Next Article in Journal
Feasibility of Polyclonal Avian Immunoglobulins (IgY) as Prophylaxis against Human Norovirus Infection
Next Article in Special Issue
An Update on Current Antiviral Strategies to Combat Human Cytomegalovirus Infection
Previous Article in Journal
Structural Insights into Common and Host-Specific Receptor-Binding Mechanisms in Algal Picorna-like Viruses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Adoptive Immunotherapy for Prophylaxis and Treatment of Cytomegalovirus Infection

by
Christopher P. Ouellette
Division of Pediatric Infectious Diseases and Host Defense Program, Nationwide Children’s Hospital, Columbus, OH 43205, USA
Viruses 2022, 14(11), 2370; https://doi.org/10.3390/v14112370
Submission received: 5 October 2022 / Revised: 22 October 2022 / Accepted: 25 October 2022 / Published: 27 October 2022
(This article belongs to the Special Issue Antiviral Treatment for Cytomegalovirus Diseases)

Abstract

:
Cytomegalovirus (CMV), a member of the Herpesviridae family, is frequent among hematopoietic cell transplant (HCT) and solid organ transplant (SOT) recipients in absence of antiviral prophylaxis, and is a major cause of morbidity and mortality in these vulnerable populations. Antivirals such ganciclovir, valganciclovir, and foscarnet are the backbone therapies, however drug toxicity and antiviral resistance may render these agents suboptimal in treatment. Newer therapies such as letermovir and maribavir have offered additional approaches for antiviral prophylaxis as well as treatment of drug resistant CMV infection, though may be limited by cost, drug intolerance, or toxicity. Adoptive immunotherapy, the transfer of viral specific T-cells (VSTs), offers a new approach in treatment of drug-resistant or refractory viral infections, with early clinical trials showing promise with respect to efficacy and safety. In this review, we will discuss some of the encouraging results and challenges of widespread adoption of VSTs in care of immunocompromised patients, with an emphasis on the clinical outcomes for treatment and prophylaxis of CMV infection among high-risk patient populations.

1. Introduction

Cytomegalovirus (CMV) is a ubiquitous virus responsible for significant mortality and morbidity in immunocompromised patients [1]. CMV infection affects 60–80% of solid organ transplant (SOT) and 70% of seropositive hematopoietic cell transplant (HCT) recipients in the absence of antiviral prophylaxis [2,3,4]. Risk factors associated with CMV reactivation include seropositivity of the recipient, reduced intensity conditioning, graft-versus-host disease (GVHD), human leukocyte antigens (HLA-B14, HLA-DRB1*01 and HLA-DRB1*13), donor activating killer immunoglobulin-like receptors (KIR) and use of high dose corticosteroids [2,3,5]. For SOT recipients, patients receiving lung and small intestine transplants have the highest risk of CMV disease, while heart recipients have a lower risk and renal transplant recipients have the lowest risk [6]. Among HCT and SOT recipients, a complex interplay between CMV infection, transplant type, GVHD and graft rejection, and the resultant net state of immunosuppression exists, creating a challenging environment of both infection and immunomodulatory management [7]. Current management focuses on reduction in immunosuppression and initiation of antiviral therapies including ganciclovir, cidofovir, foscarnet, and valganciclovir [8]. Use of these agents, while necessary, carries substantial side effect profiles including marrow suppression and nephrotoxicity. Furthermore, the development of resistance to antiviral drugs via mutations to CMV UL97 or UL54 genes further limits the efficacy of these agents against CMV [8]. Although the incidence of drug-resistant CMV remains low (0–8%) [9], it can be as high as 14.5% in high risk populations such as haploidentical HCT receiving antiviral prophylaxis [10]. Letermovir, recently FDA approved for antiviral prophylaxis for CMV post HCT, has not been approved for treatment of drug-resistant CMV [11]. Maribavir, a potent, selective nucleoside analog with activity among UL97 resistant CMV isolates, was recently approved by the FDA for treatment of refractory or resistant CMV infection, however, is not without issues surrounding drug intolerance or development of antiviral resistance [12,13]. While numerous antiviral therapies are available and have improved outcomes for CMV infection in immunocompromised patient populations, none address a primary issue with relation to viral disease in immunocompromised patient populations: the delay or failure of T-cell immune reconstitution. Given the high relevance to management of viral disease, promoting immune reconstitution via adoptive immunotherapy serves as a logical approach to treatment. Adoptive immunotherapy, specifically the transfer of viral-specific T-cells (VST) towards the virus of interest (ex. CMV), overcomes barriers of medical intolerance or antiviral resistance. Initial approaches using adoptive immunotherapy for treatment of infection included use donor lymphocyte infusions, however this approach (by nature of using a non-specific product) is complicated by high rates of GVHD (~60%) [14]. Advances in generation of viral specific T-cell products, though antigen selection or by ex vivo expansion, have demonstrated encouraging results with respect to efficacy and safety profiles [15,16,17,18]. In this review, we will focus on the use of adoptive immunotherapy for treatment and prophylaxis of CMV infection in immunocompromised patient populations.

2. Pathophysiology of CMV Infection and Rationale for Adoptive Immunotherapy

Cytomegalovirus, a betaherpesvirus and member of the Herpesviridae, infects a wide variety of cell types, establishing latency with episodes of periodic reactivation. Primary CMV infection induces a significant proinflammatory response (including interleukin-18 and interferon-ɣ (IFNɣ)) from innate immune cells [19]. NK cells perform a critical role in early CMV infection management, and defects of NK cell function are known to predispose to severe Herpesviridae infections [20]. Additionally, use of NK cells as adoptive immunotherapy concomitant to HCT has demonstrated an ancillary benefit in reduction of rates of CMV reactivation [21]. While the innate immune responses help temporize infection, adaptive immune responses, in particular CMV specific CD4+ and CD8+ T-cells, are critical in control of CMV infection [22]. Deficiencies or suboptimal responses from T-cell compartments have been observed to result in severe infections from herpesviruses [23]. Individuals with severe-combined immune deficiency are at life-threatening risk of severe, disseminated CMV infection [24]. Additionally, individuals with infection due to human immunodeficiency virus are at greatest risk with low CD4+ T-cell quantitative values, predisposing to risk of severe or fulminant disease [25]. Clinically, the use of functional cell-mediated immunity assessments in both HCT and SOT recipients can used in risk-assessment for viral reactivation [26,27,28]. In total, while the application of antiviral therapy has undoubtedly reduced the morbidity and mortality associated with CMV disease, reconstitution of immunity, particularly the adaptive immune response, is critical in long-term virologic control of CMV in those with compromised immune systems.

3. The Infusional Product: Weighing the Pros and Cons

The immunobiology of VST generation has been discussed in several other review articles, highlighting the intricate yet subtle differences between VST products [29,30]. Here, we will briefly discuss the generalities of the VST product generation, including pertinent advantages and limitations of each approach.
Autologous or donor derived, ex vivo expanded VST: in this approach, the subject’s (or original stem cell donor) peripheral blood mononuclear cells (PMBCs) are collected, and through repetitive antigenic (ex. pp65) and cytokine (ex. IL-2) stimulation, CMV T-cell populations are expanded ex vivo [17,31]. The resultant CMV specific T-cell product is then infused back to the patient to elicit antiviral efficacy. One primary advantage to this approach is risk minimization for graft-versus-host disease (GVHD) or graft rejection (GR) as this product is matched to the recipient marrow, however comes at the expense of cost and VST generation time.
Allogeneic, ex vivo expanded VST (aka. Off-the-Shelf VST): in this approach, a pool of VST products from varying HLA haplotypes is generated. The methodology for VST generation in this approach bears similarity to autologously expanded VSTs in that collected PBMCs undergoes repeated antigenic and cytokine stimulation to generate an expanded VST product ex vivo [15,16,32,33]. The VST product is then cryopreserved for future infusion upon demand. Several advantages to this approach are noted. While the generation of each cell product does require time, this is done in advance; cryopreservation of a VST with known HLA and anti-viral activity allows for rapid product identification and infusion. This approach also allows for development of multi-virus (i.e., combination of CMV, EBV, Adenovirus, HHV-6, BK, etc.) specific VST products for treatment of patients with polyviral detection [15,16,33]. Lastly, VST efficacy may be achieved with as little as 1 HLA match [15], improving the potential donor pool. Downsides to this approach also exist, primarily as some VST candidates may not have an available HLA-matched product among the banked VST pool. Cost also remains a large barrier to development as a large number of VST products will need to be generated to cover a broad portion of HLAs among the population that may require therapy.
Donor derived, antigen selected VST: in this approach, the original marrow donor, a matched donor, or partially matched donor, undergoes evaluation for the presence of prior viral infection (i.e., presence of CMV IgG antibody) for the virus of interest. The selected donor undergoes leukapheresis, for which PBMCs are collected and subsequently exposed to antigenic stimulation (ex. pp65 in setting of CMV) for IFNɣ production. Viral-specific T-cells are then selected utilizing a high throughput, IFNɣ capture system [18,34,35]. The resulting VST product is assessed for release criteria and infused into the patient. Several advantages exist with this approach, namely the relative rapidity of VST administration (typically about 1–2 weeks based upon donor screening and selection, with only an ~24-h time for VST generation from the donor leukapheresis). Improved HLA matching may also be achieved through this process than via use of an off-the-shelf product (i.e., utilizing parent or children as potential donors, thus achieving HLA haploidentical matching). Some downsides to this approach exist–it requires the potential donor to have prior viral T-cell immunity to the pathogen of interest (prior viral infection to CMV, for example) as the VST is a selected product that does not undergo antigenic exposure to create T-cell immunity. Similarly, as the VST product is not expanded, the total VST dose is often lower with this approach. Cost remains a relative barrier, as it requires investment in facilities and equipment for VST generation.

4. Adoptive Immunotherapy in HCT Recipients

The use of VSTs for treatment of CMV infection in HCT recipients has been well documented, ranging from case reports to phase I and II trials. Studies highlighting the use of CMV VSTs in HCT recipients are summarized in Table 1. In total, 26 studies and case reports, including >450 pediatric and adult HCT recipients with CMV infection, have been published detailing CMV VSTs as therapy. One of the first reports utilizing VSTs as therapy was by Einsele et al. [36], where 8 adult HCT recipients with refractory or resistant CMV infection received donor-derived CMV VSTs generated by ex vivo expansion. Enrolled patients received a VST cell dose of 1 × 107/m2, and in total 6 of 8 (75%) patients demonstrated virologic clearance. Importantly, no evidence of de novo GVHD was reported in these patients. Cumulatively, clinical response rates of VSTs for treatment of CMV infection or disease ranged from 33–100%. However, in considering studies reporting ≥5 patients (21 in total), 86% (18/21) documented response rates of >70% (inclusive of partial or complete response to therapy). When considering efficacy in this context, indication for treatment is quite relevant-the primary indication of CMV VSTs in the majority of studies was refractory or resistant CMV infection, thus representing individuals who already failed standard-of-care antiviral therapy with ganciclovir, valganciclovir, or foscarnet. Numerous open trials for HCT recipients are available (clinicaltrials.gov, accessed on 4 October 2022) with varying target populations, modalities of VST generation, dosing regimens, and outcome measures. Several ongoing clinical trials are underway utilizing multi-virus specific off-the-shelf VST (NCT04013802) products or cytokine capture VST products (NCT04364178) for management of CMV infection after HCT. Importantly, one phase III randomized, controlled trial (NCT04832607) addressing VST therapy for CMV infection after HCT, is underway.
Use of VSTs as antiviral prophylaxis after HCT has also been explored. The first study to report adoptive transfer of CMV specific immunity for prevention was Riddell et al. in 1992 [37], utilizing donor-derived, MHC class I restricted CD8+ T-cell clones expanded ex vivo prior to infusion. Among the 3 patients who received therapy, none developed CMV reactivation post-transplant, which was accompanied by a measurable CMV specific T-cell response post-transplant. In total, 11 reports describing CMV VSTs as prophylaxis against CMV infection have been reported in the literature, inclusive of >100 pediatric and adult HCT recipients. Interpretation of cumulative efficacy in the prophylaxis data challenging based upon various factors, including risk of CMV reactivation, timing of VST administration (relative to detection of CMV) and outcome measures. In total, a wide range of efficacy with respect to viral reactivation was reported (0–100%). However, among 5 studies reporting rates of CMV reactivation post CMV VST administration [17,38,39,40,41], CMV detection after VSTs as prophylaxis occurred in ~36% (23/64) of patients. In addition to CMV detection after VST administration, other endpoints have been assessed. Blyth et al. [17] reported on VST administration for prevention of CMV infection or reactivation after allogeneic HCT in adult and pediatric patients. In their study, donor-derived, ex vivo expanded CMV VSTs were administered on D+28 at doses of 2 × 107/m2. While their study found no difference in the rate of reactivation between individuals who received VSTs and control patients (p = 0.17), the study did note a significant reduction in peak viral load (p = 0.04) and number of patients who required antiviral therapy (36% versus 18%, p = 0.01) [17].
Table 1. Adoptive Immunotherapy for Treatment or Prophylaxis of Cytomegalovirus Infection or Disease in HCT Recipients.
Table 1. Adoptive Immunotherapy for Treatment or Prophylaxis of Cytomegalovirus Infection or Disease in HCT Recipients.
AuthorYearPatient AgeNumber of ptsPopulationVST SourceIndicationCell Dose
(Total, /m2, or /kg)
ResultsAdverse Events
Pei et al. [42]2022Adult, peds190Haplo HCTDonorRefractory CMV or at risk for progressive CMV disease1 × 103/kg–1 × 109/m2Cumulative response at 6 weeks: 89.5%No infusional toxicities, cumulative incidence of GVHD at 3 months 14.3%
Rubinstein et al. [43]2022Peds23
(CMV; n = 13 with either D+ or R+ serostatus)
Allo HCTDonorProphylaxis of infection (CMV, EBV, AdV, BK)2 × 107/m23 patients with clinically significant viremia (CMV = 1) 2 patients with GVHD
Wang et al. [44]2021Peds10Allo HCTDonorCMV disease or refractory infection0.5–1 × 108/kgAll patients with improved viral load at 4 weeksNo exacerbation of existing GVHD
Gottleib et al. [45]2021Adult11Allo HCTDonorProphylaxis of infection (CMV, EBV, AdV, VZV, Influenza, BK, A. fumigatus)Not mentioned8 of 10 patients with CMV reactivation noted before infusion, 3 developed end-organ diseaseNo infusional toxicities, 6 cases of GVHD post infusion
Celilova et al. [46]2020Peds1Allo HCT3rd partyDrug resistant CMV infection2 × 104/kgInitial virologic clearance, though recrudescent disease observedNo infusional toxicity, no GVHD
Seo et al. [47]2019Peds1Allo HCT3rd party (Haplo)CMV infection or disease2 × 107/m2Clinical and virologic improvementNo infusional toxicity, no GVHD
Abraham et al. [41]2019Peds14Allo HCTDonorCMV disease or CMV prophylaxis5 × 106–2.5 × 107/m23/4 with CMV viremia with virologic improvement, 6/7 with prophylaxis without reactivationGVHD in 6 patients in study
Alonso et al. [48]2019Peds2Allo HCT3rd party (Haplo)CMV infection or disease0.8–4.4 × 104/kgClinical and virologic improvementNo infusional toxicity, no GVHD
Kallay et al. [49]2018Peds9
(CMV = 3)
Allo HCT3rd partyCMV infection or disease7.5–16.2 × 104/kg2/3 (66%) with complete or partial virologic resolutionNo GVHD, no infusional toxicities.
Withers et al. [50]2017Adult, peds30Allo HCT3rd partyCMV infection or disease2 × 107/m228/30 (93%) with complete (23) or partial (5) response at ~8 weeksGVHD (grade II) in 2 patients
Neuenhahn et al. [51]2017Not reported16Allo HCTDonor, 3rd partyRefractory CMV infection or disease6.6 × 106 to 1.4 × 10712/15 (80%) patients with responseFever in 1 patient post infusion, GVHD (grade II-III) in 2 patients
Tzannou et al. [16]2017Adult, peds54
(CMV = 17)
Allo HCT3rd partyInfection due to CMV, EBV, AdV, BK, or HHV-62 × 107/m2Complete and partial response rate: 94.1% by week 6Fever with 1 patient, otherwise no infusional toxicities. 6 (11%) patients with new or recurrent GVHD post infusion
Pei et al. [52]2017Adult, peds32Haplo HCTDonorRefractory CMV infectionMedian ~2 × 10727/32 (84%)1 patient with grade II GVHD (had grade I GVHD prior to infusion)
Naik et. al [53]2016Adult, peds36
(CMV = 7)
Primary immune deficiency, s/p HCT (35/36)Donor, cord blood, or 3rd partyInfection with CMV, EBV, AdV, BK, or HHV-65 × 106–1.35 × 108/m2Complete or partial response in 86% of pts with CMV (n = 7)5 total events, GvHD (n = 4) associated with weaning IST.
Creidy et al. [54]2016Adult, peds15
(CMV = 10)
Allo HCTDonorCMV disease, AdV diseaseMedian 3.5 × 103/kg3 of 9 evaluable patients with decreased viral load. GvHD in 1 patient
Koehne et al. [55]2015Adult, peds16Allo HCTDonorRefractory CMV infection0.5–2 × 106/kg14/16 patients with viral load clearanceNo infusion related toxicity. No De novo GvHD.
Ma et al. [40]2015Adult10Allo HCTDonorProphylaxis for CMV, VZV, AdV reactivation or infection2 × 107/m26 patients developed reactivation, however only 1 required therapy2 cases of De Novo GVHD post T-cell infusion
Clancy et al. [56]2013Adult7Allo HCTDonorProphylaxis for CMV reactivation or infection2 × 107/m2 at 28d post HCTUnable to assess (3 patients with CMV detection prior to infusion)No infusion related toxicity, GVHD in 2 patients post infusion
Leen et al. [15]2013Adult, peds50
(CMV = 23, 19 evaluable)
Allo HCT3rd partyInfection of AdV, CMV, EBV2 × 107/m2CMV: response rate at 6 weeks: 74%All infusions well tolerated. De novo GVHD (n = 2)
Gerdemann et al. [32]2013Adult, peds10
(CMV = 5)
Allo HCT3rd partyInfection of AdV, CMV, EBV0.5–2 × 107/m2~80% success rate with respect to CMVNo infusion related events. GVHD in 1 patient.
Blyth et al. [17]2013Adult, peds50Allo HCTDonorProphylaxis for CMV reactivation or infection2 × 107/m2 after 28d post HCTReduction in need for directed CMV therapy (GCV, foscarnet), no reduction in reactivation ratesaGVHD in 12/50 (5 prior to VST infusion).
Bao et al. [57]2012Adult, peds7Allo HCTRecipientRefractory CMV viremia2.5–5 × 105/kg 6/7 with partial or complete virologic responseNo GVHD
Meij et al. [58]2012Adult, peds6Allo HCTDonorRefractory CMV viremia0.9 × 104–3.1 × 105/kgComplete virologic response in all patientsNo infusion related toxicities, no GVHD
Schmitt et al. [59]2011Adult2Allo HCTDonorCMV viremia0.37 × 105/kg–2.2 × 105/kgVirologic resolutionNo infusional related toxicities, no GVHD
Peggs et al. [35]2011Not stated18Allo HCTDonorPreemptive or prophylaxis of CMV1 × 104/kg7 patients with prophylaxis administration–no antiviral therapy required. No significant CMV related disease observed. No infusion related toxicity, GVHD in 3 patients
Feuchtinger et al. [18]2010Adult, peds18Allo HCTDonorRefractory CMV reactivation or disease≤5 × 104/kg
(Mean 2.1 × 104)
15/18 cases with clearance of viremia or significant reduction (>1 log)No infusion related events. GI bleed in 1 patient
Micklethwaite et al. [38]2008Adult12Allo HCTDonorProphylaxis for CMV infection2 × 107/m2 on D28 post HCT4 patients with CMV post infusion, though overall low level4 patients with GVHD post infusion (most associated with subtherapeutic IST levels)
Horn et al. [60]2008Adult1Haplo HCTDonorRefractory CMV infection2.5 × 105Virologic improvementNo GVHD or infusion related events
Mackinnon et al. [61]2007Not stated23 (16 with expanded product, 7 with IFNɣ selected product)Allo HCTDonorCMV reactivation or disease1 × 105/kg (expanded) or 1 × 104/kg (IFNγ selected)8 (50%) with virologic resolution (expanded), 4/5 (80%) with reduced antiviral duration of therapy (IFNɣ selected)No infusional toxicities, 3 patients with grade I GVHD
Leen et al. [33]2006Adult, peds11Allo HCT3rd partyCMV reactivation or disease5 × 106–1 × 108/m2All subjects with prolonged CMV antigen clearanceNo GVHD observed in cohort
Perruccio et al. [39]2005Adult, peds25Haplo HCTDonorProphylaxis of CMV infection1–10 × 105/kg7/25 with CMV reactivation, 2 deathsGVHD (grade II) in 1 patient
Cobbold [62]2005Not stated9Allo HCTDonorRefractory CMV viremia or primary detection1.2 × 103/kg–3.3 × 104/kg Viremia resolution in 8/9, viral load reduction in 1. GVHD (grade II) in 2 patients (though grade I GVHD noted prior to infusion)
Peggs et al. [63]2003Not stated16Allo HCTDonorCMV infection1 × 105/kgAll 16 cleared viral detection (8 while also receiving antiviral therapy). 2 episodes of CMV recurrence notedNo infusion related toxicity. GVHD in 3 patients (De novo in 2)
Einsele et al. [36]2002Adult8Allo HCTDonorRefractory CMV infection1 × 107/m26/8 with virologic clearanceNo GVHD events
Walter et al. [64]1995Adult, peds14Allo HCTDonorPrevention of CMV infection3.3 × 106–1 × 109/m2No patient developed CMV viremia or diseaseGVHD (grades I-II) in 3 patients
Riddell et al. [37]1992Adult3Allo HCTDonorPrevention of CMV infection3.3 × 106–1 × 109/m2No patient developed CMV viremia or diseaseNo infusional toxicity observed
Abbreviations: HCT, hematopoietic cell transplantation; VST, viral specific T-cell; Haplo, haploidentical; CMV, cytomegalovirus; GVHD, graft-versus-host disease; Allo, allogeneic; EBV, Epstein–Barr virus; AdV, Adenovirus; VZV, varicella zoster virus; HHV-6, human herpesvirus 6; aGVHD, acute graft-versus-host disease; IFNγ, interferon gamma.
Currently, one randomized clinical trial (NCT02108522) is underway studying the effect of an off-the-shelf, multi-virus specific T-cell product (ALVR105) for prevention of viral infection, including CMV, after allogeneic HCT. The VST product in this study was utilized for treatment of CMV infection post HCT in a Phase II, non-randomized open-label study with noted treatment success [16].
With respect to potential adverse events, the greatest concern was the development of GVHD after VST infusion. Reported rates of GVHD ranged considerably, from 0–22% among patients receiving the VST as therapy, in contrast to 4–55% who received the VST as an antiviral prophylaxis. In evaluating cumulative rates of GVHD from trials, individuals receiving VSTs as prophylaxis tended to have higher rates of GVHD post-VST infusion in comparison to those with receipt as treatment (~25% versus ~10%); however, this finding is likely confounded by the timing of VST administration post-transplant, and difficulty in differentiating GVHD development related to the VST product as opposed to that of HCT. Importantly, the grade of GVHD post-VST infusion was often mild in nature (grade I-II), with very few reports of severe morbidity or mortality post-VST infusion.

5. Experience of Adoptive Immunotherapy in SOT Recipients

The application of adoptive immunotherapy among SOT recipients is less robust than HCT recipients. The majority of patients treated with this approach have utilized Epstein–Barr Virus (EBV) specific VSTs in treatment of EBV+ PTLD disorders or viremia with variable success [65,66,67,68,69]. Studies highlighting use of CMV VSTs in SOT recipients are summarized in Table 2. The majority of reports are single cases describing success, however a recent phase I trial among adult SOT recipients noted success of autologous, ex vivo expanded CMV VSTs in treatment of CMV infection. [31] In this study by Smith et al., 13 adult SOT (lung, heart, and kidney) recipients received CMV VSTs; 11 (84.6%) demonstrated clinical or virologic response to therapy. Importantly, administration of autologously derived CMV VSTs were able to demonstrate efficacy despite infusion in the setting of ongoing maintenance immunosuppression, as well as avoiding the development of graft rejection or dysfunction. Given the potential of delivering alloreactive T-cells in SOT recipients, graft rejection is among one of the greater concerns with VST therapy. Fortunately, only 1 case report of fatal graft rejection post VST administration for CMV infection has been noted [70], however the total number of SOT recipients who’ve received CMV specific VSTs remains sparce in the literature.

6. Experience of Adoptive Immunotherapy in Other Immunocompromised Patient Popu-Lations

Limited data exist for use of adoptive immunotherapy beyond the HCT and SOT recipient. Patients with primary immunodeficiencies have received VSTs for treatment of CMV infection prior to subsequently after transplantation with complete or partial response in 86% of cases [53]. Recently, the use of a haploidentical, selected VST productfor treatment of CMV disease in a patient with rheumatoid arthritis and chronic lymphocytic leukemia was reported, noting clinical improvement after therapy [74].

7. Challenges, Limitations, and Future Directions of Adoptive Immunotherapy

Several limitations exist regarding use of adoptive immunotherapy. Due to the cellular nature of the VST product, medications with effects against T-cell activity or number have direct impact on the product. Several of these effects have been directly accounted for in the study design of protocols, specifically limiting dosing of corticosteroids to ≤0.5 mg/kg/d of prednisone equivalent, and the temporal administration of T-cell depleting agents (such as anti-thymocyte globulin and alemtuzumab, generally within 21–28 days of T-cell infusion). Ongoing immunosuppression, such as tacrolimus in HCT and SOT recipients, may also impact the overall efficacy of T-cell products. To address this, development of calcineurin-resistant VSTs have begun though have yet to enter clinical trials [75,76,77].
Several unanswered questions remain regarding VST therapies. One important question revolves around the durability of the infused product within the host. Leen at al. studied this by evaluating T-cell receptor rearrangement to identify the infusional product among the host T-cell population, noting persistence of the infusional product for up to 6 weeks in studied patients [15]. However, the duration of VST persistence varied among patients in this study. Amazingly, sequencing of T-cell receptor Beta chain (TCRB) to track VST persistence has noted detectable VST populations in recipients for up to 4 years post infusion [78]. Nevertheless, the duration of VST persistence remains relatively unknown, and is likely further impacted based upon qualities of the infusional product (ex. autologous versus HLA mismatched), as well as numerous recipient related factors (ex. presence of and treatment for GVHD/graft rejection, baseline immunosuppressive medications, etc.).
Several other critical questions remain regarding VSTs and the host, specifically “when” (i.e., timing) these cells should be administered and “who” is the appropriate candidate. The majority of clinical trials have focused on individuals with refractory or resistant CMV infection, providing therapy to those failing first line therapies before proceeding to VSTs. Contemporary studies have sought to include treatment for individuals earlier into the disease course, utilizing the VST option earlier alongside standard-of-care antiviral therapies. Several studies have included the use of VSTs as a prophylactic approach among HCT recipients. However, inclusion and exclusion criteria have varied substantially among studies, leading to difficulties in interpretating the appropriate target populations for VSTs. Additionally, variations in cell dose, use of multiple versus single-dosing approaches, use of multi-virus specific VST products, and other confounding factors (including administration of standard-of-care antiviral therapy, concomitant immunosuppressive therapies, etc.) make interpretation of efficacy challenging. Lastly, and importantly, publication bias may impact the appearance of VST efficacy.
To address this, randomized, placebo-controlled trials are needed to evaluate the true efficacy and safety of VSTs. One such trial is underway for CMV infection after allogeneic HCT (NCT04832607), and several others are enrolling for alternative disease processes such as BK polyomavirus (NCT04390113) and Adenovirus (NCT05179057). Lastly, an additional trial assessing prophylaxis against CMV, EBV, Adenovirus, BK and JC polyomaviruses, and human herpesvirus 6 (NCT05305040) in currently enrolling. Importantly, these last three mentioned clinical trials are all utilizing the same partially HLA-matched, off-the-shelf multi-virus VST product (ALVR105) for study. Additional randomized, controlled trials evaluating the various VST generation methodologies (ex. autologously derived VST, antigen-selected VST, multi-virus versus single-virus specific) in context of the degree of HLA-matching, dosing regimens (ex. total dose, single versus multiple dosing), and recipient characteristics (ex. HCT versus SOT) are needed to assess true efficacy and safety among the varying clinical contexts. Nevertheless, despite these limitations, adoptive immunotherapy has shown promise in the setting of prophylactic administration as well as for treatment of CMV infection, particularly in settings of refractory or resistant CMV disease.

Funding

No grant or honorarium was procured for publication of this manuscript.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

Author C.P.O. is a site principal investigator for the clinical trial “Haploidentical Donor CMV Specific CTL to Treat CMV Reactivation or Infection after Solid Organ & HCT” (NCT03665675).

References

  1. Teira, P.; Battiwalla, M.; Ramanathan, M.; Barrett, A.J.; Ahn, K.W.; Chen, M.; Green, J.S.; Saad, A.; Antin, J.H.; Savani, B.N.; et al. Early cytomegalovirus reactivation remains associated with increased transplant-related mortality in the current era: A CIBMTR analysis. Blood 2016, 127, 2427–2438. [Google Scholar] [CrossRef] [PubMed]
  2. Acar, K.; Aki, S.Z.; Ozkurt, Z.N.; Bozdayi, G.; Rota, S.; Turkoz Sucak, G. Factors associated with cytomegalovirus reactivation following allogeneic hematopoietic stem cell transplantation: Human leukocyte antigens might be among the risk factors. Turk. J. Haematol. 2014, 31, 276–285. [Google Scholar] [CrossRef] [PubMed]
  3. Cook, M.; Briggs, D.; Craddock, C.; Mahendra, P.; Milligan, D.; Fegan, C.; Darbyshire, P.; Lawson, S.; Boxall, E.; Moss, P. Donor KIR genotype has a major influence on the rate of cytomegalovirus reactivation following T-cell replete stem cell transplantation. Blood 2006, 107, 1230–1232. [Google Scholar] [CrossRef] [Green Version]
  4. Humar, A.; Snydman, D.; the AST Infectious Diseases Community of Practice. Cytomegalovirus in solid organ transplant recipients. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2009, 9 (Suppl. S4), S78–S86. [Google Scholar] [CrossRef] [PubMed]
  5. Roemhild, A.; Reinke, P. Virus-specific T-cell therapy in solid organ transplantation. Transpl. Int. 2016, 29, 515–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Kowalsky, S.; Arnon, R.; Posada, R. Prevention of cytomegalovirus following solid organ transplantation: A literature review. Pediatr. Transplant. 2013, 17, 499–509. [Google Scholar] [CrossRef] [PubMed]
  7. Barrett, A.J.; Bollard, C.M. The coming of age of adoptive T-cell therapy for viral infection after stem cell transplantation. Ann. Transl. Med. 2015, 3, 62. [Google Scholar] [CrossRef]
  8. El Chaer, F.; Shah, D.P.; Chemaly, R.F. How I treat resistant cytomegalovirus infection in hematopoietic cell transplantation recipients. Blood 2016, 128, 2624–2636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Hantz, S.; Garnier-Geoffroy, F.; Mazeron, M.C.; Garrigue, I.; Merville, P.; Mengelle, C.; Rostaing, L.; Saint Marcoux, F.; Essig, M.; Rerolle, J.P.; et al. Drug-resistant cytomegalovirus in transplant recipients: A French cohort study. J. Antimicrob. Chemother. 2010, 65, 2628–2640. [Google Scholar] [CrossRef] [Green Version]
  10. Shmueli, E.; Or, R.; Shapira, M.Y.; Resnick, I.B.; Caplan, O.; Bdolah-Abram, T.; Wolf, D.G. High rate of cytomegalovirus drug resistance among patients receiving preemptive antiviral treatment after haploidentical stem cell transplantation. J. Infect. Dis. 2014, 209, 557–561. [Google Scholar] [CrossRef] [PubMed]
  11. Marty, F.M.; Ljungman, P.; Chemaly, R.F.; Maertens, J.; Dadwal, S.S.; Duarte, R.F.; Haider, S.; Ullmann, A.J.; Katayama, Y.; Brown, J.; et al. Letermovir Prophylaxis for Cytomegalovirus in Hematopoietic-Cell Transplantation. N. Engl. J. Med. 2017, 377, 2433–2444. [Google Scholar] [CrossRef] [PubMed]
  12. Papanicolaou, G.A.; Silveira, F.P.; Langston, A.A.; Pereira, M.R.; Avery, R.K.; Uknis, M.; Wijatyk, A.; Wu, J.; Boeckh, M.; Marty, F.M.; et al. Maribavir for Refractory or Resistant Cytomegalovirus Infections in Hematopoietic-cell or Solid-organ Transplant Recipients: A Randomized, Dose-ranging, Double-blind, Phase 2 Study. Clin. Infect.Dis. Off. Publ. Infect. Dis. Soc. Am. 2019, 68, 1255–1264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Chou, S.; Song, K.; Wu, J.; Bo, T.; Crumpacker, C. Drug resistance mutations and associated phenotypes detected in clinical trials of maribavir for treatment of cytomegalovirus infection. J. Infect. Dis. 2020, 226, 576–584. [Google Scholar] [CrossRef] [PubMed]
  14. Frey, N.V.; Porter, D.L. Graft-versus-host disease after donor leukocyte infusions: Presentation and management. Best Pract. Res. Clin. Haematol. 2008, 21, 205–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Leen, A.M.; Bollard, C.M.; Mendizabal, A.M.; Shpall, E.J.; Szabolcs, P.; Antin, J.H.; Kapoor, N.; Pai, S.Y.; Rowley, S.D.; Kebriaei, P.; et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood 2013, 121, 5113–5123. [Google Scholar] [CrossRef] [PubMed]
  16. Tzannou, I.; Papadopoulou, A.; Naik, S.; Leung, K.; Martinez, C.A.; Ramos, C.A.; Carrum, G.; Sasa, G.; Lulla, P.; Watanabe, A.; et al. 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. 2017, 35, 3547–3557. [Google Scholar] [CrossRef] [PubMed]
  17. Blyth, E.; Clancy, L.; Simms, R.; Ma, C.K.; Burgess, J.; Deo, S.; Byth, K.; Dubosq, M.C.; Shaw, P.J.; Micklethwaite, K.P.; et al. Donor-derived CMV-specific T cells reduce the requirement for CMV-directed pharmacotherapy after allogeneic stem cell transplantation. Blood 2013, 121, 3745–3758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Feuchtinger, T.; Opherk, K.; Bethge, W.A.; Topp, M.S.; Schuster, F.R.; Weissinger, E.M.; Mohty, M.; Or, R.; Maschan, M.; Schumm, M.; et al. Adoptive transfer of pp65-specific T cells for the treatment of chemorefractory cytomegalovirus disease or reactivation after haploidentical and matched unrelated stem cell transplantation. Blood 2010, 116, 4360–4367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. van de Berg, P.J.; Heutinck, K.M.; Raabe, R.; Minnee, R.C.; Young, S.L.; van Donselaar-van der Pant, K.A.; Bemelman, F.J.; van Lier, R.A.; ten Berge, I.J. Human cytomegalovirus induces systemic immune activation characterized by a type 1 cytokine signature. J. Infect Dis. 2010, 202, 690–699. [Google Scholar] [CrossRef] [Green Version]
  20. Biron, C.A.; Byron, K.S.; Sullivan, J.L. Severe herpesvirus infections in an adolescent without natural killer cells. N. Engl. J. Med. 1989, 320, 1731–1735. [Google Scholar] [CrossRef] [PubMed]
  21. Ciurea, S.O.; Schafer, J.R.; Bassett, R.; Denman, C.J.; Cao, K.; Willis, D.; Rondon, G.; Chen, J.; Soebbing, D.; Kaur, I.; et al. Phase 1 clinical trial using mbIL21 ex vivo-expanded donor-derived NK cells after haploidentical transplantation. Blood 2017, 130, 1857–1868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wills, M.R.; Carmichael, A.J.; Mynard, K.; Jin, X.; Weekes, M.P.; Plachter, B.; Sissons, J.G. The human cytotoxic T-lymphocyte (CTL) response to cytomegalovirus is dominated by structural protein pp65: Frequency, specificity, and T-cell receptor usage of pp65-specific CTL. J. Virol. 1996, 70, 7569–7579. [Google Scholar] [CrossRef] [Green Version]
  23. Ferreira, V.H.; Kumar, D.; Humar, A. Deep Profiling of the CD8+ T-cell Compartment Identifies Activated Cell Subsets and Multifunctional Responses Associated with Control of Cytomegalovirus Viremia. Transplantation 2019, 103, 613–621. [Google Scholar] [CrossRef] [PubMed]
  24. Bonilla, F.A.; Khan, D.A.; Ballas, Z.K.; Chinen, J.; Frank, M.M.; Hsu, J.T.; Keller, M.; Kobrynski, L.J.; Komarow, H.D.; Mazer, B.; et al. Practice parameter for the diagnosis and management of primary immunodeficiency. J. Allergy Clin. Immunol. 2015, 136, 1186–1205.e78. [Google Scholar] [CrossRef] [Green Version]
  25. Gerard, L.; Leport, C.; Flandre, P.; Houhou, N.; Salmon-Ceron, D.; Pepin, J.M.; Mandet, C.; Brun-Vezinet, F.; Vilde, J.L. Cytomegalovirus (CMV) viremia and the CD4+ lymphocyte count as predictors of CMV disease in patients infected with human immunodeficiency virus. Clin. Infect.Dis. Off. Publ. Infect. Dis. Soc. Am. 1997, 24, 836–840. [Google Scholar] [CrossRef] [Green Version]
  26. Kumar, D.; Chin-Hong, P.; Kayler, L.; Wojciechowski, D.; Limaye, A.P.; Osama Gaber, A.; Ball, S.; Mehta, A.K.; Cooper, M.; Blanchard, T.; et al. A prospective multicenter observational study of cell-mediated immunity as a predictor for cytomegalovirus infection in kidney transplant recipients. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2019, 19, 2505–2516. [Google Scholar] [CrossRef] [PubMed]
  27. Chemaly, R.F.; El Haddad, L.; Winston, D.J.; Rowley, S.D.; Mulane, K.M.; Chandrasekar, P.; Avery, R.K.; Hari, P.; Peggs, K.S.; Kumar, D.; et al. Cytomegalovirus (CMV) Cell-Mediated Immunity and CMV Infection after Allogeneic Hematopoietic Cell Transplantation: The REACT Study. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2020, 71, 2365–2374. [Google Scholar] [CrossRef]
  28. Nesher, L.; Shah, D.P.; Ariza-Heredia, E.J.; Azzi, J.M.; Siddiqui, H.K.; Ghantoji, S.S.; Marsh, L.Y.; Michailidis, L.; Makedonas, G.; Rezvani, K.; et al. Utility of the Enzyme-Linked Immunospot Interferon-gamma-Release Assay to Predict the Risk of Cytomegalovirus Infection in Hematopoietic Cell Transplant Recipients. J. Infect Dis. 2016, 213, 1701–1707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Houghtelin, A.; Bollard, C.M. Virus-Specific T Cells for the Immunocompromised Patient. Front. Immunol. 2017, 8, 1272. [Google Scholar] [CrossRef] [Green Version]
  30. Shafat, M.S.; Mehra, V.; Peggs, K.S.; Roddie, C. Cellular Therapeutic Approaches to Cytomegalovirus Infection Following Allogeneic Stem Cell Transplantation. Front. Immunol. 2020, 11, 1694. [Google Scholar] [CrossRef] [PubMed]
  31. Smith, C.; Beagley, L.; Rehan, S.; Neller, M.A.; Crooks, P.; Solomon, M.; Holmes-Liew, C.L.; Holmes, M.; McKenzie, S.C.; Hopkins, P.; et al. Autologous Adoptive T-cell Therapy for Recurrent or Drug-resistant Cytomegalovirus Complications in Solid Organ Transplant Recipients: A Single-arm Open-label Phase I Clinical Trial. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2019, 68, 632–640. [Google Scholar] [CrossRef] [PubMed]
  32. Gerdemann, U.; Katari, U.L.; Papadopoulou, A.; Keirnan, J.M.; Craddock, J.A.; Liu, H.; Martinez, C.A.; Kennedy-Nasser, A.; Leung, K.S.; Gottschalk, S.M.; et al. Safety and clinical efficacy of rapidly-generated trivirus-directed T cells as treatment for adenovirus, EBV, and CMV infections after allogeneic hematopoietic stem cell transplant. Mol. Ther. J. Am. Soc. Gene Ther. 2013, 21, 2113–2121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Leen, A.M.; Myers, G.D.; Sili, U.; Huls, M.H.; Weiss, H.; Leung, K.S.; Carrum, G.; Krance, R.A.; Chang, C.C.; Molldrem, J.J.; et al. Monoculture-derived T lymphocytes specific for multiple viruses expand and produce clinically relevant effects in immunocompromised individuals. Nat. Med. 2006, 12, 1160–1166. [Google Scholar] [CrossRef] [PubMed]
  34. Auletta, J.J.; Sanchez, P.J.; Meyer, E.K.; O’Donnell, L.C.; Cassady, K.A.; Ouellette, C.P.; Hecht, S.; Diaz, A.; Pavlek, L.R.; Salamon, D.P.; et al. Adjuvant haploidentical virus-specific T lymphocytes for treatment of disseminated adenovirus infection in a premature infant. J. Allergy Clin. Immunol. 2019, 144, 594–597.e4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Peggs, K.S.; Thomson, K.; Samuel, E.; Dyer, G.; Armoogum, J.; Chakraverty, R.; Pang, K.; Mackinnon, S.; Lowdell, M.W. Directly selected cytomegalovirus-reactive donor T cells confer rapid and safe systemic reconstitution of virus-specific immunity following stem cell transplantation. Clin. Infect. Dis. Off. Publ. Infect. Dis. Soc. Am. 2011, 52, 49–57. [Google Scholar] [CrossRef] [PubMed]
  36. Einsele, H.; Roosnek, E.; Rufer, N.; Sinzger, C.; Riegler, S.; Loffler, J.; Grigoleit, U.; Moris, A.; Rammensee, H.G.; Kanz, L.; et al. Infusion of cytomegalovirus (CMV)-specific T cells for the treatment of CMV infection not responding to antiviral chemotherapy. Blood 2002, 99, 3916–3922. [Google Scholar] [CrossRef] [Green Version]
  37. Riddell, S.R.; Watanabe, K.S.; Goodrich, J.M.; Li, C.R.; Agha, M.E.; Greenberg, P.D. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science 1992, 257, 238–241. [Google Scholar] [CrossRef]
  38. Micklethwaite, K.P.; Clancy, L.; Sandher, U.; Hansen, A.M.; Blyth, E.; Antonenas, V.; Sartor, M.M.; Bradstock, K.F.; Gottlieb, D.J. Prophylactic infusion of cytomegalovirus-specific cytotoxic T lymphocytes stimulated with Ad5f35pp65 gene-modified dendritic cells after allogeneic hemopoietic stem cell transplantation. Blood 2008, 112, 3974–3981. [Google Scholar] [CrossRef]
  39. Perruccio, K.; Tosti, A.; Burchielli, E.; Topini, F.; Ruggeri, L.; Carotti, A.; Capanni, M.; Urbani, E.; Mancusi, A.; Aversa, F.; et al. Transferring functional immune responses to pathogens after haploidentical hematopoietic transplantation. Blood 2005, 106, 4397–4406. [Google Scholar] [CrossRef]
  40. Ma, C.K.; Blyth, E.; Clancy, L.; Simms, R.; Burgess, J.; Brown, R.; Deo, S.; Micklethwaite, K.P.; Gottlieb, D.J. 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 2015, 17, 1406–1420. [Google Scholar] [CrossRef]
  41. Abraham, A.A.; John, T.D.; Keller, M.D.; Cruz, C.R.N.; Salem, B.; Roesch, L.; Liu, H.; Hoq, F.; Grilley, B.J.; Gee, A.P.; et al. Safety and feasibility of virus-specific T cells derived from umbilical cord blood in cord blood transplant recipients. Blood Adv. 2019, 3, 2057–2068. [Google Scholar] [CrossRef] [PubMed]
  42. Pei, X.Y.; Zhao, X.Y.; Liu, X.F.; Mo, X.D.; Lv, M.; Xu, L.P.; Wang, Y.; Chang, Y.J.; Zhang, X.H.; Liu, K.Y.; et al. Adoptive therapy with cytomegalovirus-specific T cells for cytomegalovirus infection after haploidentical stem cell transplantation and factors affecting efficacy. Am. J. Hematol. 2022, 97, 762–769. [Google Scholar] [CrossRef] [PubMed]
  43. Rubinstein, J.D.; Lutzko, C.; Leemhuis, T.; Zhu, X.; Pham, G.; Ray, L.; Thomas, S.; Dourson, C.; Wilhelm, J.; Lane, A.; et al. Scheduled administration of virus-specific T cells for viral prophylaxis after pediatric allogeneic stem cell transplant. Blood Adv. 2022, 6, 2897–2907. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, X.; Yu, U.; Yang, C.; Wang, C.; Zhang, X.; Li, Y.; Li, C.; Wen, F.; Liu, S. Cytomegalovirus (CMV)-specific cytotoxic T lymphocyte therapy resolve CMV diseases and refractory CMV infections in paediatric recipients of allogeneic haematopoietic stem cell transplantation. Bone Marrow Transpl. 2022, 57, 271–275. [Google Scholar] [CrossRef]
  45. Gottlieb, D.J.; Clancy, L.E.; Withers, B.; McGuire, H.M.; Luciani, F.; Singh, M.; Hughes, B.; Gloss, B.; Kliman, D.; Ma, C.K.K.; et al. Prophylactic antigen-specific T-cells targeting seven viral and fungal pathogens after allogeneic haemopoietic stem cell transplant. Clin. Transl. Immunol. 2021, 10, e1249. [Google Scholar] [CrossRef]
  46. Celilova, S.; Toret, E.; Adakli Aksoy, B.; Ovali, E.; Bozkurt, C. CMV-specific T-Cells for Treatment of CMV Infection after Hematopoietic Stem Cell Transplantation in a Pediatric Case: First Application in Turkey. Turk. J. Haematol. 2020, 37, 65–67. [Google Scholar] [CrossRef]
  47. Seo, S.; Smith, C.; Fraser, C.; Patheja, R.; Shah, S.P.; Rehan, S.; Crooks, P.; Neller, M.A.; Khanna, R. Adoptive T-cell therapy for pediatric cytomegalovirus-associated retinitis. Blood Adv. 2019, 3, 1774–1777. [Google Scholar] [CrossRef]
  48. Alonso, L.; Rudilla, F.; Gimeno, R.; Codinach, M.; Blanco, M.; Querol, S.; Diaz de Heredia, C. Successful treatment of post-transplant CMV meningoencephalitis with third-party CMV virus-specific T cells: Lessons learned. Pediatr. Transplant. 2019, 23, e13584. [Google Scholar] [CrossRef]
  49. Kallay, K.; Kassa, C.; Reti, M.; Karaszi, E.; Sinko, J.; Goda, V.; Strehn, A.; Csordas, K.; Horvath, O.; Szederjesi, A.; et al. Early Experience with CliniMACS Prodigy CCS (IFN-gamma) System in Selection of Virus-specific T Cells from Third-party Donors for Pediatric Patients with Severe Viral Infections after Hematopoietic Stem Cell Transplantation. J. Immunother. 2018, 41, 158–163. [Google Scholar] [CrossRef]
  50. Withers, B.; Blyth, E.; Clancy, L.E.; Yong, A.; Fraser, C.; Burgess, J.; Simms, R.; Brown, R.; Kliman, D.; Dubosq, M.C.; et al. Long-term control of recurrent or refractory viral infections after allogeneic HSCT with third-party virus-specific T cells. Blood Adv. 2017, 1, 2193–2205. [Google Scholar] [CrossRef] [PubMed]
  51. Neuenhahn, M.; Albrecht, J.; Odendahl, M.; Schlott, F.; Dossinger, G.; Schiemann, M.; Lakshmipathi, S.; Martin, K.; Bunjes, D.; Harsdorf, S.; et al. Transfer of minimally manipulated CMV-specific T cells from stem cell or third-party donors to treat CMV infection after allo-HSCT. Leukemia 2017, 31, 2161–2171. [Google Scholar] [CrossRef] [PubMed]
  52. Pei, X.Y.; Zhao, X.Y.; Chang, Y.J.; Liu, J.; Xu, L.P.; Wang, Y.; Zhang, X.H.; Han, W.; Chen, Y.H.; Huang, X.J. Cytomegalovirus-Specific T-Cell Transfer for Refractory Cytomegalovirus Infection after Haploidentical Stem Cell Transplantation: The Quantitative and Qualitative Immune Recovery for Cytomegalovirus. J. Infect Dis. 2017, 216, 945–956. [Google Scholar] [CrossRef] [Green Version]
  53. Naik, S.; Nicholas, S.K.; Martinez, C.A.; Leen, A.M.; Hanley, P.J.; Gottschalk, S.M.; Rooney, C.M.; Hanson, I.C.; Krance, R.A.; Shpall, E.J.; et al. Adoptive immunotherapy for primary immunodeficiency disorders with virus-specific T lymphocytes. J. Allergy Clin. Immunol. 2016, 137, 1498–1505.e1491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Creidy, R.; Moshous, D.; Touzot, F.; Elie, C.; Neven, B.; Gabrion, A.; Leruez-Ville, M.; Maury, S.; Ternaux, B.; Nisoy, J.; et al. Specific T cells for the treatment of cytomegalovirus and/or adenovirus in the context of hematopoietic stem cell transplantation. J. Allergy Clin. Immunol. 2016, 138, 920–924.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Koehne, G.; Hasan, A.; Doubrovina, E.; Prockop, S.; Tyler, E.; Wasilewski, G.; O’Reilly, R.J. Immunotherapy with Donor T Cells Sensitized with Overlapping Pentadecapeptides for Treatment of Persistent Cytomegalovirus Infection or Viremia. Biol. Blood Marrow Transplant. 2015, 21, 1663–1678. [Google Scholar] [CrossRef] [Green Version]
  56. Clancy, L.E.; Blyth, E.; Simms, R.M.; Micklethwaite, K.P.; Ma, C.K.; Burgess, J.S.; Antonenas, V.; Shaw, P.J.; Gottlieb, D.J. Cytomegalovirus-specific cytotoxic T lymphocytes can be efficiently expanded from granulocyte colony-stimulating factor-mobilized hemopoietic progenitor cell products ex vivo and safely transferred to stem cell transplantation recipients to facilitate immune reconstitution. Biol. Blood Marrow Transplant. 2013, 19, 725–734. [Google Scholar] [CrossRef] [Green Version]
  57. Bao, L.; Cowan, M.J.; Dunham, K.; Horn, B.; McGuirk, J.; Gilman, A.; Lucas, K.G. Adoptive immunotherapy with CMV-specific cytotoxic T lymphocytes for stem cell transplant patients with refractory CMV infections. J. Immunother. 2012, 35, 293–298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Meij, P.; Jedema, I.; Zandvliet, M.L.; van der Heiden, P.L.; van de Meent, M.; van Egmond, H.M.; van Liempt, E.; Hoogstraten, C.; Kruithof, S.; Veld, S.; et al. Effective treatment of refractory CMV reactivation after allogeneic stem cell transplantation with in vitro-generated CMV pp65-specific CD8+ T-cell lines. J. Immunother. 2012, 35, 621–628. [Google Scholar] [CrossRef]
  59. Schmitt, A.; Tonn, T.; Busch, D.H.; Grigoleit, G.U.; Einsele, H.; Odendahl, M.; Germeroth, L.; Ringhoffer, M.; Ringhoffer, S.; Wiesneth, M.; et al. Adoptive transfer and selective reconstitution of streptamer-selected cytomegalovirus-specific CD8+ T cells leads to virus clearance in patients after allogeneic peripheral blood stem cell transplantation. Transfusion 2011, 51, 591–599. [Google Scholar] [CrossRef] [PubMed]
  60. Horn, B.; Bao, L.; Dunham, K.; Stamer, M.; Adler, S.; Cowan, M.; Lucas, K. Infusion of cytomegalovirus specific cytotoxic T lymphocytes from a sero-negative donor can facilitate resolution of infection and immune reconstitution. Pediatr. Infect Dis. J. 2009, 28, 65–67. [Google Scholar] [CrossRef]
  61. Mackinnon, S.; Thomson, K.; Verfuerth, S.; Peggs, K.; Lowdell, M. Adoptive cellular therapy for cytomegalovirus infection following allogeneic stem cell transplantation using virus-specific T cells. Blood Cells Mol. Dis. 2008, 40, 63–67. [Google Scholar] [CrossRef] [PubMed]
  62. Cobbold, M.; Khan, N.; Pourgheysari, B.; Tauro, S.; McDonald, D.; Osman, H.; Assenmacher, M.; Billingham, L.; Steward, C.; Crawley, C.; et al. Adoptive transfer of cytomegalovirus-specific CTL to stem cell transplant patients after selection by HLA-peptide tetramers. J. Exp. Med. 2005, 202, 379–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Peggs, K.S.; Verfuerth, S.; Pizzey, A.; Khan, N.; Guiver, M.; Moss, P.A.; Mackinnon, S. Adoptive cellular therapy for early cytomegalovirus infection after allogeneic stem-cell transplantation with virus-specific T-cell lines. Lancet 2003, 362, 1375–1377. [Google Scholar] [CrossRef]
  64. Walter, E.A.; Greenberg, P.D.; Gilbert, M.J.; Finch, R.J.; Watanabe, K.S.; Thomas, E.D.; Riddell, S.R. 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, 1038–1044. [Google Scholar] [CrossRef]
  65. Sun, Q.; Burton, R.; Reddy, V.; Lucas, K.G. Safety of allogeneic Epstein-Barr virus (EBV)-specific cytotoxic T lymphocytes for patients with refractory EBV-related lymphoma. Br. J. Haematol. 2002, 118, 799–808. [Google Scholar] [CrossRef] [PubMed]
  66. Haque, T.; Amlot, P.L.; Helling, N.; Thomas, J.A.; Sweny, P.; Rolles, K.; Burroughs, A.K.; Prentice, H.G.; Crawford, D.H. Reconstitution of EBV-specific T cell immunity in solid organ transplant recipients. J. Immunol. 1998, 160, 6204–6209. [Google Scholar]
  67. Comoli, P.; Labirio, M.; Basso, S.; Baldanti, F.; Grossi, P.; Furione, M.; Vigano, M.; Fiocchi, R.; Rossi, G.; Ginevri, F.; et al. Infusion of autologous Epstein-Barr virus (EBV)-specific cytotoxic T cells for prevention of EBV-related lymphoproliferative disorder in solid organ transplant recipients with evidence of active virus replication. Blood 2002, 99, 2592–2598. [Google Scholar] [CrossRef] [Green Version]
  68. Gandhi, M.K.; Wilkie, G.M.; Dua, U.; Mollee, P.N.; Grimmett, K.; Williams, T.; Whitaker, N.; Gill, D.; Crawford, D.H. Immunity, homing and efficacy of allogeneic adoptive immunotherapy for posttransplant lymphoproliferative disorders. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2007, 7, 1293–1299. [Google Scholar] [CrossRef]
  69. Khanna, R.; Bell, S.; Sherritt, M.; Galbraith, A.; Burrows, S.R.; Rafter, L.; Clarke, B.; Slaughter, R.; Falk, M.C.; Douglass, J.; et al. Activation and adoptive transfer of Epstein-Barr virus-specific cytotoxic T cells in solid organ transplant patients with posttransplant lymphoproliferative disease. Proc. Natl. Acad. Sci. USA 1999, 96, 10391–10396. [Google Scholar] [CrossRef] [Green Version]
  70. Brestrich, G.; Zwinger, S.; Fischer, A.; Schmuck, M.; Rohmhild, A.; Hammer, M.H.; Kurtz, A.; Uharek, L.; Knosalla, C.; Lehmkuhl, H.; et al. Adoptive T-cell therapy of a lung transplanted patient with severe CMV disease and resistance to antiviral therapy. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2009, 9, 1679–1684. [Google Scholar] [CrossRef]
  71. Miele, M.; Gallo, A.; Di Bella, M.; Timoneri, F.; Barbera, F.; Sciveres, M.; Riva, S.; Grossi, P.; Conaldi, P.G. Successful Use of Heterologous CMV-Reactive T Lymphocyte to Treat Severe Refractory Cytomegalovirus (CMV) Infection in a Liver Transplanted Patient: Correlation of the Host Antiviral Immune Reconstitution with CMV Viral Load and CMV miRNome. Microorganisms 2021, 9, 684. [Google Scholar] [CrossRef] [PubMed]
  72. Macesic, N.; Langsford, D.; Nicholls, K.; Hughes, P.; Gottlieb, D.J.; Clancy, L.; Blyth, E.; Micklethwaite, K.; Withers, B.; Majumdar, S.; et al. Adoptive T cell immunotherapy for treatment of ganciclovir-resistant cytomegalovirus disease in a renal transplant recipient. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2015, 15, 827–832. [Google Scholar] [CrossRef]
  73. Holmes-Liew, C.L.; Holmes, M.; Beagley, L.; Hopkins, P.; Chambers, D.; Smith, C.; Khanna, R. Adoptive T-cell immunotherapy for ganciclovir-resistant CMV disease after lung transplantation. Clin. Transl. Immunol. 2015, 4, e35. [Google Scholar] [CrossRef] [PubMed]
  74. Yuste, J.R.; Lopez-Diaz de Cerio, A.; Rifon, J.; Moreno, C.; Panizo, M.; Inoges, S. Adoptive T-cell therapy with CD45RA-depleted donor in the treatment of cytomegalovirus disease in immunocompromised non-transplant patients. Antivir. Ther. 2019, 24, 313–319. [Google Scholar] [CrossRef] [PubMed]
  75. Amini, L.; Wagner, D.L.; Rossler, U.; Zarrinrad, G.; Wagner, L.F.; Vollmer, T.; Wendering, D.J.; Kornak, U.; Volk, H.D.; Reinke, P.; et al. CRISPR-Cas9-Edited Tacrolimus-Resistant Antiviral T Cells for Advanced Adoptive Immunotherapy in Transplant Recipients. Mol. Ther. J. Am. Soc. Gene Ther. 2021, 29, 32–46. [Google Scholar] [CrossRef] [PubMed]
  76. De Angelis, B.; Dotti, G.; Quintarelli, C.; Huye, L.E.; Zhang, L.; Zhang, M.; Pane, F.; Heslop, H.E.; Brenner, M.K.; Rooney, C.M.; et al. Generation of Epstein-Barr virus-specific cytotoxic T lymphocytes resistant to the immunosuppressive drug tacrolimus (FK506). Blood 2009, 114, 4784–4791. [Google Scholar] [CrossRef] [Green Version]
  77. Ricciardelli, I.; Brewin, J.; Lugthart, G.; Albon, S.J.; Pule, M.; Amrolia, P.J. Rapid generation of EBV-specific cytotoxic T lymphocytes resistant to calcineurin inhibitors for adoptive immunotherapy. Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. Transpl. Surg. 2013, 13, 3244–3252. [Google Scholar] [CrossRef] [Green Version]
  78. Keller, M.D.; Darko, S.; Lang, H.; Ransier, A.; Lazarski, C.A.; Wang, Y.; Hanley, P.J.; Davila, B.J.; Heimall, J.R.; Ambinder, R.F.; et al. T-cell receptor sequencing demonstrates persistence of virus-specific T cells after antiviral immunotherapy. Br. J. Haematol. 2019, 187, 206–218. [Google Scholar] [CrossRef]
Table 2. Adoptive Immunotherapy for Treatment of Cytomegalovirus Infection or Disease in SOT Recipients.
Table 2. Adoptive Immunotherapy for Treatment of Cytomegalovirus Infection or Disease in SOT Recipients.
AuthorYearPatient AgeNumber of ptsPopulationVST SourceIndicationCell Dose
(Total, /m2, or /kg)
ResultsAdverse Events
Miele et al. [71]2021Peds1SOT (liver)3rd party donor)CMV disease (pneumonitis)1 × 106/kgDecreased viral load, No infusion related toxicity, graft function stable
Smith et al. [31]2019Adult21 enrolled (13 received therapy)SOT
Renal, 4
Lung 8
Heart 1
RecipientCMV disease or persistent viremia1–2 × 107/m211/13 (84.6%) with virologic or clinical improvement. No infusion related toxicities, graft function stable
Macesic et al. [72]2015Adult1SOT (renal)3rd party donorCMV disease1.6 × 107/m2Decreased viral load, asymptomaticNo infusion related toxicity, graft function stable.
Holmes-Liew et al. [73]2015Adult1SOT (lung)RecipientCMV disease (hepatitis)12 × 107 divided over 4 dosesCMV viral load undetectable, resolved end organ diseaseNo infusion related toxicity, no graft rejection
Brestrich et al. [70]2008Adult1SOT (lung), CVID pre SOTRecipientCMV disease (pneumonitis)1 × 107/m2Symptomatic improvement, decreased viral loadNo infusion related toxicity, later death from graft failure
Abbreviations: SOT, Solid organ transplantation; VST, viral specific T-cell; CMV, cytomegalovirus; CVID; common variable immune deficiency.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ouellette, C.P. Adoptive Immunotherapy for Prophylaxis and Treatment of Cytomegalovirus Infection. Viruses 2022, 14, 2370. https://doi.org/10.3390/v14112370

AMA Style

Ouellette CP. Adoptive Immunotherapy for Prophylaxis and Treatment of Cytomegalovirus Infection. Viruses. 2022; 14(11):2370. https://doi.org/10.3390/v14112370

Chicago/Turabian Style

Ouellette, Christopher P. 2022. "Adoptive Immunotherapy for Prophylaxis and Treatment of Cytomegalovirus Infection" Viruses 14, no. 11: 2370. https://doi.org/10.3390/v14112370

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop