two cases (patients 1 and 2) who received weekly IVIG (5 mg) for 2 weeks, in addition to antivirals and steroids, showed significant improvement within 3-4 weeks. ...
Article
Full-text available
IntroductionSevere viral keratitis with hypopyon and retrocorneal plaques is easily misdiagnosed as it mimics fungal or bacterial keratitis and is more likely to undergo emergency therapeutic penetrating keratoplasty (TPK) in the presence of active infection, resulting in poor outcomes. This case series offers some important insights for the management strategy of severe viral keratitis.Methods This retrospective case series involved five patients with unilateral severe infectious keratitis with hypopyon over 3 mm and retrocorneal endothelial plaques. Testing for corneal sensation, microscopy, culture, and anterior segment optical coherence tomography (ASOCT) were performed.ResultsAt presentation, all five cases had visual acuity of counting fingers (CF) or worse, large centrally located ulcer with clean ulcer surface, hypopyon, and retrocorneal plaques with reduced corneal sensation. ASOCT demonstrated the presence of retrocorneal plaques with clear space between corneal endothelium and plaques. All cases received systemic and topical ganciclovir and topical steroids. Two patients received intravenous immunoglobulin (IVIG) weekly for 2 weeks. Complete resolution was achieved in all cases except one patient who underwent TPK because of diffuse anterior synechiae of the iris, with recurrence of infection after 10 days. The patients who received IVIG showed resolution earlier compared to the other patients.Conclusions Evaluation of characteristics of retrocorneal plaques by ASOCT and reduced corneal sensation are valuable aids in diagnosis of complicated viral keratitis. Conservative medical management may be a viable option even in severe cases. IVIG may have a role in speedy resolution of severe cases, but more research is needed to confirm this.
... Management of patients with ganciclovir resistant CMV is recommended with high dose ganciclovir, foscarnet, letermovir, leflunomide, and CMV-immunoglobulin. [8] Financial support and sponsorship Nil. ...
Article
Cytomegalovirus (CMV) poses a significant threat to solid organ transplant recipients (SOTR). The incidence of CMV disease in SOTR varies according to immunosuppressive therapy, antiviral prophylaxis, donor and recipient serologic compatibility, and the transplanted organ: 9% to 23%, 22% to 29% and 8% to 32% after heart, liver and kidney transplant, respectively. CMV retinitis (CMVR) is a rare manifestation of CMV with a high risk of blindness. Infection may vary in severity, from initially clinically silent cases to full-blown advanced changes involving the eye. The most characteristic effects are changes in the retina, which usually begin at the retina's periphery and are asymptomatic, then these changes spread toward the center as the disease progresses and impairs vision. We describe CMV vitritis and retinitis in a 74-year-old patient after heart transplantation conducted in 1992. The first symptom of the disease was low vision in the left eye. Initially no blood viremia was observed; then the CMV viral load in the blood and vitreous body of the right eye was 2454 and 26 million IU/mL.Despite the initiation of treatment (intravitreal and then intravenous ganciclovir), the inflammatory process progressed rapidly and vision in the left eye was lost, although functional visual acuity in the right eye was maintained. Systemic antiviral therapy with intravenous ganciclovir lasted 6 weeks until the eradication of CMV viremia. The patient was on prophylactic therapy with oral valganciclovir for 12 months. A clinically silent course of CMVR delays diagnosis and therapy. Therefore, it is recommended that all SOTR undergo periodic ophthalmologic control to avoid delayed diagnosis.
Article
Hosts with compromised or naive immune systems, such as individuals living with HIV/AIDS, transplant recipients, and fetuses, are at the highest risk for complications from cytomegalovirus (CMV) infection. Despite substantial progress in prevention, diagnostics, and treatment, CMV continues to negatively impact both solid-organ transplant (SOT) and hematologic cell transplant (HCT) recipients. In this article, we summarize important developments in the field over the past 10 years and highlight new approaches and remaining challenges to the optimal control of CMV infection and disease in transplant settings.
Article
Full-text available
Following a year of valganciclovir prophylaxis, a lung transplant recipient developed cytomegalovirus (CMV) infection that became resistant to ganciclovir, as confirmed by detection of UL97 kinase mutation M460V and a previously uncharacterized UL54 DNA polymerase mutation L516P. The latter mutation is now shown to confer ganciclovir and cidofovir resistance. As predicted from the viral genotype, foscarnet therapy was effective, but resumption of valganciclovir as secondary prophylaxis resulted in a plasma viral load rebound to 3.6 log10 copies/mL several weeks later. Valganciclovir was then replaced by letermovir, resulting in gradual viral load reduction in the first 5 weeks to below the quantitation limit (2.7 log10 copies/mL) for one week, followed by 10 weeks of rising viral loads reaching 4.3 log10 copies/mL while on letermovir. At this point, CMV genotypic testing revealed UL56 mutation C325Y, which confers absolute resistance to letermovir. Retreatment with foscarnet was successful. This case adds to the considerable list of proven ganciclovir resistance mutations, and provides an early experience with letermovir resistance after off‐label therapeutic use. This experience is consistent with in vitro observations of rapid emergence of letermovir‐resistant CMV after drug exposure. This article is protected by copyright. All rights reserved.
Article
Full-text available
Intravenous ganciclovir and, increasingly, oral valganciclovir are now considered the mainstay of treatment for cytomegalovirus (CMV) infection or CMV disease. Under certain circumstances, CMV immunoglobulin (CMVIG) may be an appropriate addition or, indeed, alternative. Data on monotherapy with CMVIG are limited, but encouraging, for example in cases of ganciclovir intolerance. In cases of recurrent CMV in thoracic transplant patients after a disease- and drug-free period, adjunctive CMVIG can be considered in patients with hypogammaglobulinemia. Antiviral-resistant CMV, which is more common among thoracic organ recipients than in other types of transplant, can be an indication for introduction of CMVIG, particularly in view of the toxicity associated with other options, such as foscarnet. Due to a lack of controlled trials, decision-making is based on clinical experience. In the absence of a robust evidence base, it seems reasonable to consider the use of CMVIG to treat CMV in adult or pediatric thoracic transplant patients with ganciclovir-resistant infection, or in serious or complicated cases. The latter can potentially include (i) treatment of severe clinical manifestations, such as pneumonitis or eye complications; (ii) patients with a positive biopsy in end organs, such as the lung or stomach; (iii) symptomatic cases with rising polymerase chain reaction values (for example, higher than 5.0 log10) despite antiviral treatment; (iv) CMV disease or CMV infection or risk factors, such as CMV-IgG-negative serostatus; (vi) ganciclovir intolerance; (vii) patients with hypogammaglobulinemia.
Article
Cytomegalovirus is one of the most common opportunistic infections that affect the outcome of solid organ transplantation. This updated guideline from the American Society of Transplantation Infectious Diseases Community of Practice provides evidence‐based and expert recommendations for screening, diagnosis, prevention and treatment of CMV in solid organ transplant recipients. CMV serology to detect immunoglobulin G remains as the standard method for pre‐transplant screening of donors and transplant candidates. Antiviral prophylaxis and pre‐emptive therapy are the mainstays of CMV prevention. The lack of a widely‐applicable viral load threshold for diagnosis and preemptive therapy is highlighted, as a result of variability of CMV nucleic acid testing, even in the contemporary era when calibrators are standardized. Valganciclovir and intravenous ganciclovir remain as drugs of choice for CMV management. Strategies for managing drug‐resistant CMV infection are presented. There is increasing use of CMV‐specific cell‐mediated immune assays to stratify the risk of CMV infection after solid organ transplantation, but their role in optimizing CMV prevention and treatment efforts have yet to be demonstrated. Specific issues related to pediatric transplant recipients are discussed. This article is protected by copyright. All rights reserved.
Article
Despite advances in prevention and treatment, cytomegalovirus (CMV) infection and disease remain an expected problem in solid organ transplant recipients. Because of the effect of immunosuppressing medications, CMV primary, secondary, and reactivated infection requires antiviral medications to prevent serious direct and indirect effects of the virus. Side effects and drug resistance, however, often limit the capacity of traditional antiviral therapies. This article updates the clinician on current and promising approaches to the management and control of CMV in the solid organ transplant recipient.
Article
Despite advances in preventive strategies, cytomegalovirus (CMV) infection remains a major complication in solid organ and hematopoietic cell transplant recipients. CMV infection may fail to respond to commercially available antiviral therapies, with or without demonstrating genotypic mutation(s) known to be associated with resistance to these therapies. This lack of response has been termed "resistant/refractory CMV" and is a key focus of clinical trials of some investigational antiviral agents. In order to provide consistent criteria for future clinical trials and outcomes research, the CMV Resistance Working Group of the CMV Drug Development Forum (consisting of scientists, clinicians, regulatory officials, and industry representatives from the US, Canada, and Europe) has undertaken establishing standardized consensus definitions of "resistant" and "refractory" CMV. These definitions have emerged from the Working Group's review of the available virologic and clinical literature and will be subject to reassessment and modification based on results of future studies.
Article
Background Cytomegalovirus (CMV) infection remains a common complication after allogeneic hematopoietic-cell transplantation. Letermovir is an antiviral drug that inhibits the CMV–terminase complex. Methods In this phase 3, double-blind trial, we randomly assigned CMV-seropositive transplant recipients, 18 years of age or older, in a 2:1 ratio to receive letermovir or placebo, administered orally or intravenously, through week 14 after transplantation; randomization was stratified according to trial site and CMV disease risk. Letermovir was administered at a dose of 480 mg per day (or 240 mg per day in patients taking cyclosporine). Patients in whom clinically significant CMV infection (CMV disease or CMV viremia leading to preemptive treatment) developed discontinued the trial regimen and received anti-CMV treatment. The primary end point was the proportion of patients, among patients without detectable CMV DNA at randomization, who had clinically significant CMV infection through week 24 after transplantation. Patients who discontinued the trial or had missing end-point data at week 24 were imputed as having a primary end-point event. Patients were followed through week 48 after transplantation. Results From June 2014 to March 2016, a total of 565 patients underwent randomization and received letermovir or placebo beginning a median of 9 days after transplantation. Among 495 patients with undetectable CMV DNA at randomization, fewer patients in the letermovir group than in the placebo group had clinically significant CMV infection or were imputed as having a primary end-point event by week 24 after transplantation (122 of 325 patients [37.5%] vs. 103 of 170 [60.6%], P<0.001). The frequency and severity of adverse events were similar in the two groups overall. Vomiting was reported in 18.5% of the patients who received letermovir and in 13.5% of those who received placebo; edema in 14.5% and 9.4%, respectively; and atrial fibrillation or flutter in 4.6% and 1.0%, respectively. The rates of myelotoxic and nephrotoxic events were similar in the letermovir group and the placebo group. All-cause mortality at week 48 after transplantation was 20.9% among letermovir recipients and 25.5% among placebo recipients. Conclusions Letermovir prophylaxis resulted in a significantly lower risk of clinically significant CMV infection than placebo. Adverse events with letermovir were mainly of low grade. (Funded by Merck; ClinicalTrials.gov number, NCT02137772; EudraCT number, 2013-003831-31.)
Article
Letermovir is a human cytomegalovirus (CMV) terminase inhibitor that was clinically effective in a Phase III prevention trial. In vitro studies have shown that viral mutations conferring letermovir resistance map primarily to the UL56 component of the terminase complex and uncommonly to UL89. After serial culture of a baseline CMV laboratory strain under letermovir, mutation was observed in a third terminase component in 2 experiments, both resulting in amino acid substitution P91S in gene UL51 and adding to a pre-existing UL56 mutation. Recombinant phenotyping indicated that P91S alone conferred 2.1-fold increased letermovir resistance (EC50) over baseline, and when combined with UL56 mutation S229F or R369M, multiplied the level of resistance conferred by those mutations by 3.5–7.7-fold. Similarly a combination of UL56 mutations S229F, L254F and L257I selected in the same experiment conferred 54-fold increased letermovir EC50 over baseline, but 290-fold when combined with UL51 P91S. The P91S mutant was not perceptibly growth impaired. Although pUL51 is essential for normal function of the terminase complex, its biological significance is not well understood. Letermovir resistance mutations mapping to 3 separate genes, and their multiplier effect on the level of resistance, suggest that the terminase components interactively contribute to the structure of a letermovir antiviral target. The diagnostic importance of the UL51 P91S mutation arises from its potential to augment the letermovir resistance of some UL56 mutations at low fitness cost.
Article
Cytomegalovirus (CMV) is a ubiquitous DNA herpes virus that causes significant morbidity and mortality in immunocompromised individuals. CMV retinitis is a potentially blinding manifestation of CMV infection that was commonly seen in advanced acquired immunodeficiency syndrome (AIDS) in the era before modern combination antiretroviral therapy era, but is also recognized in patients with immune deficiency from multiple causes. The advent of and advances in antiretroviral therapies for human immunodeficiency virus have decreased the incidence of CMV retinitis by over 90% among AIDS patients, and improved visual outcomes in those affected. The diagnosis is generally a clinical one, and treatment modalities include systemic and intravitreal antiviral medications. Retinal detachment and immune recovery uveitis are sight-threatening complications of CMV retinitis that require specific treatments.
Article
Cytomegalovirus (CMV) infection is a significant complication in hematopoietic cell transplantation (HCT) recipients. Four antiviral drugs are used for preventing or treating CMV: ganciclovir, valganciclovir, foscarnet, and cidofovir. With prolonged and repeated use of these drugs, CMV can become resistant to standard therapy, resulting in increased morbidity and mortality, especially in HCT recipients. Antiviral drug resistance should be suspected when CMV viremia (DNAemia or antigenemia) fails to improve or continue to increase after 2 weeks of appropriately dosed and delivered antiviral therapy. CMV resistance is diagnosed by detecting specific genetic mutations. UL97 mutations confer resistance to ganciclovir and valganciclovir, and a UL54 mutation confers multidrug resistance. Risk factors for resistance include prolonged or previous anti-CMV drug exposure or inadequate dosing, absorption, or bioavailability. Host risk factors include type of HCT and degree of immunosuppression. Depending on the genotyping results, multiple strategies can be adopted to treat resistant CMV infections, albeit no randomized clinical trials exist so far, after reducing immunosuppression (if possible): ganciclovir dose escalation, ganciclovir and foscarnet combination, and adjunct therapy such as CMV-specific cytotoxic T-lymphocyte infusions. Novel therapies such as maribavir, brincidofovir, and letermovir should be further studied for treatment of resistant CMV.