Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to...

37
1 Transplantation Publish Ahead of Print DOI: 10.1097/TP.0000000000002893 Impact of CMV reactivation, treatment approaches and immune reconstitution in a nonmyeloablative tolerance induction protocol in Cynomolgus macaques Paula Alonso-Guallart, DVM, 1 Raimon Duran-Struuck, DVM, PhD, 1,2 Jonah S. Zitsman, BS, 1 Stephen Sameroff, BS, 3 Marcus Pereira, MD, MPH, 4 Jeffrey Stern, MD, 1 Erik Berglund, MD, PhD, 1 Nathaly Llore, MD, 1 Genevieve Pierre, BS, 1 Emily Lopes, BS, 1 Sigal B. Kofman, BS, 1 Makenzie Danton, MS, 1 Hugo P. Sondermeijer, MD, 1,5 David Woodland, MD, 1 Yojiro Kato, MD, PhD, 1 Dilrukshi K. Ekanayake-Alper, DVM, PhD, 1 Alina C. Iuga, MD, 6 Cheng-Shie Wuu, PhD, 7 Anette Wu, MD, MPH, 1 W. Ian Lipkin, MD, 3 Rafal Tokarz, PhD, 3 Megan Sykes, MD, 1,8,9 and Adam Griesemer , MD 1,9 1 Columbia Center for Translational Immunology, Department of Medicine, Columbia University Medical Center, New York, NY. 2 Department of Pathobiology, University of Pennsylvania, Philadelphia, PA. 3 Center for Infection and Immunity, Mailman School of Public Health, Columbia University Medical Center, New York, NY. 4 Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, USA. 5 Department of Physiology, Maastricht University, Maastricht, Netherlands. 6 Department of Pathology and Cell Biology, Columbia University, New York, NY, USA 7 Department of Radiation Oncology, Columbia University Medical Center, New York, NY. 8 Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY. ACCEPTED Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Transcript of Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to...

Page 1: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

1

Transplantation Publish Ahead of Print

DOI: 10.1097/TP.0000000000002893

Impact of CMV reactivation, treatment approaches and immune reconstitution in a

nonmyeloablative tolerance induction protocol in Cynomolgus macaques

Paula Alonso-Guallart, DVM,1 Raimon Duran-Struuck, DVM, PhD,

1,2 Jonah S. Zitsman, BS,

1

Stephen Sameroff, BS,3 Marcus Pereira, MD, MPH,

4 Jeffrey Stern, MD,

1 Erik Berglund, MD,

PhD,1 Nathaly Llore, MD,

1 Genevieve Pierre, BS,

1 Emily Lopes, BS,

1 Sigal B. Kofman, BS,

1

Makenzie Danton, MS,1 Hugo P. Sondermeijer, MD,

1,5 David Woodland, MD,

1 Yojiro Kato,

MD, PhD,1 Dilrukshi K. Ekanayake-Alper, DVM, PhD,

1 Alina C. Iuga, MD,

6 Cheng-Shie Wuu,

PhD,7 Anette Wu, MD, MPH,

1 W. Ian Lipkin, MD,

3 Rafal Tokarz, PhD,

3 Megan Sykes, MD,

1,8,9

and Adam Griesemer , MD1,9

1 Columbia Center for Translational Immunology, Department of Medicine, Columbia University

Medical Center, New York, NY.

2 Department of Pathobiology, University of Pennsylvania, Philadelphia, PA.

3 Center for Infection and Immunity, Mailman School of Public Health, Columbia University

Medical Center, New York, NY.

4 Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center,

New York, NY, USA.

5 Department of Physiology, Maastricht University, Maastricht, Netherlands.

6 Department of Pathology and Cell Biology, Columbia University, New York, NY, USA

7 Department of Radiation Oncology, Columbia University Medical Center, New York, NY.

8 Department of Microbiology and Immunology, Columbia University Medical Center, New

York, NY.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 2: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

2

9 Department of Surgery, Columbia University Medical Center, New York, NY.

Corresponding Author: Paula Alonso-Guallart, DVM, Columbia Center for Translational

Immunology, Columbia University Medical Center, 650 West 168th Street, 17-1705, New York,

NY 10032 ([email protected]).

Author Contributions

PAG, RDS, MS and AG performed research, analyzed data and wrote the manuscript. JSZ, JS,

EB, NL, GP, EL, SBK, MD, HPS, DW, YK, DKE and AW performed research, collected

samples and analyzed data. SS, RT and WIL established and performed CMV PCR assays. MP

participated in research and CMV assessment. AI analyzed histology specimens. CSW

performed animal conditioning.

Disclosure of Conflicts of Interest: Authors do not report any conflict of interest pertaining to

the work shown in this manuscript.

Funding: Funding for these studies was provided by NIH grant R01OD017949, by startup funds

from Columbia University Departments of Medicine and Surgery (to MS and RDS), the Banting

Foundation (to MS), the Columbia University core award (to RDS), and the Irving Pilot

Translational science award for new investigators (to RDS). These studies used the resources of

the Herbert Irving Comprehensive Cancer Center Flow Cytometry Shared Resources funded in

part through Center Grant P30CA013696 and the Diabetes and Endocrinology Research Center

Flow Core Facility funded in part through Center Grant 5P30DK063608.

Running title: Impact of CMV and treatment in Cynomolgus macaques

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 3: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

3

Abbreviations:

CMV: Cytomegalovirus

MCM: Mauritius Cynomolgus macaque

BMT: Bone marrow transplant

MHC: Major histocompatibility complex

NHP: Nonhuman primate

CyA: Cyclosporine A

GCV: Ganciclovir

VGC: Valganciclovir

GVHD: Graft-versus-host disease

Treg: Regulatory T cell

TBI: Total body irradiation

TI: Thymic irradiation

ATG: Anti-thymocyte globulin

qPCR: Quantitative polymerase chain reaction

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 4: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

4

Abstract

Background: Cytomegalovirus (CMV) infection is a serious complication in immunosuppressed

patients, specifically transplant recipients. Here, we describe the development and use of an

assay to monitor the incidence and treatment of CMV viremia in a Cynomolgus macaque model

of bone marrow transplantation (BMT) for tolerance induction. We address the correlation

between the course of viremia and immune reconstitution.

Methods: Twenty-one animals received a nonmyeloablative conditioning regimen. Seven

received cyclosporine A (CyA) for 28 days and fourteen received rapamycin. A CMV PCR assay

was developed and run twice per week to monitor viremia. Nineteen recipients were CMV

seropositive before BMT. Immune reconstitution was monitored through flow cytometry and

CMV viremia was tracked via qPCR.

Results: Recipients developed CMV viremia during the first month post-BMT. Two animals

developed uncontrollable CMV disease. CMV reactivation occurred earlier in CyA-treated

animals compared to those receiving rapamycin. Post-BMT, T-cell counts remained significantly

lower compared to pretransplant levels until CMV reactivation, at which point they increased

during the viremic phase and approached pretransplant levels three months post-BMT.

Management of CMV required treatment before viremia reached 10,000 copies/mL, otherwise

clinical symptoms were observed. High doses of ganciclovir resolved the viremia, which could

subsequently be controlled with valganciclovir.

Conclusion: We developed an assay to monitor CMV in Cynomolgus macaques. CMV

reactivation occurred in 100% of seropositive animals in this model. Rapamycin delayed CMV

reactivation and ganciclovir treatment was effective at high doses. As in humans, CD8+ T cells

proliferated during CMV viremia.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 5: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

5

1. Introduction

Cytomegalovirus (CMV) is a -herpesvirus that affects a high percentage (50-90%) of the

human population.1,2

After primary infection, CMV remains latent in immunocompetent

individuals. Clinically significant disease can occur during primary infection in seronegative

individuals or as reactivation of latent infection in immunocompromised individuals, such as

immunosuppressed transplant recipients. CMV is a particularly frequent complication in bone

marrow transplant (BMT) recipients, where treatment with myelosuppressive antiviral drugs is

undesirable during stem cell engraftment.3 CMV may have additional impact on the outcomes of

bone marrow transplant recipients. Cellular immunity through T cell responses is the most

important pathway for controlling CMV replication.4-9

Virus-specific memory T cells have been

associated with bone marrow (BM) rejection or graft-versus-host disease (GVHD) at the time of

CMV infection,10

suggesting that viremia may induce heterologous immunity to the donor or

recipient, respectively. Early detection and control of CMV in BMT patients is therefore crucial

for optimal clinical outcomes.

Our laboratory has utilized Mauritian origin Cynomolgus macaques (MCM) as a translational

model to study the ability to induce transplantation tolerance through long-term mixed

hematopoietic chimerism across major histocompatibility complex (MHC) barriers. Our current

tolerance induction protocol uses a nonmyeloablative preparative regimen along with a short

course (28 days) of cyclosporine A (CyA) or rapamycin monotherapy, BMT and in vitro

expanded polyclonal recipient regulatory T cell (Treg) infusions.11

In our MCM model, as in

humans, we found that CMV primary infection or reactivation is a significant complication after

BMT. We therefore developed a qPCR assay to monitor CMV viremia in our animals.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 6: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

6

Here, we report the results of different treatment approaches for the control of CMV infection in

MCM, the effect of the use of CyA versus rapamycin on CMV reactivation timing and on the

reconstitution of CD4+ and CD8+ T cell numbers and naïve/memory phenotypes in the context

of CMV reactivation and control.

2. Materials and Methods

2.1 Animals

Adult Mauritian origin Cynomolgus macaques were used in this study (Charles River Primates,

Wilmington, MA; Sanofi-Synthelabo, Bridgewater, NJ; Bioculture Group, Glenmoore, PA).

Recipient and donor pairs were selected for ABO compatibility and mismatching of Cynomolgus

leukocyte antigens.12

Cytomegalovirus serology was assessed prior to transplant (VRL

Laboratories). All animals were negative for B virus, simian T-lymphotropic virus, simian

retrovirus, simian immunodeficiency virus, simian varicella virus and malaria. All macaques

were housed at the Institute of Comparative Medicine (Columbia University Medical Center,

New York, NY). This facility holds a current USDA registration, PHS assurance and is

AAALAC accredited. All surgical and experimental procedures were approved by the Columbia

University Institutional Animal Care and Use Committee. All postoperative animals were

continuously monitored by a team of veterinarians and pediatric surgeons, providing

uninterrupted medical care day-and-night.

2.2 Development of Cynomolgus CMV quantitative polymerase chain reaction (qPCR) and

CMV monitoring

A quantitative PCR assay was designed to detect a fragment of the DNA polymerase gene of

Cynomolgus cytomegalovirus (CMV). Using Primer3 software, ten primer pairs were designed

and tested in a gel-based assay. To generate template material for testing, we used the EasyMag

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 7: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

7

extraction platform (Biomerieux, Marcy-l'Étoile, France) to extract DNA from serum and brain

of a Cynomolgus macaque with clinical signs of systemic CMV reactivation. From the initial

tests, we selected one optimal primer pair, consisting of primers CMV-Forward-

GATGGGACCGCTCAAGTTTC, CMV-Reverse-TGACGGTAGCGAGGAGACAA, and

CMV-Probe-(Fam) GGTCGATGGGGTTTTGACTCACGA (Tam). To generate a quantified

standard for the assay, we cloned a PCR product containing the primer and probe binding sites

into the PGem T-easy (Promega, Madison, WI) ligation and vector system. Plasmids were

isolated and purified using the PureLink plasmid miniprep kit (ThermoFisher, Waltham,

MA). The concentration of the standard was quantified, and then serially diluted. These

serial dilutions were used to optimize the qPCR assay using TaqMan Universal PCR

Master Mix (ThermoFisher) and to test the sensitivity of the primers and probe. The

assay had an efficiency of 95% and a sensitivity of >5 copies/4µl of DNA.

For CMV testing, 200µl of serum were extracted on the EasyMag platform and eluted in

40µl of DNA using the Qiagen QIAamp DNA Mini Kit (Qiagen, Hilden, Germany). PCR

reactions were run in triplicate using 4µl of DNA along with serially diluted standards

and a no-template control on a ViiA7 Real-Time PCR machine (Applied Biosystems,

Foster City, CA). Results were analyzed using QuantStudio Real-Time PCR software

(Applied Biosystems). Results of the assay were multiplied by 50 to yield DNA

copies/mL of serum.

2.3 Cell sorting and flow cytometric analysis

Whole blood was lysed and labeled with a combination of the following monoclonal antibodies:

MHC I-PE-Cy7 (G46-2.6, BD Biosciences, San Jose, CA), CD3-PerCP-Cy5.5 (SP34-2, BD

Biosciences), CD4-BV510 (L200, BD Horizon, Franklin Lakes, NJ), CD4-APC (L200, BD

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 8: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

8

Pharmingen, Franklin Lakes, NJ), CD8-APC (RPA-T8, BD Biosciences), CD8-BV421 (RPA-

T8, BD Biosciences), CD95-PE (DX2, BD Biosciences), CD28-Pacific Blue (CD28.2,

BioLegend, San Diego, CA). Samples were acquired using FACSCanto II or LSRFortessa (BD

Bioscience). Samples were analyzed using FCS Express (De Novo Software, Glendale, CA).

CD4+ and CD8+ T cells were defined as CD20-CD3+CD4+ or CD20-CD3+CD8+ cells,

respectively. NK cells were defined as CD20-CD3-CD56+CD8+ cells.13,14

Naïve T cells were

defined as CD28+CD95-. Central and effector memory T cells were defined as CD28+CD95+

and CD28-CD95+, respectively.

2.4 Conditioning regimen

Recipients were conditioned with 2.5-3 Gy of total body irradiation (TBI) (Table 1) administered

on days -6 and -5, 7 Gy of thymic irradiation (TI) on day -1, T-cell depletion with anti-horse

anti-thymocyte globulin (ATG, Pfizer, New York, NY) on days -2, -1 and 0, co-stimulation

blockade with anti-CD40L (NHP Reagent Resource, Boston, MA) (day 0, 2, 5, 7, 9 and 12) and

a short course of immunosuppression post-BMT (either CyA (Novartis, Basel, Switzerland) or

rapamycin (LC Laboratories, Woburn, MA)). BM was administered from a MHC-mismatched

donor ± in vitro expanded autologous Tregs. CyA levels were maintained between 200-400

ng/mL and rapamycin levels between 20-30 ng/mL for 28 days and tapered to 0 thereafter.11

Animals treated with CyA were included in Group A if they received Tregs or Group B if they

did not, and rapamycin-treated animals that received Tregs were included in Group C or Group

D if they did not receive Tregs.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 9: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

9

2.5 Major histocompatibility complex (MHC) genotyping

Comprehensive MHC genotypes were determined by the Genetic Services Unit of the Wisconsin

National Primate Research Center at the University of Wisconsin-Madison

(http://www.primate.wisc.edu/wprc/services/genetics.html). Genomic DNA isolated from whole

blood samples served as templates for PCR with a panel of primers that flank the highly

polymorphic peptide binding domains encoded by exon 2 of MHC class I (Mafa-A, -B, -I, -E)

and class II (Mafa-DRB, -DQA, -DQB, -DPA and -DPB) loci. These PCR products were

generated with Fluidigm Access Arrays that allow all reactions to be multiplexed in a single

experiment. After cleanup and pooling, these products were sequenced on an Illumina MiSeq

instrument and the resulting sequence reads mapped against a custom database of Mauritian

Cynomolgus macaque class I and class II.15

2.6 Statistics

Data were analyzed applying two-tailed paired or unpaired Student’s t-Test using GraphPad

Prism 7.05 (San Diego, CA). P values of ≤0.05 were considered statistically significant (*= p ≤

0.05, **= p ≤ 0.01, ***= p ≤ 0.001, ****= p ≤ 0.0001, NS = Not significant).

3. Results:

3.1 Hematologic effects of the BMT preparative regimen and CMV reactivation

postconditioning

Our MCM transplant tolerance induction protocol across MHC barriers included BMT ± infusion

of in vitro expanded polyclonal recipient Tregs. In addition, prior to the transplant, the animals

received a preparative regimen that included a T cell depleting agent (ATG), TBI and TI in

addition to CyA (n=7) or rapamycin (n=14) for a minimum of 28 days (cf., Materials and

Methods).11

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 10: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

10

Effects of this preparative regimen included transient leukopenia, anemia, and

thrombocytopenia. All hematopoietic cell lineages reached their nadir approximately two weeks

post-TBI (Figure 1A, 1B, 1C). In addition, CD4+ T cell, CD8+ T cell and NK cell counts

decreased significantly compared to the preconditioning levels (Figure 1D,1E, and 1F

respectively) and the recovery of these populations was slower compared to the white blood cell

count (WBC), hematocrit and platelets. We compared the effects of the regimen with

posttransplant CyA vs rapamycin with and without expanded Tregs on hematologic recovery

during the first month post-BMT (Groups A through D). All four groups responded with a

similar pattern, with the exception of Treg-treated animals receiving CyA (Group A), where

anemia and thrombocytopenia were more pronounced than the other groups (Figure 1A-1F and

Table 2).

Twenty of 21 animals that underwent conditioning and BMT developed CMV viremia between

day 0 and 19 in the case of CyA-treated recipients and between day 4 and 30 if they received

rapamycin as posttransplant immunosuppression (Table 1). In some cases, when CMV viremia

exceeded >10,000 copies/mL, clinical signs were observed (lethargy, loss of appetite, tremor).

Two of the CyA recipients were euthanized due to uncontrollable CMV disease before the qPCR

assay was developed and the coordination of antiviral treatment was perfected. Histology from

the first animal, 90-47, showed acute meningitis featuring meningeal neutrophilic infiltrates as

well as cytomegalic cells (Figure 2A) and the cerebral cortex showed scattered neurons with

cytomegalic changes including nuclear and cytoplasmic inclusions, that were positive on CMV

immunostaining (Figure 2B), which correlated with the clinical findings. This animal’s CD4 and

CD8 counts remained under 400 cells/µL until sacrifice (Figure 2C). Due to these outcomes, our

qPCR assay was developed to monitor MCM CMV in the subsequent animals (cf., Materials and

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 11: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

11

Methods) and antiviral treatment was instituted in all animals that developed CMV viremia. The

second animal that developed uncontrollable CMV disease (90-39) was seronegative before the

transplant conditioning. After the recipient developed a primary infection from the donor, whose

serologic testing revealed it converted to CMV+ just prior to donation, CMV copies increased

rapidly, reaching >600,000 copies/mL (Figure 2D). The animal had clinical signs of CMV

disease that included Bell’s palsy. Ganciclovir (GCV) treatment was started at 10 mg/kg BID IV

on day 17, increased to 12.5 mg/kg BID on day 25 and switched later to valganciclovir (VGC)

after a poor response to treatment, but the animal did not recover and was euthanized. The lack

of successful CMV control was perhaps in part due to the low T cell counts from the

conditioning regimen (Figure 2D), the lack of preexisting immunity against CMV and the timing

of antiviral treatment.

3.2 Rapamycin delayed the use of antivirals post-BMT compared to CyA.

Since the early CMV reactivation in the animals that received CyA post-BMT had a detrimental

effect on bone marrow engraftment due to the toxic effect of CMV and antivirals on donor stem

cells as we have previously shown,11

we next sought to evaluate whether an alternate

maintenance immunosuppressant would affect the kinetics of CMV reactivation. We chose to

replace CyA with rapamycin given that rapamycin has been reported to reduce the risk of CMV

infection in organ transplant recipients16

while favoring the proliferation and survival of Tregs

that are part of the conditioning regimen being tested in our tolerance induction protocol.17,18

We

performed 14 BMTs with rapamycin including Treg-treated animals and controls (no Tregs

infused) (Groups C and D). Animals receiving rapamycin had a trend toward delayed CMV

reactivation detected through qPCR analysis compared to those receiving CyA (15.86±2.62

(n=14) vs 6.4±3.4 days (n=5) respectively, p=0.0687) (Figure 2E). Therefore, the use of

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 12: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

12

rapamycin instead of CyA permitted a delay in the use of antiviral treatment while potentially

providing a more favorable environment for Tregs.

3.3 Lower rapamycin levels correlated with CMV reactivation prior to antiviral treatment

Animals in this study received rapamycin doses targeted at a level of 20-30 ng/mL during the

first 30 days, followed by a taper during the ensuing three weeks. In some cases, the rapamycin

levels reached lower levels than desired (during the first month post-BMT) and we noted that

CMV viremia started to increase when the rapamycin levels dropped to ~10 ng/mL if GCV had

not previously been started (Figure 2F). These data support the antiviral activity of rapamycin in

MCM and its use as an immunosuppressant post-BMT, permitting a delay in antiviral use.

3.4 T cell counts during CMV viremia and recovery

We studied the T cell recovery in blood at different time points (Figure 3A) and their correlation

with CMV viremia (Figure 3B). We observed an expected and significant decrease in the T cell

counts during the first month post-BMT compared to the pretransplant levels for both the CD4

and the CD8 populations (p<0.0001) due to T cell depleting therapy. The decrease in T cells was

similar for the rapamycin and CyA recipients (Figure 3A). T cell counts started to recover during

the second month post-BMT, although not yet reaching pre-BMT levels. Although CD8+ T cells

reached lower absolute numbers/L in blood than CD4+ T cells, CD8+ T cell recovery occurred

earlier than CD4+ T cell recovery. By the third month post-BMT, CD8 counts were comparable

to those pre-BMT. On average, recipients that received rapamycin and CyA recovered their T

cell counts at a similar rate, with no significant difference between these two groups

posttransplant (Figure 3A).

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 13: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

13

T cell number analysis was performed at different stages of CMV reactivation to determine if

CMV influenced T cell proliferation. Both prior to and at the time of CMV reactivation, T cell

levels were significantly lower compared to the pretransplant counts (Figure 3B and Table 1). By

the time CMV became undetectable in serum, while animals were on GCV treatment, we

observed an increase in the CD4 and CD8 counts. At that point, we kept the animals on GCV

because T cell counts were still low. When CD8 counts surpassed 500 cells/L the GCV

treatment was switched to VGC in order to protect from reactivation and T cell numbers

remained relatively stable for over two weeks (Figure 3B).

3.5 Expansion of naïve and memory T cells

We further studied the T cell reconstitution during CMV reactivation in the rapamycin-treated

recipients based on their naïve (CD28+CD95-), central memory (CD28+CD95+) and effector

memory (CD28-CD95+) phenotype19

in CD8+ and CD4+ cells (Figure 4A). Prior to

conditioning, T cells with naïve phenotype were the predominate subset among both CD4+ and

CD8+ T cells. Following conditioning and transplantation, the naïve CD4+ and CD8+ T cell

percentages were markedly reduced. In contrast to CD4+ T cells, which shifted toward a central

memory phenotype, the percentage of both central and effector memory phenotypes increased

among CD8+ cells (Figure 4B and 4C). The trends for absolute counts mirrored the trends for the

percentage of naïve and memory cells for both CD4 and CD8 T cells (Figure 4D and 4E,

respectively).

3.6 CMV clearance and antiviral treatment

We studied different preemptive treatment approaches for MCM that received this tolerance

induction regimen. In one recipient (90-7), GCV at a dose of 12.5 mg/kg BID controlled viremia

after initiation at close to 200,000 copies/mL, but CMV reactivated late despite maintenance

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 14: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

14

treatment with VGC that was started to allow oral drug administration (Figure 5A). Here we

focused on the CD8+ T cell counts as have been shown to be fundamental for the control of

CMV. Viremia decreased to 4,000 copies/mL after CD8+ T cells increased to >350 cells/µL

while the animal was maintained on VGC. In a second animal (90-39), antiviral treatment with

GCV (starting at 10 mg/kg up to 15 mg/kg BID) was initiated when the viral load exceeded

150,000 copies/mL but was unable to control viremia that eventually peaked at >600,000

copies/mL (Figure 5B). In addition, it has been our experience that foscarnet alone (90 mg/kg)

did not prevent CMV reactivation (Figure 5C). Furthermore, in one animal (90-15) the

combination of foscarnet at 90 mg/kg BID with GCV at low doses (5 mg/kg BID) intended to

limit myelosuppression likewise failed to control CMV reactivation (Figure 5D). Overall, we

found that CMV clearance was efficient with GCV at 12.5 mg/kg BID when treatment was

instituted before CMV viremia rose above 10,000 copies/mL and maintained at this dose until

viremia was undetectable for >2 weeks and CD8+ T cell numbers exceeded 500 copies/mL in the

peripheral blood (e.g. Figure 5E).

Based on this initial experience, when the rapamycin-treated animals reactivated CMV, GCV

treatment was initiated at 12.5 mg/kg BID when viremia reached >1,000 copies/mL. After CMV

in serum was tested negative for two consecutive weeks and the CD8+ cells recovered >500

cells/µl, GCV was switched to VGC. CMV was controlled successfully with this strategy, which

frequently allowed us to delay antiviral treatment with myelosuppressive drugs during the first

few weeks after BMT (Figure 6).

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 15: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

15

4. Discussion

Human CMV is a ubiquitous virus, highly prevalent among humans.1,2

CMV remains the most

important opportunistic pathogen in immunocompromised individuals such as BMT recipients.20-

23 The CMV status of the donor and recipient plays a major role in the development of viremia,

with seronegative recipients receiving transplants from seropositive donors having the highest

risk of disease. In addition, the intensity of recipient immune system manipulation has a major

impact on the development of opportunistic infections. Despite the highly species-specific nature

of each CMV strain, NHPs represent an excellent model for the study of CMV as the genomic

organization and coding potential closely resembles human CMV, facilitating mechanistic

studies of viral pathogenesis and its effect on the host immune response.24-26

The CMV viruses

that are endemic in NHPs and humans are evolutionarily related.27

Studies have compared the

structure and genetics of CMV virus in Cynomolgus macaques to those isolated from Rhesus

macaques and humans. They demonstrated that the Cynomolgus CMV glycoprotein B amino

acid sequence is 88% homologous to rhesus and 76% homologous to human glycoprotein B,

respectively.24

Of the 262 open reading frames identified in Cynomolgus CMV, 137 are

homologous to human CMV.28

Here, we present the outcomes of CMV infection in Cynomolgus macaque recipients that

underwent a nonmyeloablative conditioning regimen, BMT with or without in vitro expanded

polyclonal recipient Tregs and a short course of immunosuppression monotherapy with either

CyA or rapamycin.11

In our study, all seropositive recipients reactivated CMV post-BMT

irrespective of the immunosuppressive therapy. Our qPCR assay was able to detect and monitor

CMV viremia, which allowed us to initiate treatment only once viremia was detected, thus

avoiding the use of myelosuppressive antivirals during the period of stem cell engraftment.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 16: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

16

We studied several approaches to control CMV in our MCM transplant tolerance induction

protocol. High levels of CMV viremia were difficult to control and became lethal unless

promptly treated. Foscarnet as prophylaxis did not effectively inhibit CMV viremia in our model.

We found that GCV at 12.5 mg/kg BID was able to control CMV infection if instituted soon

after viremia was detected and before the viremia reached 10,000 copies/mL. Lower GCV doses

were not sufficient to clear the viremia. Long-term CMV control was maintained when VGC was

given once the viremia cleared and CD8 counts were >500 cells/µL. Thus, we show here in our

MCM model that CMV can be controlled by high doses of GCV followed by VGC after viremia

clears and CD8 T cell counts recover.

Rapamycin is an immunosuppressant with reported antiviral properties that may be effective

against CMV.16

We compared the serologic titers and response to treatment in animals that

received CyA versus rapamycin. We found that animals receiving rapamycin had delayed CMV

reactivation compared to CyA-treated animals. By delaying the CMV reactivation, rapamycin

used with our preemptive treatment protocol delayed the use of myelosuppressive antiviral

treatment, thus allowing the donor BM additional time to engraft without being affected by the

toxic effect of the drugs, representing a promising approach in BMT recipients.29

The beneficial

effects on CMV viremia that we observed after changing from CyA to rapamycin must be

balanced with the knowledge of the different side effects and toxicities of each drug. While,

CyA is associated with hypertension, diabetes, nephrotoxicity, neurotoxicity, liver toxicity, and

diarrhea, rapamycin can cause impaired wound healing and angiogenesis, mouth ulcers,

thrombocytopenia, peripheral edema, arthralgia, and interstitial pneumonitis. Thus, the

appropriate immunosuppression for a particular patient must be chosen after considering how the

drugs may affect their overall health status.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 17: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

17

Our conditioning protocol, including ATG-based T-cell depletion, uniformly resulted in

substantial CD4+ and CD8+ T cell reduction. It was during this lymphopenic period that CMV

reactivation occurred. We found that T cell recovery, predominantly by CD8+ T cells, started at

the peak of CMV viremia and progressed rapidly until the virus became undetectable in the

serum. The expansion occurred mostly in central and effector memory T cells. There are several

potential explanations for this pattern of T cell expansion. First, lymphopenia-driven expansion

is known to drive the division of T cells that then assume a memory phenotype. However, CMV

is also known to drive T cell recovery after T cell depletion. In a clinical study of kidney

recipients that received rabbit ATG, when T cell recovery was compared between recipients

based on their CMV serostatus, CD8+ T cells were shown to repopulate faster (1-2 months) in

those patients that reactivated CMV compared to seronegative recipients (taking up to two

years).30

The CD8+ T cells had an effector and memory phenotype, suggesting that memory T

cell expansion was driven by CMV replication. CMV reactivation was also reported in a BMT

study performed in Rhesus macaques that sought to induce mixed chimerism. Similar to our

study, that group also showed that the repopulating CD8+ T cells shifted from a naïve to a

central and effector phenotype during CMV infection.31

These findings could suggest that CMV

infection promoted T cell expansion that in turn caused immune activation against CMV and

potentially the graft due to heterologous immunity. Consistent with this possibility, we

previously reported that the only Cynomolgus recipient that remained CMV negative in a prior

study of BMT achieved a markedly increased level and of duration chimerism compared to the

rest of the group.11

However, in order to rigorously study the effect of CMV on BM engraftment

and rejection, we would require a large CMV seronegative control group. Unfortunately, CMV-

seronegative MCM are extremely rare making a study of this nature infeasible.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 18: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

18

Novel approaches are currently being studied for the prevention and control of CMV to avoid or

decrease the requirements for conventional antiviral therapies. Adoptive transfer of donor CMV-

specific T cells offers an alternative to restore CMV immunity that would reduce the need for the

conventional antiviral treatments and their side effects.32-34

In addition, they provide an

alternative in the face of CMV drug-resistance, which is currently a growing problem.35,36

One

potential pitfall of this strategy is the possibility of heterologous immunity of anti-CMV T cells

and precipitation of rejection by the use of cellular immunotherapy.37

Alternatively, CMV

vaccines represent an approach being investigated for CMV prevention.38

Although there are

currently no licensed CMV vaccines, there is an increasing interest in this approach and vaccines

are under clinical development. CMV-specific CD8+ and CD4+ T cell recovery is associated

with protection against CMV.39

Therefore, monitoring the CMV-specific T cell population might

be a strategic approach to predict CMV reactivation in BMT recipients and improve the timing

for the initiation of antiviral treatment.40,41

The use of tetrameric complexes of HLA molecules

loaded with a CMV peptide has been investigated in human BMT recipients to monitor the

recovery of CMV-specific CD8+ T cells posttransplant.6 Findings support the need for CMV-

specific CD8+ T cells for CMV protection and suggest that graft-origin CMV-specific memory T

cells contributed to CMV protection. The development of nonhuman primate tetramers and other

assays for the study of these populations in MCM transplant models would facilitate the

determination of whether or not CMV represents a barrier to the induction of transplantation

tolerance.

Our treatment strategy with GCV starting at the time of viremia reaching 1,000 copies/mL with

eventual transition to VGC was successful in controlling CMV in infected animals. This strategy

is also adopted in the clinic in BMT recipients, where antiviral treatment is started after CMV

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 19: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

19

detection. Unlike solid organ transplantation, where CMV prophylaxis is an option, the early use

of antivirals is avoided in BMT recipients due to the myelosuppressive component of these

drugs. We propose that for solid organ transplantation studies in macaques, prophylactic dosing

with GCV be administered if T cell depletion is utilized. For BMT studies, we propose a rigorous

monitoring protocol for CMV reactivation and initiation of GCV at 12.5 mg/kg BID once

viremia exceeds 1,000 copies/mL. Other groups that study BMT in NHPs have used cidofovir

prophylaxis during the peritransplant period. They found a significant incidence of CMV

reactivation, consistent with our experience that it was unable to suppress viremia completely,

and therefore frequent monitoring for viremia is still required.31

Recently, a new CMV

prophylaxis approach that lacks the toxic effects of the currently available antiviral drugs,

letermovir, has been developed.42,43

Letermovir inhibits CMV replication by binding to

components of the terminase complex (UL51, UL56 or both). It is now clinically available and

clinical trials showed that prophylaxis with letermovir resulted in a significant lower risk of

clinically CMV infection compared to those that received placebo. Further studies in MCM are

necessary to establish its potential in this model.

In conclusion, CMV infection remains a challenge for BMT recipients. Viremia and antiviral

therapy not only interfere with BM engraftment, hindering successful outcomes, but also cause

morbidity and mortality in immunocompromised recipients. Similar results were observed in our

Cynomolgus macaque transplant tolerance induction protocol, where recipients developed CMV

viremia that progressed to clinical CMV disease if not treated promptly. Additionally, the

immune response to CMV may contribute to immune activation, thus preventing the

development of chimerism and donor-specific tolearance.11

Newer antivirals without bone

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 20: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

20

marrow toxicity are a promising approach to avoid CMV reactivation and its consequent adverse

effects.

Acknowledgments

ATG used in this study was generously provided by Pfizer.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 21: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

21

References:

1. Staras SA, Dollard SC, Radford KW, et al. Seroprevalence of cytomegalovirus infection in the

United States, 1988-1994. Clin Infect Dis. 2006;43(9):1143-1151.

2. Xu GJ, Kula T, Xu Q, et al. Viral immunology. Comprehensive serological profiling of human

populations using a synthetic human virome. Science. 2015;348(6239):aaa0698.

3. Azevedo LS, Pierrotti LC, Abdala E, et al. Cytomegalovirus infection in transplant recipients.

Clinics (Sao Paulo). 2015;70(7):515-523.

4. Li CR, Greenberg PD, Gilbert MJ, et al. Recovery of HLA-restricted cytomegalovirus (CMV)-

specific T-cell responses after allogeneic bone marrow transplant: correlation with CMV disease

and effect of ganciclovir prophylaxis. Blood. 1994;83(7):1971-1979.

5. Cwynarski K, Ainsworth J, Cobbold M, et al. Direct visualization of cytomegalovirus-specific

T-cell reconstitution after allogeneic stem cell transplantation. Blood. 2001;97(5):1232-1240.

6. Gratama JW, van Esser JW, Lamers CH, et al. Tetramer-based quantification of

cytomegalovirus (CMV)-specific CD8+ T lymphocytes in T-cell-depleted stem cell grafts and

after transplantation may identify patients at risk for progressive CMV infection. Blood.

2001;98(5):1358-1364.

7. Singhal S, Shaw JC, Ainsworth J, et al. Direct visualization and quantitation of

cytomegalovirus-specific CD8+ cytotoxic T-lymphocytes in liver transplant patients.

Transplantation. 2000;69(11):2251-2259.

8. Hassan-Walker AF, Vargas Cuero AL, Mattes FM, et al. CD8+ cytotoxic lymphocyte

responses against cytomegalovirus after liver transplantation: correlation with time from

transplant to receipt of tacrolimus. J Infect Dis. 2001;183(6):835-843.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 22: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

22

9. Jin X, Demoitie MA, Donahoe SM, et al. High frequency of cytomegalovirus-specific

cytotoxic T-effector cells in HLA-A*0201-positive subjects during multiple viral coinfections. J

Infect Dis. 2000;181(1):165-175.

10. Amir AL, D'Orsogna LJ, Roelen DL, et al. Allo-HLA reactivity of virus-specific memory T

cells is common. Blood. 2010;115(15):3146-3157.

11. Duran-Struuck R, Sondermeijer HP, Bühler L, et al. Effect of Ex Vivo-Expanded Recipient

Regulatory T Cells on Hematopoietic Chimerism and Kidney Allograft Tolerance Across MHC

Barriers in Cynomolgus Macaques. Transplantation. 2017;101(2):274-283.

12. Budde ML, Wiseman RW, Karl JA, et al. Characterization of Mauritian cynomolgus

macaque major histocompatibility complex class I haplotypes by high-resolution

pyrosequencing. Immunogenetics. 2010;62(11-12):773-780.

13. Vargas-Inchaustegui DA, Demberg T, Robert-Guroff M. A CD8alpha(-) subpopulation of

macaque circulatory natural killer cells can mediate both antibody-dependent and antibody-

independent cytotoxic activities. Immunology. 2011;134(3):326-340.

14. Zitsman JS, Alonso-Guallart P, Ovanez C, et al. Distinctive Leukocyte Subpopulations

According to Organ Type in Cynomolgus Macaques. Comp Med. 2016;66(4):308-323.

15. Karl JA, Bohn PS, Wiseman RW, et al. Major histocompatibility complex class I haplotype

diversity in Chinese rhesus macaques. G3 (Bethesda). 2013;3(7):1195-1201.

16. Demopoulos L, Polinsky M, Steele G, et al. Reduced risk of cytomegalovirus infection in

solid organ transplant recipients treated with sirolimus: a pooled analysis of clinical trials.

Transplant Proc. 2008;40(5):1407-1410. ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 23: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

23

17. Sugiyama H, Maeda Y, Nishimori H, et al. Mammalian target of rapamycin inhibitors permit

regulatory T cell reconstitution and inhibit experimental chronic graft-versus-host disease. Biol

Blood Marrow Transplant. 2014;20(2):183-191.

18. Zhao T, Yang C, Qiu Y, et al. Comparison of regulatory T cells and FoxP3-positive T-cell

subsets in the peripheral blood of renal transplant recipients with sirolimus versus cyclosporine:

a preliminary study. Transplant Proc. 2013;45(1):148-152.

19. Nadazdin O, Boskovic S, Murakami T, et al. Phenotype, distribution and alloreactive

properties of memory T cells from cynomolgus monkeys. Am J Transplant. 2010;10(6):1375-

1384.

20. Meyers JD, Flournoy N, Thomas ED. Nonbacterial pneumonia after allogeneic marrow

transplantation: a review of ten years' experience. Rev Infect Dis. 1982;4(6):1119-1132.

21. Meyers JD, Flournoy N, Thomas ED. Risk factors for cytomegalovirus infection after human

marrow transplantation. J Infect Dis. 1986;153(3):478-488.

22. Ljungman P, Brand R, Hoek J, et al. Donor cytomegalovirus status influences the outcome of

allogeneic stem cell transplant: a study by the European group for blood and marrow

transplantation. Clin Infect Dis. 2014;59(4):473-481.

23. Teira P, Battiwalla M, Ramanathan M, et al. Early cytomegalovirus reactivation remains

associated with increased transplant-related mortality in the current era: a CIBMTR analysis.

Blood. 2016;127(20):2427-2438.

24. Ambagala AP, Marsh A, Chan J, et al. Isolation and characterization of cynomolgus macaque

(Macaca fascicularis) cytomegalovirus (CyCMV). Virology. 2011;412(1):125-135.

25. Powers C, Früh K. Rhesus CMV: an emerging animal model for human CMV. Med

Microbiol Immunol. 2008;197(2):109-115.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 24: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

24

26. Yue Y, Barry PA. Rhesus cytomegalovirus a nonhuman primate model for the study of

human cytomegalovirus. Adv Virus Res. 2008;72:207-226.

27. Russell JN, Marsh AK, Willer DO, et al. A novel strain of cynomolgus macaque

cytomegalovirus: implications for host-virus co-evolution. BMC Genomics. 2016;17:277.

28. Marsh AK, Willer DO, Ambagala APN, et al. Genomic Sequencing and Characterization of

Cynomolgus Macaque Cytomegalovirus. J Virol. 2011;85(24):12995-13009.

29. Piñana JL, Perez-Pitarch A, Guglieri-Lopez B, et al. Sirolimus exposure and the occurrence

of cytomegalovirus DNAemia after allogeneic hematopoietic stem cell transplantation. Am J

Transplant. 2018;18(12):2885-2894.

30. Havenith SH, Remmerswaal EB, Bemelman FJ, et al. Rapid T cell repopulation after rabbit

anti-thymocyte globulin (rATG) treatment is driven mainly by cytomegalovirus. Clin Exp

Immunol. 2012;169(3):292-301.

31. Zheng HB, Watkins B, Tkachev V, et al. The Knife's Edge of Tolerance: Inducing Stable

Multilineage Mixed Chimerism but With a Significant Risk of CMV Reactivation and Disease in

Rhesus Macaques. Am J Transplant. 2017;17(3):657-670.

32. Peggs KS, Thomson K, Samuel E, et al. Directly selected cytomegalovirus-reactive donor T

cells confer rapid and safe systemic reconstitution of virus-specific immunity following stem cell

transplantation. Clin Infect Dis. 2011;52(1):49-57.

33. Blyth E, Clancy L, Simms R, et al. Donor-derived CMV-specific T cells reduce the

requirement for CMV-directed pharmacotherapy after allogeneic stem cell transplantation.

Blood. 2013;121(18):3745-3758. ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 25: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

25

34. Walter EA, Greenberg PD, Gilbert MJ, et al. Reconstitution of cellular immunity against

cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the

donor. N Engl J Med. 1995;333(16):1038-1044.

35. Macesic N, Langsford D, Nicholls K, et al. Adoptive T cell immunotherapy for treatment of

ganciclovir-resistant cytomegalovirus disease in a renal transplant recipient. Am J Transplant.

2015;15(3):827-832.

36. Holmes-Liew CL, Holmes M, Beagley L, et al. Adoptive T-cell immunotherapy for

ganciclovir-resistant CMV disease after lung transplantation. Clin Transl Immunology.

2015;4(3):e35.

37. Che JW, Daniels KA, Selin LK, Welsh RM. Heterologous Immunity and Persistent Murine

Cytomegalovirus Infection. J Virol. 2017;91(2):e01386-16.

38. Schleiss MR. Cytomegalovirus vaccines under clinical development. J Virus Erad.

2016;2(4):198-207.

39. Gerna G, Lilleri D, Chiesa A, et al. Virologic and immunologic monitoring of

cytomegalovirus to guide preemptive therapy in solid-organ transplantation. Am J Transplant.

2011;11(11):2463-2471.

40. Baldanti F, Lilleri D, Gerna G. Monitoring human cytomegalovirus infection in transplant

recipients. J Clin Virol. 2008;41(3):237-241.

41. Legendre C, Pascual M. Improving outcomes for solid-organ transplant recipients at risk

from cytomegalovirus infection: late-onset disease and indirect consequences. Clin Infect Dis.

2008;46(5):732-740.

42. Marty FM, Maertens J, Badshah C. Letermovir Prophylaxis for Cytomegalovirus. N Engl J

Med. 2018;378(10):963-965.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 26: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

26

43. Marty FM, Ljungman P, Chemaly RF, et al. Letermovir Prophylaxis for Cytomegalovirus in

Hematopoietic-Cell Transplantation. N Engl J Med. 2017;377(25):2433-2444.

44. Caldwell RG, Marshall P, Fishel J. Method validation and reference range values for a

peripheral blood immunophenotyping assay in non-human primates. J Immunotoxicol.

2016;13(1):64-76.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 27: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

27

Figure legends:

Figure 1. Hematological effects of the BMT preparative regimen

Mean of (A) WBC, (B) hematocrit, (C) platelets, (D) CD4+ T cells, (E) CD8+ T cells, (F) and

NK cells at different time points after BMT in recipients receiving a nonmyeloablative

conditioning regimen. Each colored line represents a different group based on the

immunosuppressive therapy (CyA or rapamycin) and the administration of Tregs: Group A

[green], CyA and Tregs (n=4); Group B [blue], CyA (n=3); Group C [red], rapamycin and Tregs

(n=9); Group D [black], rapamycin (n=5). NK cell data is only available for the rapamycin-

treated recipients. Dotted lines represent normal ranges.44

Figure 2. Effects of CMV viremia and reactivation timing

Histological findings in the (A) meninges and (B) cerebral cortex of a CMV+ recipient (90-47)

that developed uncontrollable CMV disease. C) CD4+ T cell (green) and CD8+ T cell (blue)

counts for 90-47. D) CMV level (red), CD4+ T cell (green), and CD8+ T cell (blue) counts of

recipient 90-39. Antiviral treatment with ganciclovir (blue bar) and valganciclovir (green bar) is

indicated at the top of the graph. E) CMV reactivation time point for recipients receiving

rapamycin (red, n=14) or CyA (blue, n=5) as immunosuppressant. F) Correlation between the

rapamycin level and the day of CMV reactivation posttransplant.

Figure 3. T cell counts during CMV viremia and recovery

A) CD4+ and CD8+ T cell counts at different time points post-BMT in rapamycin-treated (red,

n=14) and CyA-treated (blue, n=7) recipients. Two-tailed unpaired Student’s t-Test was run

between the different time points including the rapamycin and CyA animals within each

comparison. Two-tailed unpaired Student’s t-Test was run between the rapamycin and CyA

recipients at each time point. B) Mean ± SEM of CD4+ and CD8+ T cell counts in rapamycin-

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 28: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

28

treated (red, n=14) and CyA-treated (blue, n=5) recipients in correlation with the CMV

reactivation and clearance time points. Two-tailed unpaired Student’s t-Test was run between the

different time points including the rapamycin and CyA animals within each comparison. 90-1

(CMV seronegative) and 90-47 (CMV was not assessed) were excluded.

Figure 4. Evolution of naïve and memory T cells post-BMT.

A) Schema of the analysis of naïve and memory T cells based on the CD28 and CD95 marker.

Mean ± SEM of the percentage of naïve (blue), central memory (green) and effector memory

(red) cells within B) CD4+ and C) CD8+ T cells in correlation with the CMV reactivation and

clearance time points in recipients receiving rapamycin (n=14). Mean ± SEM of naïve (blue),

central memory (green) and effector memory (red) cell counts within D) CD4+ and E) CD8+ T

cells over time in recipients receiving rapamycin (n=14).

Figure 5. Antiviral approaches in CyA-treated animals

CD8+ T cell counts (black), CMV viremia (red), and antiviral treatment (valganciclovir [green],

ganciclovir [blue], foscarnet [magenta]) in CyA-treated recipients (A through E, n=5). 90-1

(CMV seronegative) and 90-47 (CMV was not assessed) were excluded.

Figure 6. Antiviral treatment in rapamycin-treated animals

CD8+ T cell counts (black), CMV viremia (red), and antiviral treatment (valganciclovir [green],

ganciclovir [blue], foscarnet [magenta], cidofovir [light blue]) in rapamycin-treated recipients

(n=13). Recipient BM12B did not receive antiviral treatment.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 29: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

29

Table 1. CMV reactivation time point and antiviral treatment

Animal

ID

Immunosu

ppressive

Therapy

TBI

(cGy)

Day of

Reactivation

Day of CMV

>1,000

copies/mL

Day of CMV

>10,000

copies/mL

Day of Initiation

of Antiviral

Treatment

Antiviral

Treatment

Cause of

Euthanasia

90-39 CyA 300 0 0 12 17 GCV, VGC CMV viremia

90-15 CyA 300 19 19 23 23 GCV, VGC, Foscarnet

Scientific Endpoint

6C64 CyA 300 5 5 19 2 Foscarnet, GCV, VGC

Scientific Endpoint

6C1 CyA 300 7 12 NA 7 GCV Scientific Endpoint

90-47 CyA 300 NA NA NA NA NA CMV viremia

90-7 CyA 300 1 1 19 10 GCV, VGC Scientific

Endpoint

90-1 CyA 300 NA NA NA NA NA Scientific

Endpoint

SA196A Rapamycin 300 8 8 29 30 GCV Scientific

Endpoint

AM538C Rapamycin 250 26 26 34 34 GCV Scientific

Endpoint

AK746F Rapamycin 250 26 26 28 29 GCV Scientific

Endpoint

BF418F Rapamycin 250 6 NA NA 13 GCV,

Foscarnet,

Cidofovir

Scientific

Endpoint

AG531J Rapamycin 250 16 20 NA 20 GCV Scientific

Endpoint

BM12B Rapamycin 250 29 NA NA NA NA Scientific

Endpoint

AF201G Rapamycin 250 8 26 NA 19 GCV GVHD

V59M Rapamycin 250 10 NA NA 25 GCV GVHD

AJ606D Rapamycin 250 29 33 NA 36 GCV Scientific

Endpoint

AH260F Rapamycin 250 9 9 32 32 GCV Scientific Endpoint

AT468G Rapamycin 250 11 NA NA 17 GCV,

Foscarnet

Renal failure/

Infection

BY648F Rapamycin 250 10 NA NA 16 GCV Scientific

Endpoint

AP532B Rapamycin 250 30 49 NA 43 GCV GVHD

F813M Rapamycin 250 4 28 36 27 GCV GVHD

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 30: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

30

“Day” represents the number of days after conditioning was completed at which the first CMV positive

sample was detected (Day of Reactivation), CMV copies/mL threshold was reached (Day of CMV >1,000

copies/mL or Day of CMV >10,000 copies/mL), or the time point when the antiviral treatment was

started (Day of initiation of antiviral treatment). 90-1 (CMV seronegative), 90-47 and BM12B did not

receive antiviral treatment. NA, not applicable; CMV, cytomegalovirus; CyA, cyclosporine A; TBI, total

body irradiation; GCV, ganciclovir; VGC, valganciclovir.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 31: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

31

Table 2. T-test analysis between different groups depending on the administration of Tregs and

rapamycin or CyA.

Groups WBC Hematocrit Platelets

Treg-CyA (A) vs

Control-CyA (B)

ns **

Control-CyA (B)

**

Control-CyA (B)

Treg-CyA (A) vs

Treg-Rapamycin (C)

ns ***

Treg-Rapamycin (C)

****

Treg-Rapamycin (C)

Treg-CyA (A) vs

Control-Rapamycin (D)

*

Treg-CyA (A)

***

Control-Rapamycin (D)

***

Control-Rapamycin (D)

Control-CyA (B) vs

Treg-Rapamycin (C)

ns *

Control-CyA (B)

*

Control-CyA (B)

Control-CyA (B) vs

Control-Rapamycin (D)

ns *

Control-CyA (B)

*

Control-CyA (B)

Treg-Rapamycin (C) vs

Control-Rapamycin (D)

*

Treg-Rapamycin (C)

ns ns

Two-tailed paired Student’s t-Test analysis was performed between each group for the first 30 days of

the study (*= p ≤ 0.05, **= p ≤ 0.01, ***= p ≤ 0.001, ****= p ≤ 0.0001, ns = not significant). Recipients

were classified as “Group A” (received Tregs and CyA), “Group B” (CyA), “Group C” (Tregs and

rapamycin), and “Group D” (rapamycin). WBC, white blood cell count. The group with higher counts is

indicated in the case of a significant difference.

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 32: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

32

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 33: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

33

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 34: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

34

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 35: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

35

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 36: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

36

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

Page 37: Transplantation Publish Ahead of Print DOI: … · 2019-08-12 · model to study the ability to induce transplantation tolerance through long-term mixed ... Recipient and donor pairs

37

ACCEPTED

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.