Abstract
Background:
Herpes simplex virus (HSV) 1 and 2, varicella zoster virus (VZV), and human herpesvirus 6 (HHV-6) cause severe infections in immunocompromised hosts. Interventions to optimize virus-specific adaptive immunity may have advantages over antivirals in the prophylaxis and treatment of these infections.
Aims:
We sought to review adaptive immune responses and methods for assessing and replenishing cellular and humoral immunity to HSV, VZV and HHV-6 in solid organ transplant (SOT) and hematopoietic cell transplant (HCT) recipients.
Sources:
We searched PubMed for relevant studies on immune responses to HSV, VZV and HHV-6 as well as studies describing methods for evaluating and restoring cell-mediated immunity to other double stranded DNA viruses in transplant recipients. Recent studies, randomized controlled trials, and investigations highlighting key concepts in clinical virology were prioritized for inclusion.
Content:
We describe the mechanisms of adaptive immunity to HSV, VZV and HHV-6, and limitations of antivirals as prophylaxis and treatment for these infections in SOT and HCT recipients. We review methods for measuring and restoring cellular immunity to double stranded DNA viruses, their potential applications to management of HSV, VZV, and HHV-6 in immunocompromised hosts, and barriers to clinical use. Vaccination and virus-specific T cell therapies are discussed in detail.
Implications:
The growing repertoire of diagnostic and therapeutic techniques focused on virus-specific adaptive immunity provides a novel approach to management of viral infections in transplant recipients. Investigations to optimize such interventions specifically in HSV, VZV, and HHV-6 are needed.
Graphical Abstract
Introduction
The human herpesviruses are a group of eight double stranded DNA viruses that establish lifelong latency in human cells following primary infection (1). Reactivation of latent herpesviruses is facilitated by loss of regulatory cellular immunity and may occur any time after primary infection. The immunocompromised state associated with solid organ transplant (SOT) and hematopoietic cell transplant (HCT) increases the risk for severe disease during both primary infection and viral reactivation, and transplant recipients are prone to frequent episodes of the latter (1). Decades of research have centered on immune responses to cytomegalovirus (CMV) and Epstein-Barr virus (EBV) in transplant recipients, while adaptive immunity to non-EBV, non-CMV herpesviruses are seldom the focus of large investigations. The majority of adults worldwide are infected with herpes simplex virus (HSV) 1 and 2, varicella zoster virus (VZV) and/or human herpesvirus 6 (HHV-6), and these infections may cause significant morbidity and occasional mortality following SOT or HCT (2–4). Severe sequalae from human herpesvirus 7 (HHV-7) and human herpesvirus 8 (HHV-8) are rare in transplant recipients (3). In this review, we describe adaptive immunity to HSV, VZV and HHV-6, discuss methods for evaluating virus-specific cellular responses, highlight limitations of available antiviral therapies, and explore novel mechanisms for restoring humoral in SOT and HCT recipients.
Overview of adaptive immunity in HSV, VZV and HHV-6 infections
Herpes simplex viruses
The herpes simplex viruses are α-herpesviruses that most commonly infect the oral or genital mucosa during primary infection and establish latency in the trigeminal nerve (oral HSV) or sacral dorsal root ganglia (genital HSV). HSV-1 and HSV-2 infect approximately 60% and 15–20% of adults by age 50, respectively (5, 6). Most clinical HSV infections in human immunodeficiency virus (HIV)-negative immunocompromised adults are due to HSV-1 and are characterized by mucocutaneous disease in the orofacial region (6, 7). Immunocompromised hosts are predisposed to more severe manifestations of HSV, including dissemination with visceral organ involvement (6, 8).
Cell-mediated immunity (CMI) has a key role in maintaining HSV latency (9, 10). In both SOT and HCT recipients, risk of HSV-1 reactivation is highest in the early post-transplant period when cellular immunity is most impaired (6, 7). Prior to routine antiviral prophylaxis, HSV-1 mucocutaneous disease occurred in up to 80% of seropositive HCT recipients and 15% of seropositive SOT recipients in the first several months following transplant (6, 11–13). Impaired T cell immunity also facilitates emergence of acyclovir resistant HSV during appropriately dosed prophylaxis, reflecting opportunities for viral mutants to arise during unchecked viral replication and the persistence of less fit variants that would otherwise be eliminated in immunocompetent hosts (14, 15). Cross-placental transfer of maternal HSV antibodies is a major mechanism of protection against neonatal HSV among infants born to seropositive mothers, suggesting that humoral immunity (HI) plays a key role during primary infection (16, 17). HI likely has a lesser role in the maintenance of HSV latency and isolated hypogammaglobinemia has not been associated with an increased risk of HSV reactivation (8, 18).
Varicella Zoster Virus
VZV is a ubiquitous α-herpesvirus that infected nearly all children before the advent of childhood varicella vaccination (VARIVAX®) in 1995 (19). Primary infection (varicella) manifests as a characteristic chickenpox rash, after which VZV establishes latency in ganglionic neurons. Thirty percent of VZV-infected individuals experience viral reactivation, manifested as herpes zoster ([HZ], shingles) (20). The incidence of HZ is up to 10 times higher in immunocompromised patients (21). Approximately 15% of HZ cases result in post-herpetic neuralgia (PHN), which is carries significant morbidity and is more common among immunocompromised persons (22).
CMI has central role in maintaining VZV latency. The first investigation of VZV-specific donor T cell infusion for seropositive allogeneic HCT recipients demonstrated limited viral reactivation during the post-transplant period, supporting a model of HZ pathogenesis in which cyclical viral reactivation is subclinical when CMI is intact (23). VZV-specific CMI has been inversely correlated with HZ incidence and related complications, including PHN (24). The role of HI in the prevention of HZ is less clear and likely less critical. In a study of 12,522 adults of whom 401 developed HZ over a three year period, VZV-specific CMI, but not VZV-specific antibody titers, correlated with protection against HZ (24).
Human Herpesvirus 6
HHV-6 (roseolavirus) is a β-herpesvirus that consists of two subspecies, HHV-6A and HHV-6B. Nearly all clinical manifestations of HHV-6 are due to HHV-6B, which infects >95% of people during early childhood (25). Primary HHV-6B infection may be asymptomatic or present with a mild, rash-associated febrile illness known as “exanthema subitem” or “roseola” (26). HHV-6B reactivation occurs in 30–50% of allogeneic HCT recipients, often during the pre-engraftment and early post-engraftment periods (3). HHV-6B in HCT recipients is associated with a variety of complications, including limbic encephalitis (1–8% of HCT recipients), which, when not fatal, frequently leads to severe and prolonged disability (3). HHV-6B reactivation occurs in up to one-third of patients in the first 12 weeks after liver transplantation, but HHV-6B encephalitis in SOT recipients is rare (3, 27).
As with HSV and VZV, CMI plays a greater role in controlling HHV-6 reactivation compared to HI. CD8 knockout, but not B cell deficiency, causes rapidly fatal infection in mice infected with murine roseolavirus, a β-herpesvirus related to HHV-6 (28). In HCT recipients, post-transplant HHV-6B viral loads are inversely correlated with the absolute number of HHV6B specific CD4 T cells contained in the graft, and higher proportions of perforin-expressing CD8 T cells are associated with HHV-6B clearance in viremic patients (28–30).
Limitations of antivirals
In SOT recipients, prophylaxis with acyclovir, valacyclovir or famciclovir is recommended in HSV and/or VZV seropositive patients for 1 month post-transplant and during treatment for rejection (6). HCT recipients remain at risk for VZV reactivation for months to years following transplant, and antiviral prophylaxis is extended for at least one year after HCT (7). HHV-6 prophylaxis is not recommended in SOT or HCT recipients (25). Plasma surveillance and/or preemptive therapy is not recommended for HSV, VZV or HHV-6, though antivirals should be given when there is high suspicion for disease (6, 7, 25).
Antivirals are not universally successful in preventing or treating herpesvirus infections. Breakthrough HSV infections occur in up to 10% of SOT and HCT recipients receiving prophylaxis, particularly if antivirals are underdosed for renal function or when absorption of oral agents is inadequate (6, 7). Emergence of HSV antiviral resistance while on adequately dosed prophylaxis may render pharmacotherapy ineffective, and HHV-6 encephalitis is associated with prolonged morbidity even in patients who receive targeted antiviral therapy (2, 4, 31). HZ after discontinuation of routine prophylaxis is common late after HCT and occurs in 26% of umbilical cord blood transplant recipients within 5 years of transplant (4). While acyclovir and valacyclovir are relatively safe and inexpensive, high pill burden, medication fatigue and transitions between healthcare teams limit long-term adherence (4).
Antiviral prophylaxis may have other unintended consequences that result in poor immune reconstitution. For example, CMV prevention with prophylactic letermovir (HCT) or valganciclovir (SOT) is associated with reduced CMV-specific polyfunctional T cell responses beyond day 100 compared to preemptive treatment approaches, potentially leaving patients vulnerable to a higher incidence of late CMV reactivation and disease (32–34). Clinically, such rebound effects have not been demonstrated after prolonged antiviral prophylaxis for VZV or HSV despite reduced VZV-specific lymphocyte proliferation among HCT recipients exposed to acyclovir (35, 36). Diagnostic tools that assess virus-specific immunity could identify patients at highest risk for viral infections and guide the necessary duration of prophylaxis, and therapies that restore immune function offer an alternative to antivirals for both prophylaxis and treatment of herpesviruses (Table 1).
Table 1:
Methods for assessing and restoring adaptive immunity to HSV, VZV, HHV-6
Diagnostics | Prophylaxis and therapeutics | ||||
---|---|---|---|---|---|
HI assay (serologies) | CMI assays | Vaccines | Immunoglobulins | Virus-specific T cells | |
HSV-1 & 2 | Widely available; used to assess for evidence of latent infection prior to SOT and HCT | Not available for routine clinical use | Limited to clinical trials for prevention of primary infection | Not indicated for primary prevention, prophylaxis, or treatment | Not developed for use in humans |
VZV | Widely available; used to assess for evidence of latent infection prior to SOT and HCT | Not available for routine clinical use | Primary infection (varicella): live-attenuated varicella vaccine either alone (VARIVAX®) or in combination with measles, mumps, and rubella (MMRV, ProQuad®) | Varicella zoster globulin (VARIZIG®) or pooled intravenous immunoglobulin (IVIG) for post-exposure prophylaxis in VZV seronegative immunocompromised persons | Not developed for use in humans |
Reactivation (herpes zoster): RZV (SHINGRIX®); ZVL (ZOSTAVAX®) no longer available in the US | Not indicated for prevention of reactivation or treatment of zoster | ||||
HHV-6 | Not recommended for routine clinical use | Not available for routine clinical use | Not available for prevention of primary infection or reactivation | Not indicated for primary prevention, prophylaxis, or treatment | Under investigation for prophylaxis and treatment in HCT recipients |
Abbreviations: cell mediated immunity (CMI), hematopoietic cell transplant (HCT), herpes simplex virus (HSV), human herpes virus 6 (HHV-6), humoral immunity (HI), recombinant zoster vaccine (RZV), solid organ transplant (SOT), varicella zoster virus (VZV), zoster vaccine live (ZVL)
Assessing humoral and cellular immune responses
Assessing humoral immunity
Measurement of virus-specific immunoglobulins in clinical practice is rarely performed to evaluate humoral immunity. Instead, HSV and VZV-specific immunoglobulins are used to detect latent infection and identify patients at risk of reactivation following HCT and SOT (6). Antibody tests for HHV-6 are not readily available and not recommended in transplant recipients (25). Furthermore, titers of HSV, VZV or HHV-6-specific immunoglobulins measured by enzyme-linked immunoassay (ELISA) do not necessarily correlate with neutralizing activity (16, 37, 38). Some experts use total immunoglobulin G (IgG) levels as a marker of global infection risk, but hypogammaglobulinemia without concurrent lymphopenia does not correlate with risk of VZV, HSV or HHV-6B reactivation or disease (39, 40).
Assessing cellular immunity
Absolute CD4 T cell count is often measured as a global assessment of immune reconstitution in transplant recipients but may not accurately reflect virus-specific CMI (4, 41). Cytokine release assays that measure virus-specific CMI have potential to predict risk of herpesvirus infections. Commercial and in-house enzyme-linked immunospot (ELISPOT) assays, which detect interferon-γ release by purified peripheral blood mononuclear cells (PBMCs) after incubation with virus-specific peptides or antigen lysates, have been developed to assess CMV-CMI, though are unable to differentiate CD4 and CD8 responses (42, 43). The ratio of IFN- γ producing cells to PBMC is measured, but thresholds for positivity vary between individual assays (42). Prospective studies of CMV-ELISPOT assays in HCT and kidney transplant recipients have demonstrated that positive results, indicative of in vitro CMV-CMI, have high negative predictive values (>93%) but low positive predictive values (<30%) for CS-CMVi (43, 44). Among 241 CMV seropositive allogeneic HCT recipients, CMV-CMI was low in 94% of patients who experienced CS-CMVi in the first 6 months after transplant (43).
Such CMI assays for HSV, VZV and HHV-6 are not clinically available but could help providers understand an individual’s risk for viral infections and guide duration of HSV and VZV pharmacologic prophylaxis following transplant or after immunization, particularly among HCT recipients with profound T cell dysfunction. VZV-CMI has been correlated with protection against HZ in vaccine clinical trials, suggesting that VZV-CMI assays may predict safe termination of VZV prophylaxis after HCT (24). Specific thresholds associated with protection need to be established before CMI assays for non-CMV herpesviruses can be integrated into clinical practice.
Restoring humoral and cellular immunity
Active immunization (vaccination)
Vaccination to promote endogenous humoral and cellular immunity is one approach to restoring virus-specific immune responses. VZV is the only herpesvirus for which licensed vaccines are currently available. Vaccines for the prevention of HSV and HHV-6 primary infection have been developed but efforts have not focused on immunocompromised populations (16, 38). In addition to the live attenuated vaccine given to children to prevent varicella, two vaccines have been developed for the prevention of HZ in adults: a live attenuated vaccine (zoster vaccine live [ZVL], ZOSTAVAX®) and an adjuvanted recombinant glycoprotein E subunit vaccine (recombinant zoster vaccine [RZV], SHINGRIX®). RZV has greater vaccine efficacy than ZVL for the prevention of HZ and is the only zoster vaccine available in the United States (45, 46). Unlike ZVL, RZV does not carry the risk of disseminated VZV infection and is therefore safer in immunocompromised populations. The United States Food and Drug Administration has approved RZV for immunocompromised adults aged 18 and older (47).
SOT and autologous HCT recipients receiving RZV in clinical trials demonstrated similar or slightly reduced humoral and cellular immune responses compared to immunocompetent adults (20, 45, 48–52). In a placebo-controlled clinical trial of RZV in autologous HCT recipients, vaccine efficacy was ~70% and RZV elicited humoral and cellular responses in 67% and 93% of vaccinees, respectively (48). RZV may be less immunogenic and less effective in allogeneic HCT recipients. In an observational series of allogeneic HCT recipients who received RZV at a median of 7 months post-transplant, humoral response occurred in only 3/18 (18%) patients (53). In a separate cohort of allogeneic HCT recipients who received 2 doses of RZV and subsequently discontinued antiviral prophylaxis, 3 (2%) developed HZ within 6 months of the second dose (53). Notably, data on the safety, immunogenicity, and efficacy of RZV in the first 6–12 months after allogeneic HCT or SOT are limited, and RZV has not eliminated the need for antiviral prophylaxis in the early post-transplant periods. Passive immunization strategies may be more effective in the early periods after HCT and SOT until adequate immune reconstitution is achieved and documented with virus-specific assays.
Immune globulins
Passive immunization with pooled intravenous (or subcutaneous) immune globulin (IVIG) replenishes humoral immunity and is used to prevent bacterial and sinopulmonary infections in patients with a variety of immunocompromising conditions but does not have a major role in management of HSV, VZV or HHV-6 (6, 7, 25, 39). Immune globulin preparations containing high titers of anti-VZV IgG are recommended to reduce the attack rate and prevent complications of varicella in VZV-seronegative immunocompromised patients after VZV exposure but are not routinely used to prevent reactivation of or treat infections due to HSV, VZV or HHV-6 in seropositive immunocompromised populations (6, 7, 25, 54).
Virus-specific T cells (VSTs)
Single or repeated virus-specific T cell (VST) infusions are a promising passive immunization strategy to replenish cellular immunity in immunocompromised patients. In HCT recipients, donor lymphocyte infusions are limited by graft versus host reactions due to the population of donor T cells recognizing recipient antigens. Selected infusion of CD4 and CD8 VSTs, or other approaches such as virus-specific NK cells or T cells with engineered receptors, may provide CMI while avoiding off-target T cell alloreactivity (55, 56). VST products were first designed to target individual viruses, including CMV, EBV, BK virus, and adenovirus (57). More recently, multivirus VST products that recognize a collection of double stranded DNA viruses, including HHV-6B, are being studied for prophylaxis and treatment of viral infections in high-risk HCT recipients, and have been administered for drug-refractory HHV-6B encephalitis in a limited number of patients (56, 58, 59). HSV-1 specific VSTs have been generated in vitro, but there are no reports of HSV or VZV specific T cell use in humans to date (56, 60). No serious adverse events have been causally linked to VST infusion, though GVHD and cytokine release syndrome are theoretical risks (56). Despite VSTs’ potential to revolutionize the management of viral infections in immunocompromised populations, evidence from placebo-controlled trials is not yet available, and there are several challenges to VST implementation in clinical practice (Table 2).
Table 2:
Advantages and limitations of virus-specific T cells (VSTs) for the prophylaxis and treatment of HSV, VZV and HHV-6 in immunocompromised hosts
Advantages of VSTs | Limitations of VSTs | |
---|---|---|
Implementation | No pill burden, intended for short-term use as a “bridge” to immune reconstitution | Costs associated with production and storage |
Does not require enteral absorption or dose adjustment for renal function | Infusion center needs (logistics of scheduling, travel for patients) | |
Stored banks of “off the shelf” products from third party donors with a variety of HLA types avoid treatment delays related to finding a suitably matched donor (56) | HLA matching between VST donor, HCT donor, and recipient | |
Safety | Well-tolerated in small clinical trials | Theoretical risk of GVHD or CRS |
Efficacy | Option for HHV-6 prophylaxis; complementary or alternative strategy for treatment of HHV-6 or acyclovir resistant HSV | Existing antivirals have established efficacy in management of HSV and VZV |
Enthusiasm for research on cell-based therapies following the success of CAR-T cells for treatment of B cell malignancies | Impact of corticosteroids, T cell depleting antibodies and other immunosuppressants on efficacy | |
Reported success in treatment of HHV- 6B encephalitis, under active investigation for prophylaxis in allogeneic HCT recipients (56, 59) | Large, placebo-controlled trials lacking |
Abbreviations: chimeric antigen receptor modified-T (CAR-T), cytokine release syndrome (CRS), graft versus host disease (GVHD), human herpesvirus 6 (HHV-6), herpes simplex virus (HSV), varicella zoster virus (VZV), virus-specific T cell
Conclusions
HSV, VZV and HHV-6 continue to cause severe disease in patients with impaired cellular immunity. While antiviral prophylaxis has reduced the incidence of VZV and HSV infections following HCT and SOT, there are several limitations of antivirals in terms of safety, efficacy, and implementation. Laboratory methods assessing virus-specific CMI immunity have potential to guide the duration VZV prophylaxis following HCT, but clinical correlates of protection need to be established before routine use. Interventions focusing on measuring and optimizing an individual’s cellular immunity offer a precision medicine approach for infection prevention and management. Highly immunogenic vaccines may be valuable in some immunocompromised patients, and initiatives to develop vaccines that prevent HSV and HHV-6B reactivation are needed. However, in patients with the most profound deficits in CMI, active immunization is unlikely to induce protective immunity. VSTs are a promising tool to reduce viral reactivation and replication in highly immunocompromised patients awaiting CMI recovery. Overcoming the impact of immunosuppressants on VST expansion and function should be a focus of future investigations.
Funding:
This work was supported by the National Institute of Allergy and Infectious Diseases (T32AI118690 to M.R.H) and the National Cancer Institute (U01CA247548 to J.A.H) of the National Institutes of Health. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Conflict of interest disclosures:
M.R.H. received speaking honoraria from Cigna LifeSource and Thermo Fisher Scientific. J.A.H received consulting fees from Gilead Sciences, Allovir, Takeda and research support from Takeda, Allovir, Deverra Thereapeutics, and Gilead. K.M.A. has no relevant interests to disclose.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Johnson DC, Hill AB. 1998. Herpesvirus evasion of the immune system. Curr Top Microbiol Immunol 232:149–77. [DOI] [PubMed] [Google Scholar]
- 2.Sahoo F, Hill JA, Xie H, Leisenring W, Yi J, Goyal S, Kimball LE, Lee I, Seo S, Davis C, Pergam SA, Flowers ME, Liaw KL, Holmberg L, Boeckh M. 2017. Herpes Zoster in Autologous Hematopoietic Cell Transplant Recipients in the Era of Acyclovir or Valacyclovir Prophylaxis and Novel Treatment and Maintenance Therapies. Biol Blood Marrow Transplant 23:505–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hill JA, Zerr DM. 2014. Roseoloviruses in transplant recipients: clinical consequences and prospects for treatment and prevention trials. Curr Opin Virol 9:53–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Xue E, Xie H, Leisenring WM, Kimball LE, Goyal S, Chung L, Blazevic R, Maltez B, Edwards A, Dahlberg AE, Salit RB, Delaney C, Pergam SA, Boeckh M, Milano F, Hill JA. 2021. High Incidence of Herpes Zoster After Cord Blood Hematopoietic Cell Transplant Despite Longer Duration of Antiviral Prophylaxis. Clin Infect Dis 72:1350–1357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.McQuillan G, Kruszon-Moran D, Flagg EW, Paulose-Ram R. 2018. Prevalence of Herpes Simplex Virus Type 1 and Type 2 in Persons Aged 14–49: United States, 2015–2016. NCHS Data Brief:1–8. [PubMed] [Google Scholar]
- 6.Lee DH, Zuckerman RA, Practice AIDCo. 2019. Herpes simplex virus infections in solid organ transplantation: Guidelines from the American Society of Transplantation Infectious Diseases Community of Practice. Clin Transplant 33:e13526. [DOI] [PubMed] [Google Scholar]
- 7.Styczynski J, Reusser P, Einsele H, de la Camara R, Cordonnier C, Ward KN, Ljungman P, Engelhard D, Leukemia SECoIi. 2009. Management of HSV, VZV and EBV infections in patients with hematological malignancies and after SCT: guidelines from the Second European Conference on Infections in Leukemia. Bone Marrow Transplant 43:757–70. [DOI] [PubMed] [Google Scholar]
- 8.Corey L, Spear PG. 1986. Infections with herpes simplex viruses (1). N Engl J Med 314:686–91. [DOI] [PubMed] [Google Scholar]
- 9.Ouwendijk WJ, Laing KJ, Verjans GM, Koelle DM. 2013. T-cell immunity to human alphaherpesviruses. Curr Opin Virol 3:452–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Freeman ML, Sheridan BS, Bonneau RH, Hendricks RL. 2007. Psychological stress compromises CD8+ T cell control of latent herpes simplex virus type 1 infections. J Immunol 179:322–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Pass RF, Whitley RJ, Whelchel JD, Diethelm AG, Reynolds DW, Alford CA. 1979. Identification of patients with increased risk of infection with herpes simplex virus after renal transplantation. J Infect Dis 140:487–92. [DOI] [PubMed] [Google Scholar]
- 12.Greenberg MS, Friedman H, Cohen SG, Oh SH, Laster L, Starr S. 1987. A comparative study of herpes simplex infections in renal transplant and leukemic patients. J Infect Dis 156:280–7. [DOI] [PubMed] [Google Scholar]
- 13.Saral R, Burns WH, Laskin OL, Santos GW, Lietman PS. 1981. Acyclovir prophylaxis of herpes-simplex-virus infections. N Engl J Med 305:63–7. [DOI] [PubMed] [Google Scholar]
- 14.Chakrabarti S, Pillay D, Ratcliffe D, Cane PA, Collingham KE, Milligan DW. 2000. Resistance to antiviral drugs in herpes simplex virus infections among allogeneic stem cell transplant recipients: risk factors and prognostic significance. J Infect Dis 181:2055–8. [DOI] [PubMed] [Google Scholar]
- 15.Langston AA, Redei I, Caliendo AM, Somani J, Hutcherson D, Lonial S, Bucur S, Cherry J, Allen A, Waller EK. 2002. Development of drug-resistant herpes simplex virus infection after haploidentical hematopoietic progenitor cell transplantation. Blood 99:1085–8. [DOI] [PubMed] [Google Scholar]
- 16.Belshe RB, Leone PA, Bernstein DI, Wald A, Levin MJ, Stapleton JT, Gorfinkel I, Morrow RL, Ewell MG, Stokes-Riner A, Dubin G, Heineman TC, Schulte JM, Deal CD, Women HTf. 2012. Efficacy results of a trial of a herpes simplex vaccine. N Engl J Med 366:34–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Brown ZA, Benedetti J, Ashley R, Burchett S, Selke S, Berry S, Vontver LA, Corey L. 1991. Neonatal herpes simplex virus infection in relation to asymptomatic maternal infection at the time of labor. N Engl J Med 324:1247–52. [DOI] [PubMed] [Google Scholar]
- 18.Dropulic LK, Cohen JI. 2011. Severe viral infections and primary immunodeficiencies. Clin Infect Dis 53:897–909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Shaw J, Gershon AA. 2018. Varicella Virus Vaccination in the United States. Viral Immunol 31:96–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hirzel C, L’Huillier AG, Ferreira VH, Marinelli T, Ku T, Ierullo M, Miao C, Schmid DS, Juvet S, Humar A, Kumar D. 2021. Safety and immunogenicity of adjuvanted recombinant subunit herpes zoster vaccine in lung transplant recipients. Am J Transplant 21:2246–2253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.McKay SL, Guo A, Pergam SA, Dooling K. 2020. Herpes Zoster Risk in Immunocompromised Adults in the United States: A Systematic Review. Clin Infect Dis 71:e125–e134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Forbes HJ, Thomas SL, Smeeth L, Clayton T, Farmer R, Bhaskaran K, Langan SM. 2016. A systematic review and meta-analysis of risk factors for postherpetic neuralgia. Pain 157:30–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ma CK, Blyth E, Clancy L, Simms R, Burgess J, Brown R, Deo S, Micklethwaite KP, Gottlieb DJ. 2015. Addition of varicella zoster virus-specific T cells to cytomegalovirus, Epstein-Barr virus and adenovirus tri-specific T cells as adoptive immunotherapy in patients undergoing allogeneic hematopoietic stem cell transplantation. Cytotherapy 17:1406–20. [DOI] [PubMed] [Google Scholar]
- 24.Asada H 2019. VZV-specific cell-mediated immunity, but not humoral immunity, correlates inversely with the incidence of herpes zoster and the severity of skin symptoms and zoster-associated pain: The SHEZ study. Vaccine 37:6776–6781. [DOI] [PubMed] [Google Scholar]
- 25.Ward KN, Hill JA, Hubacek P, de la Camara R, Crocchiolo R, Einsele H, Navarro D, Robin C, Cordonnier C, Ljungman P, (ECIL) ECoIiL. 2019. Guidelines from the 2017 European Conference on Infections in Leukaemia for management of HHV-6 infection in patients with hematologic malignancies and after hematopoietic stem cell transplantation. Haematologica 104:2155–2163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Cermelli C, Fabio G, Montorsi M, Sabbatini AM, Portolani M. 1996. Prevalence of antibodies to human herpesviruses 6 and 7 in early infancy and age at primary infection. New Microbiol 19:1–8. [PubMed] [Google Scholar]
- 27.Fernández-Ruiz M, Kumar D, Husain S, Lilly L, Renner E, Mazzulli T, Moussa G, Humar A. 2015. Utility of a monitoring strategy for human herpesviruses 6 and 7 viremia after liver transplantation: a randomized clinical trial. Transplantation 99:106–13. [DOI] [PubMed] [Google Scholar]
- 28.Hanson DJ, Hill JA, Koelle DM. 2018. Advances in the Characterization of the T-Cell Response to Human Herpesvirus-6. Front Immunol 9:1454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.de Pagter PJ, Boelens JJ, Jacobi R, Schuurman R, Nanlohy NM, Sanders EA, van Baarle D. 2013. Increased proportion of perforin-expressing CD8+T-cells indicates control of herpesvirus reactivation in children after stem cell transplantation. Clin Immunol 148:92–8. [DOI] [PubMed] [Google Scholar]
- 30.Hanson DJ, Xie H, Zerr DM, Leisenring WM, Jerome KR, Huang ML, Stevens-Ayers T, Boeckh M, Koelle DM, Hill JA. 2021. Donor-Derived CD4+ T Cells and Human Herpesvirus 6B Detection After Allogeneic Hematopoietic Cell Transplantation. J Infect Dis 223:709–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Limaye AP, Bakthavatsalam R, Kim HW, Kuhr CS, Halldorson JB, Healey PJ, Boeckh M. 2004. Late-onset cytomegalovirus disease in liver transplant recipients despite antiviral prophylaxis. Transplantation 78:1390–6. [DOI] [PubMed] [Google Scholar]
- 32.Singh N, Winston DJ, Razonable RR, Lyon GM, Silveira FP, Wagener MM, Stevens-Ayers T, Edmison B, Boeckh M, Limaye AP. 2020. Effect of Preemptive Therapy vs Antiviral Prophylaxis on Cytomegalovirus Disease in Seronegative Liver Transplant Recipients With Seropositive Donors: A Randomized Clinical Trial. JAMA 323:1378–1387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hill JA, Zamora D, Xie H, Thur LA, Delaney C, Dahlberg A, Pergam SA, Leisenring WM, Boeckh M, Milano F. 2021. Delayed-onset cytomegalovirus infection is frequent after discontinuing letermovir in cord blood transplant recipients. Blood Adv 5:3113–3119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zamora D, Duke ER, Xie H, Edmison BC, Akoto B, Kiener R, Stevens-Ayers T, Wagner R, Mielcarek M, Leisenring WM, Jerome KR, Schiffer JT, Finak G, De Rosa SC, Boeckh M. 2021. Cytomegalovirus-specific T-cell reconstitution following letermovir prophylaxis after hematopoietic cell transplantation. Blood 138:34–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Boeckh M, Kim HW, Flowers ME, Meyers JD, Bowden RA. 2006. Long-term acyclovir for prevention of varicella zoster virus disease after allogeneic hematopoietic cell transplantation--a randomized double-blind placebo-controlled study. Blood 107:1800–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ljungman P, Wilczek H, Gahrton G, Gustavsson A, Lundgren G, Lönnqvist B, Ringdén O, Wahren B. 1986. Long-term acyclovir prophylaxis in bone marrow transplant recipients and lymphocyte proliferation responses to herpes virus antigens in vitro. Bone Marrow Transplant 1:185–92. [PubMed] [Google Scholar]
- 37.Haumont M, Jurdan M, Kangro H, Jacquet A, Massaer M, Deleersnyder V, Garcia L, Bosseloir A, Bruck C, Bollen A, Jacobs P. 1997. Neutralizing antibody responses induced by varicella-zoster virus gE and gB glycoproteins following infection, reactivation or immunization. J Med Virol 53:63–8. [DOI] [PubMed] [Google Scholar]
- 38.Wang B, Hara K, Kawabata A, Nishimura M, Wakata A, Tjan LH, Poetranto AL, Yamamoto C, Haseda Y, Aoshi T, Munakata L, Suzuki R, Komatsu M, Tsukamoto R, Itoh T, Nishigori C, Saito Y, Matozaki T, Mori Y. 2020. Tetrameric glycoprotein complex gH/gL/gQ1/gQ2 is a promising vaccine candidate for human herpesvirus 6B. PLoS Pathog 16:e1008609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hill JA, Giralt S, Torgerson TR, Lazarus HM. 2019. CAR-T - and a side order of IgG, to go? - Immunoglobulin replacement in patients receiving CAR-T cell therapy. Blood Rev 38:100596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Ueda M, Berger M, Gale RP, Lazarus HM. 2018. Immunoglobulin therapy in hematologic neoplasms and after hematopoietic cell transplantation. Blood Rev 32:106–115. [DOI] [PubMed] [Google Scholar]
- 41.Thomson KJ, Hart DP, Banerjee L, Ward KN, Peggs KS, Mackinnon S. 2005. The effect of low-dose aciclovir on reactivation of varicella zoster virus after allogeneic haemopoietic stem cell transplantation. Bone Marrow Transplant 35:1065–9. [DOI] [PubMed] [Google Scholar]
- 42.Kotton CN, Kumar D, Caliendo AM, Huprikar S, Chou S, Danziger-Isakov L, Humar A, Group TTSICC. 2018. The Third International Consensus Guidelines on the Management of Cytomegalovirus in Solid-organ Transplantation. Transplantation 102:900–931. [DOI] [PubMed] [Google Scholar]
- 43.Chemaly RF, El Haddad L, Winston DJ, Rowley SD, Mulane KM, Chandrasekar P, Avery RK, Hari P, Peggs KS, Kumar D, Nath R, Ljungman P, Mossad SB, Dadwal SS, Blanchard T, Shah DP, Jiang Y, Ariza-Heredia E. 2020. Cytomegalovirus (CMV) Cell-Mediated Immunity and CMV Infection After Allogeneic Hematopoietic Cell Transplantation: The REACT Study. Clin Infect Dis 71:2365–2374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kumar D, Chin-Hong P, Kayler L, Wojciechowski D, Limaye AP, Osama Gaber A, Ball S, Mehta AK, Cooper M, Blanchard T, MacDougall J, Kotton CN. 2019. A prospective multicenter observational study of cell-mediated immunity as a predictor for cytomegalovirus infection in kidney transplant recipients. Am J Transplant 19:2505–2516. [DOI] [PubMed] [Google Scholar]
- 45.Cunningham AL, Lal H, Kovac M, Chlibek R, Hwang SJ, Díez-Domingo J, Godeaux O, Levin MJ, McElhaney JE, Puig-Barberà J, Vanden Abeele C, Vesikari T, Watanabe D, Zahaf T, Ahonen A, Athan E, Barba-Gomez JF, Campora L, de Looze F, Downey HJ, Ghesquiere W, Gorfinkel I, Korhonen T, Leung E, McNeil SA, Oostvogels L, Rombo L, Smetana J, Weckx L, Yeo W, Heineman TC, Group Z-S. 2016. Efficacy of the Herpes Zoster Subunit Vaccine in Adults 70 Years of Age or Older. N Engl J Med 375:1019–32. [DOI] [PubMed] [Google Scholar]
- 46.Oxman MN, Levin MJ, Johnson GR, Schmader KE, Straus SE, Gelb LD, Arbeit RD, Simberkoff MS, Gershon AA, Davis LE, Weinberg A, Boardman KD, Williams HM, Zhang JH, Peduzzi PN, Beisel CE, Morrison VA, Guatelli JC, Brooks PA, Kauffman CA, Pachucki CT, Neuzil KM, Betts RF, Wright PF, Griffin MR, Brunell P, Soto NE, Marques AR, Keay SK, Goodman RP, Cotton DJ, Gnann JW, Loutit J, Holodniy M, Keitel WA, Crawford GE, Yeh SS, Lobo Z, Toney JF, Greenberg RN, Keller PM, Harbecke R, Hayward AR, Irwin MR, Kyriakides TC, Chan CY, Chan IS, Wang WW, Annunziato PW, Silber JL, et al. 2005. A vaccine to prevent herpes zoster and postherpetic neuralgia in older adults. N Engl J Med 352:2271–84. [DOI] [PubMed] [Google Scholar]
- 47.SHINGRIX. Administration UFaD, Published online August 2, 2021. Accessed September 21 2021. https://www.fda.gov/vaccines-blood-biologics/vaccines/shingrix.
- 48.Bastidas A, de la Serna J, El Idrissi M, Oostvogels L, Quittet P, López-Jiménez J, Vural F, Pohlreich D, Zuckerman T, Issa NC, Gaidano G, Lee JJ, Abhyankar S, Solano C, Perez de Oteyza J, Satlin MJ, Schwartz S, Campins M, Rocci A, Vallejo Llamas C, Lee DG, Tan SM, Johnston AM, Grigg A, Boeckh MJ, Campora L, Lopez-Fauqued M, Heineman TC, Stadtmauer EA, Sullivan KM, Collaborators Z-HSG. 2019. Effect of Recombinant Zoster Vaccine on Incidence of Herpes Zoster After Autologous Stem Cell Transplantation: A Randomized Clinical Trial. JAMA 322:123–133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Vink P, Ramon Torrell JM, Sanchez Fructuoso A, Kim SJ, Kim SI, Zaltzman J, Ortiz F, Campistol Plana JM, Fernandez Rodriguez AM, Rebollo Rodrigo H, Campins Marti M, Perez R, González Roncero FM, Kumar D, Chiang YJ, Doucette K, Pipeleers L, Agüera Morales ML, Rodriguez-Ferrero ML, Secchi A, McNeil SA, Campora L, Di Paolo E, El Idrissi M, López-Fauqued M, Salaun B, Heineman TC, Oostvogels L, Group Z-S. 2020. Immunogenicity and Safety of the Adjuvanted Recombinant Zoster Vaccine in Chronically Immunosuppressed Adults Following Renal Transplant: A Phase 3, Randomized Clinical Trial. Clin Infect Dis 70:181–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Dagnew AF, Ilhan O, Lee WS, Woszczyk D, Kwak JY, Bowcock S, Sohn SK, Rodriguez Macías G, Chiou TJ, Quiel D, Aoun M, Navarro Matilla MB, de la Serna J, Milliken S, Murphy J, McNeil SA, Salaun B, Di Paolo E, Campora L, López-Fauqued M, El Idrissi M, Schuind A, Heineman TC, Van den Steen P, Oostvogels L, group Z-s. 2019. Immunogenicity and safety of the adjuvanted recombinant zoster vaccine in adults with haematological malignancies: a phase 3, randomised, clinical trial and post-hoc efficacy analysis. Lancet Infect Dis 19:988–1000. [DOI] [PubMed] [Google Scholar]
- 51.Cunningham AL, Heineman TC, Lal H, Godeaux O, Chlibek R, Hwang SJ, McElhaney JE, Vesikari T, Andrews C, Choi WS, Esen M, Ikematsu H, Choma MK, Pauksens K, Ravault S, Salaun B, Schwarz TF, Smetana J, Abeele CV, Van den Steen P, Vastiau I, Weckx LY, Levin MJ, Group Z-S. 2018. Immune Responses to a Recombinant Glycoprotein E Herpes Zoster Vaccine in Adults Aged 50 Years or Older. J Infect Dis 217:1750–1760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Boeckh MJ, Arvin AM, Mullane KM, Camacho LH, Winston DJ, Morrison VA, Hurtado K, Durrand Hall J, Pang L, Su SC, Kaplan SS, Annunziato PW, Popmihajlov Z, Group VPTGaVPT. 2020. Immunogenicity of Inactivated Varicella Zoster Vaccine in Autologous Hematopoietic Stem Cell Transplant Recipients and Patients With Solid or Hematologic Cancer. Open Forum Infect Dis 7:ofaa172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Camargo JF, Lin RY, Natori Y, Anderson AD, Alencar MC, Wang TP, Morris MI, Komanduri KV. 2020. Reduced immunogenicity of the adjuvanted recombinant zoster vaccine after hematopoietic cell transplant: a pilot study. Blood Adv 4:4618–4622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zaia JA, Levin MJ, Preblud SR, Leszczynski J, Wright GG, Ellis RJ, Curtis AC, Valerio MA, LeGore J. 1983. Evaluation of varicella-zoster immune globulin: protection of immunosuppressed children after household exposure to varicella. J Infect Dis 147:737–43. [DOI] [PubMed] [Google Scholar]
- 55.Gottlieb DJ, Clancy LE, Withers B, McGuire HM, Luciani F, Singh M, Hughes B, Gloss B, Kliman D, Ma CKK, Panicker S, Bishop D, Dubosq MC, Li Z, Avdic S, Micklethwaite K, Blyth E. 2021. Prophylactic antigen-specific T-cells targeting seven viral and fungal pathogens after allogeneic haemopoietic stem cell transplant. Clin Transl Immunology 10:e1249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Tzannou I, Papadopoulou A, Naik S, Leung K, Martinez CA, Ramos CA, Carrum G, Sasa G, Lulla P, Watanabe A, Kuvalekar M, Gee AP, Wu MF, Liu H, Grilley BJ, Krance RA, Gottschalk S, Brenner MK, Rooney CM, Heslop HE, Leen AM, Omer B. 2017. Off-the-Shelf Virus-Specific T Cells to Treat BK Virus, Human Herpesvirus 6, Cytomegalovirus, Epstein-Barr Virus, and Adenovirus Infections After Allogeneic Hematopoietic Stem-Cell Transplantation. J Clin Oncol 35:3547–3557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Houghtelin A, Bollard CM. 2017. Virus-Specific T Cells for the Immunocompromised Patient. Front Immunol 8:1272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Papadopoulou A, Koukoulias K, Alvanou M, Papadopoulos VK, Bousiou Z, Kalaitzidou V, Kika FS, Papalexandri A, Mallouri D, Batsis I, Sakellari I, Anagnostopoulos A, Yannaki E. Patient risk stratification and tailored clinical management of post-transplant CMV-, EBV-, and BKV-infections by monitoring virus-specific T-cell immunity. eJHaem 2:428–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Phase 2 Multicenter, Randomized, Double-blind, Placebo-controlled Study to Assess the Safety and Efficacy of Viralym- M Compared to Placebo for the Prevention of AdV, BKV, CMV, EBV, HHV-6, and JCV Infection and/or Disease, in High-Risk Patients After Allogeneic Hematopoietic Cell Transplant. https://clinicaltrials.gov/ct2/show/NCT04693637?term=viralym&draw=2&rank=4. Accessed 29 Sep 2021.
- 60.Ma CK, Clancy L, Deo S, Blyth E, Micklethwaite KP, Gottlieb DJ. 2017. Herpes simplex virus type 1 (HSV-1) specific T-cell generation from HLA-A1- and HLA-A2-positive donors for adoptive immunotherapy. Cytotherapy 19:107–118. [DOI] [PubMed] [Google Scholar]